1
|
Poddar MS, Chu YD, Pendharkar G, Liu CH, Yeh CT. Exploring cancer-associated fibroblast-induced resistance to tyrosine kinase inhibitors in hepatoma cells using a liver-on-a-chip model. LAB ON A CHIP 2024; 24:5043-5054. [PMID: 39356081 DOI: 10.1039/d4lc00624k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/03/2024]
Abstract
Liver cancer is a significant global contributor to cancer-related mortality. Despite available targeted therapies, resistance to tyrosine kinase inhibitors (TKIs) like sorafenib and lenvatinib poses a formidable challenge. The tumor microenvironment (TME), inhabited by cancer-associated fibroblasts (CAFs), profoundly influences this resistance. To uncover the mechanisms, a 3D microfluidic chip replicating liver architecture was fabricated to probe the intricate mechanisms of TKI resistance. The chip design mirrors the hexagonal structure of liver lobules, situating liver cancer cells at the core, encircled by fibroblasts, with rigorous assessments confirming biocompatibility and consistent cell growth. After determining the IC50 values of sorafenib and lenvatinib in 2D co-culture, a transwell setup revealed drug resistance development in co-cultured cells. Within the 3D microfluidic chip, live/dead assays highlighted elevated viability under drug exposure, emphasizing fibroblast-driven drug resistance. The study identifies AHSG and CLEC3B as potential mediators of drug resistance in co-culture, significantly upregulated in the co-cultured medium. Functional tests confirmed their roles, as introducing recombinant AHSG and CLEC3B enhanced liver cancer cell resistance to sorafenib and lenvatinib in both 2D and 3D scenarios. In conclusion, by replicating the complex TME using microfluidic technology, this study sheds light on the roles of AHSG and CLEC3B as well as possible approaches for improving the effectiveness of liver cancer treatment.
Collapse
Affiliation(s)
- Madhu Shree Poddar
- Institute of Nanoengineering and Microsystems, National Tsing Hua University, Hsinchu, 30044, Taiwan, R.O.C..
| | - Yu-De Chu
- Liver Research Center, Chang Gung Memorial Hospital, Taoyuan 333, Taiwan, R.O.C..
| | - Gaurav Pendharkar
- Department of Power Mechanical Engineering, National Tsing Hua University, Hsinchu 30044, Taiwan, R.O.C
| | - Cheng-Hsien Liu
- Institute of Nanoengineering and Microsystems, National Tsing Hua University, Hsinchu, 30044, Taiwan, R.O.C..
- Department of Power Mechanical Engineering, National Tsing Hua University, Hsinchu 30044, Taiwan, R.O.C
- College of Semiconductor Research, National Tsing Hua University, Hsinchu 30044, Taiwan, R.O.C
| | - Chau-Ting Yeh
- Liver Research Center, Chang Gung Memorial Hospital, Taoyuan 333, Taiwan, R.O.C..
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital, Taoyuan 333, Taiwan, R.O.C
- Molecular Medicine Research Center, Chang Gung University, Taoyuan 333, Taiwan, R.O.C
| |
Collapse
|
2
|
Razmpour F, Daryabeygi-Khotbehsara R, Soleimani D, Asgharnezhad H, Shamsi A, Bajestani GS, Nematy M, Pour MR, Maddison R, Islam SMS. Application of machine learning in predicting non-alcoholic fatty liver disease using anthropometric and body composition indices. Sci Rep 2023; 13:4942. [PMID: 36973382 PMCID: PMC10043285 DOI: 10.1038/s41598-023-32129-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 03/22/2023] [Indexed: 03/29/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is the most common chronic liver disease, which can progress from simple steatosis to advanced cirrhosis and hepatocellular carcinoma. Clinical diagnosis of NAFLD is crucial in the early stages of the disease. The main aim of this study was to apply machine learning (ML) methods to identify significant classifiers of NAFLD using body composition and anthropometric variables. A cross-sectional study was carried out among 513 individuals aged 13 years old or above in Iran. Anthropometric and body composition measurements were performed manually using body composition analyzer InBody 270. Hepatic steatosis and fibrosis were determined using a Fibroscan. ML methods including k-Nearest Neighbor (kNN), Support Vector Machine (SVM), Radial Basis Function (RBF) SVM, Gaussian Process (GP), Random Forest (RF), Neural Network (NN), Adaboost and Naïve Bayes were examined for model performance and to identify anthropometric and body composition predictors of fatty liver disease. RF generated the most accurate model for fatty liver (presence of any stage), steatosis stages and fibrosis stages with 82%, 52% and 57% accuracy, respectively. Abdomen circumference, waist circumference, chest circumference, trunk fat and body mass index were among the most important variables contributing to fatty liver disease. ML-based prediction of NAFLD using anthropometric and body composition data can assist clinicians in decision making. ML-based systems provide opportunities for NAFLD screening and early diagnosis, especially in population-level and remote areas.
Collapse
Affiliation(s)
- Farkhondeh Razmpour
- Department of Nutrition, Faculty of Medicine, Hormozgan University of Medical Sciences, Shahid Chamran Boulevard, Bandar Abbas, Iran.
| | | | - Davood Soleimani
- Department of Nutrition, School of Nutrition Sciences and Food Technology, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Hamzeh Asgharnezhad
- Institute for Intelligent Systems Research and Innovation (IISRI), Geelong Waurn Ponds Victoria, Australia
| | - Afshar Shamsi
- Biomedical Machine Learning Lab, University of New South Whales, Sydney, Australia
- Concordia Institute for Information Systems Engineering, Concordia University, Montreal, Canada
| | - Ghasem Sadeghi Bajestani
- Department of Biomedical Engineering, Faculty of Engineering, Imam Reza International University, Mashhad, Iran
| | - Mohsen Nematy
- Metabolic Syndrome Research Center, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Ralph Maddison
- Institute for Physical Activity and Nutrition (IPAN), Deakin University, Geelong Victoria, Australia
| | | |
Collapse
|
3
|
Brown AR, Alhallak I, Simmen RCM, Melnyk SB, Heard-Lipsmeyer ME, Montales MTE, Habenicht D, Van TT, Simmen FA. Krüppel-like Factor 9 (KLF9) Suppresses Hepatocellular Carcinoma (HCC)-Promoting Oxidative Stress and Inflammation in Mice Fed High-Fat Diet. Cancers (Basel) 2022; 14:cancers14071737. [PMID: 35406507 PMCID: PMC8996893 DOI: 10.3390/cancers14071737] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 03/22/2022] [Accepted: 03/25/2022] [Indexed: 12/19/2022] Open
Abstract
Obesity, oxidative stress, and inflammation are risk factors for hepatocellular carcinoma (HCC). We examined, in mice, the effects of Krüppel-like factor 9 (KLF9) knockout on: adiposity, hepatic and systemic oxidative stress, and hepatic expression of pro-inflammatory and NOX/DUOX family genes, in a high-fat diet (HFD) context. Male and female Klf9+/+ (wild type, WT) and Klf9-/- (knockout, KO) mice were fed HFD (beginning at age 35 days) for 12 weeks, after which liver and adipose tissues were obtained, and serum adiponectin and leptin levels, liver fat content, and markers of oxidative stress evaluated. Klf9-/- mice of either sex did not exhibit significant alterations in weight gain, adipocyte size, adipokine levels, or liver fat content when compared to WT counterparts. However, Klf9-/- mice of both sexes had increased liver weight/size (hepatomegaly). This was accompanied by increased hepatic oxidative stress as indicated by decreased GSH/GSSG ratio and increased homocysteine, 3-nitrotyrosine, 3-chlorotyrosine, and 4HNE content. Decreased GSH to GSSG ratio and a trend toward increased homocysteine levels were observed in the corresponding Klf9-/- mouse serum. Gene expression analysis showed a heightened pro-inflammatory state in livers from Klf9-/- mice. KLF9 suppresses hepatic oxidative stress and inflammation, thus identifying potential mechanisms for KLF9 suppression of HCC and perhaps cancers of other tissues.
Collapse
Affiliation(s)
- Adam R. Brown
- Department of Physiology & Cell Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (A.R.B.); (I.A.); (R.C.M.S.); (M.E.H.-L.); (M.T.E.M.); (D.H.); (T.T.V.)
| | - Iad Alhallak
- Department of Physiology & Cell Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (A.R.B.); (I.A.); (R.C.M.S.); (M.E.H.-L.); (M.T.E.M.); (D.H.); (T.T.V.)
| | - Rosalia C. M. Simmen
- Department of Physiology & Cell Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (A.R.B.); (I.A.); (R.C.M.S.); (M.E.H.-L.); (M.T.E.M.); (D.H.); (T.T.V.)
- The Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Stepan B. Melnyk
- Arkansas Children’s Research Institute, Little Rock, AR 72202, USA;
| | - Melissa E. Heard-Lipsmeyer
- Department of Physiology & Cell Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (A.R.B.); (I.A.); (R.C.M.S.); (M.E.H.-L.); (M.T.E.M.); (D.H.); (T.T.V.)
| | - Maria Theresa E. Montales
- Department of Physiology & Cell Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (A.R.B.); (I.A.); (R.C.M.S.); (M.E.H.-L.); (M.T.E.M.); (D.H.); (T.T.V.)
| | - Daniel Habenicht
- Department of Physiology & Cell Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (A.R.B.); (I.A.); (R.C.M.S.); (M.E.H.-L.); (M.T.E.M.); (D.H.); (T.T.V.)
| | - Trang T. Van
- Department of Physiology & Cell Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (A.R.B.); (I.A.); (R.C.M.S.); (M.E.H.-L.); (M.T.E.M.); (D.H.); (T.T.V.)
| | - Frank A. Simmen
- Department of Physiology & Cell Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (A.R.B.); (I.A.); (R.C.M.S.); (M.E.H.-L.); (M.T.E.M.); (D.H.); (T.T.V.)
- The Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Correspondence: ; Tel.: +1-501-686-8128
| |
Collapse
|
4
|
Liu Y, Zang X, Feng K, Liu S, Zhang J, Lv Z, Xin Y, Yu M. Lipidomic Determination of Serum Lipids by Ultra-High Performance Liquid Chromatography – Mass Spectrometry (UPLC-MS) for the Characterization of Nonalcoholic Fatty Liver Disease (NAFLD). ANAL LETT 2021. [DOI: 10.1080/00032719.2021.1970760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
- YuWei Liu
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
| | - Xiaoling Zang
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
- Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Kun Feng
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
| | - Shousheng Liu
- Clinical Research Center, Qingdao Municipal Hospital, Qingdao, China
| | - Jiayuan Zhang
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
| | - Zhihua Lv
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
- Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Yongning Xin
- Department of Gastroenterology, Qingdao Municipal Hospital Group, Qingdao, China
| | - Mingming Yu
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
- Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| |
Collapse
|
5
|
Rezazadeh H, Sharifi MR, Soltani N. Insulin resistance and the role of gamma-aminobutyric acid. JOURNAL OF RESEARCH IN MEDICAL SCIENCES 2021; 26:39. [PMID: 34484371 PMCID: PMC8384006 DOI: 10.4103/jrms.jrms_374_20] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Revised: 12/09/2020] [Accepted: 02/02/2021] [Indexed: 12/17/2022]
Abstract
Insulin resistance (IR) is mentioned to be a disorder in insulin ability in insulin-target tissues. Skeletal muscle (SkM) and liver function are more affected by IR than other insulin target cells. SkM is the main site for the consumption of ingested glucose. An effective treatment for IR has two properties: An inhibition of β-cell death and a promotion of β-cell replication. Gamma-aminobutyric acid (GABA) can improve beta-cell mass and function. Multiple studies have shown that GABA decreases IR probably via increase in glucose transporter 4 (GLUT4) gene expression and prevention of gluconeogenesis pathway in the liver. This review focused on the general aspects of IR in skeletal muscle (SkM), liver; the cellular mechanism(s) lead to the development of IR in these organs, and the role of GABA to reduce insulin resistance.
Collapse
Affiliation(s)
- Hossein Rezazadeh
- Department of Physiology, School of Medicine, Isfahan University of Medical Science, Isfahan Iran
| | - Mohammad Reza Sharifi
- Department of Physiology, School of Medicine, Isfahan University of Medical Science, Isfahan Iran
| | - Nepton Soltani
- Department of Physiology, School of Medicine, Isfahan University of Medical Science, Isfahan Iran
| |
Collapse
|
6
|
Komolafe O, Buzzetti E, Linden A, Best LM, Madden AM, Roberts D, Chase TJ, Fritche D, Freeman SC, Cooper NJ, Sutton AJ, Milne EJ, Wright K, Pavlov CS, Davidson BR, Tsochatzis E, Gurusamy KS. Nutritional supplementation for nonalcohol-related fatty liver disease: a network meta-analysis. Cochrane Database Syst Rev 2021; 7:CD013157. [PMID: 34280304 PMCID: PMC8406904 DOI: 10.1002/14651858.cd013157.pub2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
BACKGROUND The prevalence of non-alcohol-related fatty liver disease (NAFLD) varies between 19% and 33% in different populations. NAFLD decreases life expectancy and increases risks of liver cirrhosis, hepatocellular carcinoma, and the requirement for liver transplantation. Uncertainty surrounds relative benefits and harms of various nutritional supplements in NAFLD. Currently no nutritional supplement is recommended for people with NAFLD. OBJECTIVES • To assess the benefits and harms of different nutritional supplements for treatment of NAFLD through a network meta-analysis • To generate rankings of different nutritional supplements according to their safety and efficacy SEARCH METHODS: We searched the Cochrane Central Register of Controlled Trials, MEDLINE, Embase, Science Citation Index Expanded, Conference Proceedings Citation Index-Science, the World Health Organization International Clinical Trials Registry Platform, and trials registers until February 2021 to identify randomised clinical trials in people with NAFLD. SELECTION CRITERIA We included only randomised clinical trials (irrespective of language, blinding, or status) for people with NAFLD, irrespective of method of diagnosis, age and diabetic status of participants, or presence of non-alcoholic steatohepatitis (NASH). We excluded randomised clinical trials in which participants had previously undergone liver transplantation. DATA COLLECTION AND ANALYSIS We performed a network meta-analysis with OpenBUGS using Bayesian methods whenever possible and calculated differences in treatments using hazard ratios (HRs), odds ratios (ORs), and rate ratios with 95% credible intervals (CrIs) based on an available-case analysis, according to National Institute of Health and Care Excellence Decision Support Unit guidance. MAIN RESULTS We included in the review a total of 202 randomised clinical trials (14,200 participants). Nineteen trials were at low risk of bias. A total of 32 different interventions were compared in these trials. A total of 115 trials (7732 participants) were included in one or more comparisons. The remaining trials did not report any of the outcomes of interest for this review. Follow-up ranged from 1 month to 28 months. The follow-up period in trials that reported clinical outcomes was 2 months to 28 months. During this follow-up period, clinical events related to NAFLD such as mortality, liver cirrhosis, liver decompensation, liver transplantation, hepatocellular carcinoma, and liver-related mortality were sparse. We did not calculate effect estimates for mortality because of sparse data (zero events for at least one of the groups in the trial). None of the trials reported that they measured overall health-related quality of life using a validated scale. The evidence is very uncertain about effects of interventions on serious adverse events (number of people or number of events). We are very uncertain about effects on adverse events of most of the supplements that we investigated, as the evidence is of very low certainty. However, people taking PUFA (polyunsaturated fatty acid) may be more likely to experience an adverse event than those not receiving an active intervention (network meta-analysis results: OR 4.44, 95% CrI 2.40 to 8.48; low-certainty evidence; 4 trials, 203 participants; direct evidence: OR 4.43, 95% CrI 2.43 to 8.42). People who take other supplements (a category that includes nutritional supplements other than vitamins, fatty acids, phospholipids, and antioxidants) had higher numbers of adverse events than those not receiving an active intervention (network meta-analysis: rate ratio 1.73, 95% CrI 1.26 to 2.41; 6 trials, 291 participants; direct evidence: rate ratio 1.72, 95% CrI 1.25 to 2.40; low-certainty evidence). Data were sparse (zero events in all groups in the trial) for liver transplantation, liver decompensation, and hepatocellular carcinoma. So, we did not perform formal analysis for these outcomes. The evidence is very uncertain about effects of other antioxidants (antioxidants other than vitamins) compared to no active intervention on liver cirrhosis (HR 1.68, 95% CrI 0.23 to 15.10; 1 trial, 99 participants; very low-certainty evidence). The evidence is very uncertain about effects of interventions in any of the remaining comparisons, or data were sparse (with zero events in at least one of the groups), precluding formal calculations of effect estimates. Data were probably because of the very short follow-up period (2 months to 28 months). It takes follow-up of 8 to 28 years to detect differences in mortality between people with NAFLD and the general population. Therefore, it is unlikely that differences in clinical outcomes are noted in trials providing less than 5 to 10 years of follow-up. AUTHORS' CONCLUSIONS The evidence indicates considerable uncertainty about effects of nutritional supplementation compared to no additional intervention on all clinical outcomes for people with non-alcohol-related fatty liver disease. Accordingly, high-quality randomised comparative clinical trials with adequate follow-up are needed. We propose registry-based randomised clinical trials or cohort multiple randomised clinical trials (study design in which multiple interventions are trialed within large longitudinal cohorts of patients to gain efficiencies and align trials more closely to standard clinical practice) comparing interventions such as vitamin E, prebiotics/probiotics/synbiotics, PUFAs, and no nutritional supplementation. The reason for the choice of interventions is the impact of these interventions on indirect outcomes, which may translate to clinical benefit. Outcomes in such trials should be mortality, health-related quality of life, decompensated liver cirrhosis, liver transplantation, and resource utilisation measures including costs of intervention and decreased healthcare utilisation after minimum follow-up of 8 years (to find meaningful differences in clinically important outcomes).
Collapse
Affiliation(s)
| | - Elena Buzzetti
- Sheila Sherlock Liver Centre, Royal Free Hospital and the UCL Institute of Liver and Digestive Health, London, UK
| | - Audrey Linden
- Division of Surgery and Interventional Science, University College London, London, UK
| | - Lawrence Mj Best
- Division of Surgery and Interventional Science, University College London, London, UK
| | - Angela M Madden
- School of Life and Medical Sciences, University of Hertfordshire, Hatfield, UK
| | - Danielle Roberts
- Division of Surgery and Interventional Science, University College London, London, UK
| | - Thomas Jg Chase
- Department of General Surgery, Homerton University Hospital NHS Foundation Trust, London, UK
| | | | - Suzanne C Freeman
- Department of Health Sciences, University of Leicester, Leicester, UK
| | - Nicola J Cooper
- Department of Health Sciences, University of Leicester, Leicester, UK
| | - Alex J Sutton
- Department of Health Sciences, University of Leicester, Leicester, UK
| | | | - Kathy Wright
- Cochrane Hepato-Biliary Group, Copenhagen Trial Unit, Centre for Clinical Intervention Research, The Capital Region of Denmark, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Chavdar S Pavlov
- Department of Therapy, I.M. Sechenov First Moscow State Medical University, Moscow, Russian Federation
| | - Brian R Davidson
- Division of Surgery and Interventional Science, University College London, London, UK
| | - Emmanuel Tsochatzis
- Sheila Sherlock Liver Centre, Royal Free Hospital and the UCL Institute of Liver and Digestive Health, London, UK
| | - Kurinchi Selvan Gurusamy
- Division of Surgery and Interventional Science, University College London, London, UK
- Department of Therapy, I.M. Sechenov First Moscow State Medical University, Moscow, Russian Federation
| |
Collapse
|
7
|
Özgür Y, Akın S, Yılmaz NG, Gücün M, Keskin Ö. Uric acid albumin ratio as a predictive marker of short-term mortality in patients with acute kidney injury. Clin Exp Emerg Med 2021; 8:82-88. [PMID: 34237812 PMCID: PMC8273677 DOI: 10.15441/ceem.20.024] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Accepted: 07/30/2020] [Indexed: 12/22/2022] Open
Abstract
OBJECTIVE We aimed to investigate uric acid and albumin ratio (UA/A) as a marker of short-term mortality in acute kidney injury (AKI). Both uric acid and albumin are strongly correlated with the development and mortality of AKI. METHODS The patients hospitalized from May 2019 to September 2019 for AKI were included in this study. The diagnostic odds ratio (DOR), Youden index (J), and the area under a receiver operating characteristic curve (AUROC) determined a cut-off UA/A ratio for mortality. Cox-regression analysis was performed to identify UA/A as a prognostic marker of the 30-day mortality rate. RESULTS A total of 171 patients with an average age of 69.20±13.0 (45.6% women) were included in the study. The average UA/A ratio was 3.3±1.5 mg/g and 2.5±1.0 mg/g in the non-survivor and survivor groups, respectively (P=0.001). The best cut-off UA/A ratio associated with mortality was determined as 2.4 mg/g with a specificity of 52% and a sensitivity of 77% (DOR, 3.6; J, 28.8; AUROC, 0.644). Thirty-day cumulative survival rates of the low and high UA/A ratio groups were 85.9±4.0% and 63.7±5.0%, respectively. The estimated survival times of the low and high UA/A ratio groups were 27.7 days (95% confidence interval [CI], 26.2-29.3) and 23.9 days (95% Cl, 22.0-25.9), respectively. CONCLUSION We found a direct correlation between 30-day mortality and UA/A ratio at initial presentation in AKI patients regardless of age, comorbidities, and clinical and laboratory findings, including albuminuria.
Collapse
Affiliation(s)
- Yasemin Özgür
- Department of Internal Medicine, Dr. Lutfi Kırdar Kartal Training and Research Hospital, University of Health Sciences, Istanbul, Turkey
| | - Seydahmet Akın
- Department of Internal Medicine, Dr. Lutfi Kırdar Kartal Training and Research Hospital, University of Health Sciences, Istanbul, Turkey
| | - Nuran Gamze Yılmaz
- Department of Internal Medicine, Dr. Lutfi Kırdar Kartal Training and Research Hospital, University of Health Sciences, Istanbul, Turkey
| | - Murat Gücün
- Department of Nephrology, Kartal Koşuyolu High Speciality Educational and Research Hospital, University of Health Sciences, Istanbul, Turkey
| | - Özcan Keskin
- Department of Internal Medicine, Dr. Lutfi Kırdar Kartal Training and Research Hospital, University of Health Sciences, Istanbul, Turkey
| |
Collapse
|
8
|
Buzzetti E, Linden A, Best LM, Madden AM, Roberts D, Chase TJG, Freeman SC, Cooper NJ, Sutton AJ, Fritche D, Milne EJ, Wright K, Pavlov CS, Davidson BR, Tsochatzis E, Gurusamy KS. Lifestyle modifications for nonalcohol-related fatty liver disease: a network meta-analysis. Cochrane Database Syst Rev 2021; 6:CD013156. [PMID: 34114650 PMCID: PMC8193812 DOI: 10.1002/14651858.cd013156.pub2] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND The prevalence of nonalcohol-related fatty liver disease (NAFLD) varies between 19% and 33% in different populations. NAFLD decreases life expectancy and increases the risks of liver cirrhosis, hepatocellular carcinoma, and requirement for liver transplantation. There is uncertainty surrounding the relative benefits and harms of various lifestyle interventions for people with NAFLD. OBJECTIVES To assess the comparative benefits and harms of different lifestyle interventions in the treatment of NAFLD through a network meta-analysis, and to generate rankings of the different lifestyle interventions according to their safety and efficacy. SEARCH METHODS We searched CENTRAL, MEDLINE, Embase, Science Citation Index Expanded, Conference Proceedings Citation Index - Science, World Health Organization International Clinical Trials Registry Platform, and trials registers until February 2021 to identify randomised clinical trials in people with NAFLD. SELECTION CRITERIA We included only randomised clinical trials (irrespective of language, blinding, or status) in people with NAFLD, whatever the method of diagnosis, age, and diabetic status of participants, or presence of non-alcoholic steatohepatitis (NASH). We excluded randomised clinical trials in which participants had previously undergone liver transplantation. DATA COLLECTION AND ANALYSIS We planned to perform a network meta-analysis with OpenBUGS using Bayesian methods and to calculate the differences in treatments using hazard ratios (HRs), odds ratios (ORs), and rate ratios (RaRs) with 95% credible intervals (CrIs) based on an available-participant analysis, according to National Institute of Health and Care Excellence Decision Support Unit guidance. However, the data were too sparse for the clinical outcomes. We therefore performed only direct comparisons (head-to-head comparisons) with OpenBUGS using Bayesian methods. MAIN RESULTS We included a total of 59 randomised clinical trials (3631 participants) in the review. All but two trials were at high risk of bias. A total of 33 different interventions, ranging from advice to supervised exercise and special diets, or a combination of these and no additional intervention were compared in these trials. The reference treatment was no active intervention. Twenty-eight trials (1942 participants) were included in one or more comparisons. The follow-up ranged from 1 month to 24 months. The remaining trials did not report any of the outcomes of interest for this review. The follow-up period in the trials that reported clinical outcomes was 2 months to 24 months. During this short follow-up period, clinical events related to NAFLD such as mortality, liver cirrhosis, liver decompensation, liver transplantation, hepatocellular carcinoma, and liver-related mortality were sparse. This is probably because of the very short follow-up periods. It takes a follow-up of 8 years to 28 years to detect differences in mortality between people with NAFLD and the general population. It is therefore unlikely that differences by clinical outcomes will be noted in trials with less than 5 years to 10 years of follow-up. In one trial, one participant developed an adverse event. There were no adverse events in any of the remaining participants in this trial, or in any of the remaining trials, which seemed to be directly related to the intervention. AUTHORS' CONCLUSIONS The evidence indicates considerable uncertainty about the effects of the lifestyle interventions compared with no additional intervention (to general public health advice) on any of the clinical outcomes after a short follow-up period of 2 months to 24 months in people with nonalcohol-related fatty liver disease. Accordingly, high-quality randomised clinical trials with adequate follow-up are needed. We propose registry-based randomised clinical trials or cohort multiple randomised clinical trials (a study design in which multiple interventions are trialed within large longitudinal cohorts of participants to gain efficiencies and align trials more closely to standard clinical practice), comparing aerobic exercise and dietary advice versus standard of care (exercise and dietary advice received as part of national health promotion). The reason for the choice of aerobic exercise and dietary advice is the impact of these interventions on indirect outcomes which may translate to clinical benefit. The outcomes in such trials should be mortality, health-related quality of life, decompensated liver cirrhosis, liver transplantation, and resource use measures including costs of intervention and decreased healthcare use after a minimum follow-up of eight years, to find meaningful differences in the clinically important outcomes.
Collapse
Affiliation(s)
- Elena Buzzetti
- Sheila Sherlock Liver Centre, Royal Free Hospital and the UCL Institute of Liver and Digestive Health, London, UK
| | - Audrey Linden
- Division of Surgery and Interventional Science, University College London, London, UK
| | - Lawrence Mj Best
- Division of Surgery and Interventional Science, University College London, London, UK
| | - Angela M Madden
- School of Life and Medical Sciences, University of Hertfordshire, Hatfield, UK
| | - Danielle Roberts
- Division of Surgery and Interventional Science, University College London, London, UK
| | - Thomas J G Chase
- Department of General Surgery, Homerton University Hospital NHS Foundation Trust, London, UK
| | - Suzanne C Freeman
- Department of Health Sciences, University of Leicester, Leicester, UK
| | - Nicola J Cooper
- Department of Health Sciences, University of Leicester, Leicester, UK
| | - Alex J Sutton
- Department of Health Sciences, University of Leicester, Leicester, UK
| | | | | | - Kathy Wright
- Cochrane Hepato-Biliary Group, Copenhagen Trial Unit, Centre for Clinical Intervention Research, The Capital Region of Denmark, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Chavdar S Pavlov
- Department of Therapy, I.M. Sechenov First Moscow State Medical University, Moscow, Russian Federation
| | - Brian R Davidson
- Division of Surgery and Interventional Science, University College London, London, UK
| | - Emmanuel Tsochatzis
- Sheila Sherlock Liver Centre, Royal Free Hospital and the UCL Institute of Liver and Digestive Health, London, UK
| | - Kurinchi Selvan Gurusamy
- Division of Surgery and Interventional Science, University College London, London, UK
- Department of Therapy, I.M. Sechenov First Moscow State Medical University, Moscow, Russian Federation
| |
Collapse
|
9
|
Frohlich J, Mazza T, Sobolewski C, Foti M, Vinciguerra M. GDF11 rapidly increases lipid accumulation in liver cancer cells through ALK5-dependent signaling. Biochim Biophys Acta Mol Cell Biol Lipids 2021; 1866:158920. [PMID: 33684566 DOI: 10.1016/j.bbalip.2021.158920] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 02/22/2021] [Accepted: 03/03/2021] [Indexed: 02/06/2023]
Abstract
Hepatocellular carcinoma (HCC) is one of the fastest-growing causes of cancer-related mortalities worldwide and this trend is mimicked by the surge of non-alcoholic fatty liver disease (NAFLD). Altered hepatic lipid metabolism promotes HCC development through inflammation and activation of oncogenes. GDF11 is a member of the TGF-β superfamily and recent data have implicated GDF11 as an anti-aging factor that can alleviate high-fat diet induced obesity, hyperglycemia, insulin resistance and NAFLD. However, its role in hepatic lipid metabolism is still not fully delineated. The aim of the present study was to characterize the role of GDF11 in hepatic and HCC cells lipid accumulation. To achieve this, we performed imaging, biochemical, lipidomic, and transcriptomic analyses in primary hepatocytes and in HCC cells treated with GDF11 to study the GDF11-activated signaling pathways. GDF11 treatment rapidly triggered ALK5-dependent SMAD2/3 nuclear translocation and elevated lipid droplets in HCC cells, but not in primary hepatocytes. In HCC cells, ALK5 inhibition hampered GDF11-mediated SMAD2/3 signaling and attenuated lipid accumulation. Using ultra-high-performance liquid chromatography/mass spectrometry, we detected increased accumulation of longer acyl-chain di/tri-acylglycerols and glycerophospholipids. Unbiased transcriptomic analysis identified TGF-β and PI3K-AKT signaling among the top pathways/cellular processes activated in GDF11 treated HCC cells. In summary, GDF11 supplementation promotes pro-lipogenic gene expression and lipid accumulation in HCC cells. Integration of our "omics" data pointed to a GDF11-induced upregulation of de novo lipogenesis through activation of ALK5/SMAD2/3/PI3K-AKT pathways. Thus, GDF11 could contribute to metabolic reprogramming and dysregulation of lipid metabolism in HCC cells, without effects on healthy hepatocytes.
Collapse
Affiliation(s)
- Jan Frohlich
- International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic
| | - Tommaso Mazza
- Bioinformatics Unit, IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Cyril Sobolewski
- Department of Cell Physiology & Metabolism and Translational Research Centre in Onco-haematology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Michelangelo Foti
- Department of Cell Physiology & Metabolism and Translational Research Centre in Onco-haematology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Manlio Vinciguerra
- International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic; Department of Translational Stem Cell Biology, Medical University of Varna, Varna, Bulgaria; Institute of Liver and Digestive Health, Division of Medicine, University College London (UCL), London, United Kingdom.
| |
Collapse
|
10
|
Long MT, Zhang X, Xu H, Liu CT, Corey KE, Chung RT, Loomba R, Benjamin EJ. Hepatic Fibrosis Associates With Multiple Cardiometabolic Disease Risk Factors: The Framingham Heart Study. Hepatology 2021; 73:548-559. [PMID: 33125745 PMCID: PMC8515503 DOI: 10.1002/hep.31608] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 09/21/2020] [Accepted: 10/06/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND AND AIMS NAFLD is increasing in prevalence and will soon be the most common chronic liver disease. Liver stiffness, as assessed by vibration-controlled transient elastography (VCTE), correlates with hepatic fibrosis, an important predictor of liver-related and all-cause mortality. Although liver fat is associated with cardiovascular risk factors, the association between hepatic fibrosis and cardiovascular risk factors is less clear. APPROACH AND RESULTS We performed VCTE, assessing controlled attenuation parameter (CAP; measure of steatosis) and liver stiffness measurement (LSM) in 3,276 Framingham Heart Study adult participants (53.9% women, mean age 54.3 ± 9.1 years) presenting for a routine study visit. We performed multivariable-adjusted logistic regression models to determine the association between LSM and obesity-related, vascular-related, glucose-related, and cholesterol-related cardiovascular risk factors. The prevalence of hepatic steatosis (CAP ≥ 290 dB/m) was 28.8%, and 8.8% had hepatic fibrosis (LSM ≥ 8.2 kPa). Hepatic fibrosis was associated with multiple cardiovascular risk factors, including increased odds of obesity (OR, 1.82; 95% CI, 1.35-2.47), metabolic syndrome (OR, 1.49; 95% CI 1.10-2.01), diabetes (OR, 2.67; 95% CI, 1.21-3.75), hypertension (OR, 1.52; 95% CI, 1.15-1.99), and low high-density lipoprotein cholesterol (OR, 1.47; 95% CI, 1.09-1.98), after adjustment for age, sex, smoking status, alcohol drinks/week, physical activity index, aminotransferases, and CAP. CONCLUSIONS In our community-based cohort, VCTE-defined hepatic fibrosis was associated with multiple cardiovascular risk factors, including obesity, metabolic syndrome, diabetes, hypertension, and high-density lipoprotein cholesterol, even after accounting for covariates and CAP. Additional longitudinal studies are needed to determine if hepatic fibrosis contributes to incident cardiovascular disease risk factors or events.
Collapse
Affiliation(s)
- Michelle T Long
- Section of GastroenterologyBoston Medical CenterBoston University School of MedicineBostonMA
| | - Xiaoyu Zhang
- Department of BiostatisticsBoston UniversityBostonMA
| | - Hanfei Xu
- Department of BiostatisticsBoston UniversityBostonMA
| | - Ching-Ti Liu
- Department of BiostatisticsBoston UniversityBostonMA
| | - Kathleen E Corey
- Liver CenterGastroenterology DivisionDepartment of MedicineMassachusetts General HospitalHarvard Medical SchoolBostonMA
| | - Raymond T Chung
- Liver CenterGastroenterology DivisionDepartment of MedicineMassachusetts General HospitalHarvard Medical SchoolBostonMA
| | - Rohit Loomba
- Division of GastroenterologyDepartment of Medicine and Division of EpidemiologyDepartment of Family and PreventiveUniversity of California at San DiegoLa JollaCA
| | - Emelia J Benjamin
- Evans Department of MedicineWhitaker Cardiovascular Institute and Cardiology SectionBoston University School of MedicineBostonMA
| |
Collapse
|
11
|
Mirhafez SR, Azimi-Nezhad M, Dehabeh M, Hariri M, Naderan RD, Movahedi A, Abdalla M, Sathyapalan T, Sahebkar A. The Effect of Curcumin Phytosome on the Treatment of Patients with Non-alcoholic Fatty Liver Disease: A Double-Blind, Randomized, Placebo-Controlled Trial. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1308:25-35. [PMID: 33861434 DOI: 10.1007/978-3-030-64872-5_3] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a global health problem with increasing prevalence among overweight and obese patients. It is strongly associated with conditions of insulin resistance including type 2 diabetes mellitus (T2DM) and obesity. It has detrimental consequences ranged from simple steatosis to irreversible hepatic fibrosis and cirrhosis. Curcumin is a dietary polyphenol with potential effect in improving NAFLD. Therefore, the aim of this trial was to examine the effect of curcumin supplementation on various aspects of NAFLD. In this trial, a total number of 80 patients were randomised to receive either curcumin at 250 mg daily or placebo for 2 months. Lipid profiles, hepatic enzymes, anthropometric indices and hepatic fat mass were assessed at the baseline and the end of the trial, and compared within the groups. The grade of hepatic steatosis, and serum aspartate aminotransferase (AST) levels were significantly reduced in the curcumin group (p = 0.015 and p = 0.007, respectively) compared to the placebo. There was also a significant reduction in high density lipoprotein (HDL) levels and anthropometric indices in both groups with no significant differences between the two groups. Low dose phospholipid curcumin supplementation each day for 2 months showed significant reduction in hepatic steatosis and enzymes in patients with NAFLD compared to placebo. Further studies of longer duration and higher dosages are needed to assess its effect on other parameters of NAFLD including cardiovascular risk.
Collapse
Affiliation(s)
- Seyed Reza Mirhafez
- Noncommunicable Diseases Research Center, Neyshabur University of Medical Sciences, Neyshabur, Iran.
| | - Mohsen Azimi-Nezhad
- Noncommunicable Diseases Research Center, Neyshabur University of Medical Sciences, Neyshabur, Iran
| | - Maryam Dehabeh
- Noncommunicable Diseases Research Center, Neyshabur University of Medical Sciences, Neyshabur, Iran
| | - Mitra Hariri
- Noncommunicable Diseases Research Center, Neyshabur University of Medical Sciences, Neyshabur, Iran
| | - Ronika Danesh Naderan
- Noncommunicable Diseases Research Center, Neyshabur University of Medical Sciences, Neyshabur, Iran
| | - Ali Movahedi
- Department of Anesthesia and Operating Room Nursing, Neyshabur University of Medical Sciences, Neyshabur, Iran
| | - Mohammed Abdalla
- Academic Diabetes, Endocrinology and Metabolism, Hull York Medical School, University of Hull, Hull, UK
| | - Thozhukat Sathyapalan
- Academic Diabetes, Endocrinology and Metabolism, Hull York Medical School, University of Hull, Hull, UK
| | - Amirhossein Sahebkar
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
- School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
- Polish Mother's Memorial Hospital Research Institute (PMMHRI), Lodz, Poland.
| |
Collapse
|
12
|
Exploring the Impact of Obesity on Health Care Resources and Coding in the Acute Hospital Setting: A Feasibility Study. Healthcare (Basel) 2020; 8:healthcare8040459. [PMID: 33158275 PMCID: PMC7711616 DOI: 10.3390/healthcare8040459] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 10/22/2020] [Accepted: 11/03/2020] [Indexed: 12/03/2022] Open
Abstract
Obesity is costly, yet there have been few attempts to estimate the actual costs of providing hospital care to the obese inpatient. This study aimed to test the feasibility of measuring obesity-related health care costs and accuracy of coding data for acute inpatients. A prospective observational study was conducted over three weeks in June 2018 in a single orthopaedic ward of a metropolitan tertiary hospital in Queensland, Australia. Demographic data, anthropometric measurements, clinical characteristics, cost of hospital encounter and coding data were collected. Complete demographic, anthropometric and clinical data were collected for all 18 participants. Hospital costing reports and coding data were not available within the study timeframe. Participant recruitment and data collection were resource-intensive, with mobility assistance required to obtain anthropometric measurements in more than half of the participants. Greater staff time and costs were seen in participants with obesity compared to those without obesity (obesity: body mass index ≥ 30), though large standard deviations indicate wide variance. Data collected suggest that obesity-related cost and resource use amongst acute inpatients require further exploration. This study provides recommendations for protocol refinement to improve the accuracy of data collected for future studies measuring the actual cost of providing hospital care to obese inpatients.
Collapse
|
13
|
Frohlich J, Kovacovicova K, Mazza T, Emma MR, Cabibi D, Foti M, Sobolewski C, Oben JA, Peyrou M, Villarroya F, Soresi M, Rezzani R, Cervello M, Bonomini F, Alisi A, Vinciguerra M. GDF11 induces mild hepatic fibrosis independent of metabolic health. Aging (Albany NY) 2020; 12:20024-20046. [PMID: 33126224 PMCID: PMC7655202 DOI: 10.18632/aging.104182] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 08/25/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS Growth Differentiation Factor 11 (GDF11) is an anti-aging factor, yet its role in liver diseases is not established. We evaluated the role of GDF11 in healthy conditions and in the transition from non-alcoholic fatty liver disease (NAFLD) to non-alcoholic steatohepatitis (NASH). RESULTS GDF11 mRNA levels positively correlated with NAFLD activity score and with CPT1, SREBP, PPARγ and Col1A1 mRNA levels, and associated to portal fibrosis, in morbidly obese patients with NAFLD/NASH. GDF11-treated mice showed mildly exacerbated hepatic collagen deposition, accompanied by weight loss and without changes in liver steatosis or inflammation. GDF11 triggered ALK5-dependent SMAD2/3 nuclear translocation and the pro-fibrogenic activation of HSC. CONCLUSIONS GDF11 supplementation promotes mild liver fibrosis. Even considering its beneficial metabolic effects, caution should be taken when considering therapeutics that regulate GDF11. METHODS We analyzed liver biopsies from a cohort of 33 morbidly obese adults with NAFLD/NASH. We determined the correlations in mRNA expression levels between GDF11 and genes involved in NAFLD-to-NASH progression and with pathological features. We also exposed wild type or obese mice with NAFLD to recombinant GDF11 by daily intra-peritoneal injection and monitor the hepatic pathological changes. Finally, we analyzed GDF11-activated signaling pathways in hepatic stellate cells (HSC).
Collapse
Affiliation(s)
- Jan Frohlich
- International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic
| | - Kristina Kovacovicova
- International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic
| | - Tommaso Mazza
- Bioinformatics Unit, IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Maria R. Emma
- Institute for Biomedical Research and Innovation, National Research Council (CNR), Palermo, Italy
| | - Daniela Cabibi
- Department of Health Promotion Sciences, Maternal and Infantile Care, Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy
| | - Michelangelo Foti
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Cyril Sobolewski
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Jude A. Oben
- Institute for Liver and Digestive Health, Division of Medicine, University College London (UCL), London, United Kingdom
| | - Marion Peyrou
- Department of Biochemistry and Molecular Biomedicine, Institute of Biomedicine of the University of Barcelona, Barcelona, Catalonia, Spain
- Institut de Recerca Hospital de la Santa Creu i Sant Pau, Barcelona, Catalonia, Spain
| | - Francesc Villarroya
- Department of Biochemistry and Molecular Biomedicine, Institute of Biomedicine of the University of Barcelona, Barcelona, Catalonia, Spain
- Institut de Recerca Hospital de la Santa Creu i Sant Pau, Barcelona, Catalonia, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición, Barcelona, Catalonia, Spain
| | - Maurizio Soresi
- Department of Health Promotion Sciences, Maternal and Infantile Care, Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy
| | - Rita Rezzani
- Anatomy and Physiopathology Division, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
- Interdepartmental University Center of Research “Adaption and Regeneration of Tissues and Organs-(ARTO)”, University of Brescia, Brescia, Italy
| | - Melchiorre Cervello
- Institute for Biomedical Research and Innovation, National Research Council (CNR), Palermo, Italy
| | - Francesca Bonomini
- Anatomy and Physiopathology Division, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
- Interdepartmental University Center of Research “Adaption and Regeneration of Tissues and Organs-(ARTO)”, University of Brescia, Brescia, Italy
| | - Anna Alisi
- Research Area for Multifactorial Diseases, Research Unit of Molecular Genetics of Complex Phenotypes, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Manlio Vinciguerra
- International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic
- Institute for Liver and Digestive Health, Division of Medicine, University College London (UCL), London, United Kingdom
| |
Collapse
|
14
|
Witjes JJ, Smits LP, Pekmez CT, Prodan A, Meijnikman AS, Troelstra MA, Bouter KEC, Herrema H, Levin E, Holleboom AG, Winkelmeijer M, Beuers UH, van Lienden K, Aron-Wisnewky J, Mannisto V, Bergman JJ, Runge JH, Nederveen AJ, Dragsted LO, Konstanti P, Zoetendal EG, de Vos W, Verheij J, Groen AK, Nieuwdorp M. Donor Fecal Microbiota Transplantation Alters Gut Microbiota and Metabolites in Obese Individuals With Steatohepatitis. Hepatol Commun 2020; 4:1578-1590. [PMID: 33163830 PMCID: PMC7603524 DOI: 10.1002/hep4.1601] [Citation(s) in RCA: 95] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 07/27/2020] [Accepted: 07/31/2020] [Indexed: 12/11/2022] Open
Abstract
The intestinal microbiota has been linked to the development and prevalence of steatohepatitis in humans. Interestingly, steatohepatitis is significantly lower in individuals taking a plant-based, low-animal-protein diet, which is thought to be mediated by gut microbiota. However, data on causality between these observations in humans is scarce. In this regard, fecal microbiota transplantation (FMT) using healthy donors is safe and is capable of changing microbial composition in human disease. We therefore performed a double-blind randomized controlled proof-of-principle study in which individuals with hepatic steatosis on ultrasound were randomized to two study arms: lean vegan donor (allogenic n = 10) or own (autologous n = 11) FMT. Both were performed three times at 8-week intervals. A liver biopsy was performed at baseline and after 24 weeks in every subject to determine histopathology (Nonalcoholic Steatohepatitis Clinical Research Network) classification and changes in hepatic gene expression based on RNA sequencing. Secondary outcome parameters were changes in intestinal microbiota composition and fasting plasma metabolomics. We observed a trend toward improved necro-inflammatory histology, and found significant changes in expression of hepatic genes involved in inflammation and lipid metabolism following allogenic FMT. Intestinal microbial community structure changed following allogenic FMT, which was associated with changes in plasma metabolites as well as markers of . Conclusion: Allogenic FMT using lean vegan donors in individuals with hepatic steatosis shows an effect on intestinal microbiota composition, which is associated with beneficial changes in plasma metabolites and markers of steatohepatitis.
Collapse
Affiliation(s)
- Julia J Witjes
- Department of Internal and Vascular Medicine Amsterdam University Medical Centers Location AMC Amsterdam the Netherlands
| | - Loek P Smits
- Department of Internal and Vascular Medicine Amsterdam University Medical Centers Location AMC Amsterdam the Netherlands
| | - Ceyda T Pekmez
- Department of Nutrition, Exercise and Sports University of Copenhagen Copenhagen Denmark
| | - Andrei Prodan
- Department of Internal and Vascular Medicine Amsterdam University Medical Centers Location AMC Amsterdam the Netherlands
| | - Abraham S Meijnikman
- Department of Internal and Vascular Medicine Amsterdam University Medical Centers Location AMC Amsterdam the Netherlands
| | - Marian A Troelstra
- Department of Radiology & Nuclear Medicine Amsterdam University Medical Centers Location AMC Amsterdam the Netherlands
| | - Kristien E C Bouter
- Department of Internal and Vascular Medicine Amsterdam University Medical Centers Location AMC Amsterdam the Netherlands
| | - Hilde Herrema
- Department of Internal and Vascular Medicine Amsterdam University Medical Centers Location AMC Amsterdam the Netherlands
| | - Evgeni Levin
- Department of Internal and Vascular Medicine Amsterdam University Medical Centers Location AMC Amsterdam the Netherlands
| | - Adriaan G Holleboom
- Department of Internal and Vascular Medicine Amsterdam University Medical Centers Location AMC Amsterdam the Netherlands
| | - Maaike Winkelmeijer
- Department of Internal and Vascular Medicine Amsterdam University Medical Centers Location AMC Amsterdam the Netherlands
| | - Ulrich H Beuers
- Department of Gastroenterology and Hepatology Amsterdam University Medical Centers Location AMC Amsterdam the Netherlands
| | - Krijn van Lienden
- Department of Radiology & Nuclear Medicine Amsterdam University Medical Centers Location AMC Amsterdam the Netherlands
| | - Judith Aron-Wisnewky
- Department of Internal and Vascular Medicine Amsterdam University Medical Centers Location AMC Amsterdam the Netherlands
| | - Ville Mannisto
- Department of Internal and Vascular Medicine Amsterdam University Medical Centers Location AMC Amsterdam the Netherlands
| | - Jacques J Bergman
- Department of Gastroenterology and Hepatology Amsterdam University Medical Centers Location AMC Amsterdam the Netherlands
| | - Jurgen H Runge
- Department of Radiology & Nuclear Medicine Amsterdam University Medical Centers Location AMC Amsterdam the Netherlands
| | - Aart J Nederveen
- Department of Radiology & Nuclear Medicine Amsterdam University Medical Centers Location AMC Amsterdam the Netherlands
| | - Lars O Dragsted
- Department of Nutrition, Exercise and Sports University of Copenhagen Copenhagen Denmark
| | - Prokopis Konstanti
- Laboratory of Microbiology Wageningen University Wageningen the Netherlands
| | - Erwin G Zoetendal
- Laboratory of Microbiology Wageningen University Wageningen the Netherlands
| | - Willem de Vos
- Laboratory of Microbiology Wageningen University Wageningen the Netherlands.,Faculty of Medicine Human Microbiome Research Program University of Helsinki Finland
| | - Joanne Verheij
- Department of Pathology Amsterdam University Medical Centers Location AMC Amsterdam the Netherlands
| | - Albert K Groen
- Department of Internal and Vascular Medicine Amsterdam University Medical Centers Location AMC Amsterdam the Netherlands.,Department of Laboratory Medicine University of Groningen University Medical Center Groningen the Netherlands
| | - Max Nieuwdorp
- Department of Internal and Vascular Medicine Amsterdam University Medical Centers Location AMC Amsterdam the Netherlands
| |
Collapse
|
15
|
Keramida G, Peters AM. FDG PET/CT of the non‐malignant liver in an increasingly obese world population. Clin Physiol Funct Imaging 2020; 40:304-319. [DOI: 10.1111/cpf.12651] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 05/11/2020] [Accepted: 06/04/2020] [Indexed: 12/19/2022]
Affiliation(s)
- Georgia Keramida
- Department of Nuclear Medicine Royal Brompton and HarefieldNHS Foundation Trust London UK
| | - A. Michael Peters
- Department of Nuclear Medicine King’s College HospitalNHS Foundation Trusts London UK
| |
Collapse
|
16
|
Pattison RJ, Esteban JP, Sempokuya T, Kewcharoen J, Kalathil S, Kuwada SK. Nonalcoholic Fatty Liver Disease: An Important Consideration for Primary Care Providers in Hawai'i. HAWAI'I JOURNAL OF HEALTH & SOCIAL WELFARE 2020; 79:180-186. [PMID: 32524096 PMCID: PMC7281344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is the most common cause of chronic liver disease worldwide. NAFLD is a broad term for both non-alcoholic fatty liver (NAFL), which describes simple fatty liver without inflammation, and non-alcoholic steatohepatitis (NASH), the more severe phenotype with hepatocellular inflammation. The population of Hawai'i is particularly vulnerable to the NAFLD and obesity epidemics due to its large proportions of high-risk ethnic minorities exposed to varying degrees of westernization. Unfortunately, primary care providers (PCPs) often face a lack of awareness on the diagnosis and disease spectrum of NAFLD. Early initiation of treatment for NAFLD is crucial to slow its progression and prevent liver-related morbidity and mortality. This review aims to raise awareness for NAFLD among PCPs in Hawai'i by summarizing the disease's epidemiology, diagnosis, and treatment. The diagnostic workup of NAFLD in the primary care setting involves exclusion of other liver disease etiologies and staging assessment of fibrosis and steatosis through non-invasive means such as serum biomarkers or elastography. Patients with overt signs and symptoms of cirrhosis or a high likelihood of advanced hepatic fibrosis should be referred to liver disease specialists. The role of PCPs in NAFLD management involves facilitating weight loss through therapeutic lifestyle modifications and treatment of comorbid cardiovascular conditions. Evidence-based pharmacologic therapies for NAFLD are available, such as vitamin E and pioglitazone, with more currently in development.
Collapse
Affiliation(s)
- Robert J. Pattison
- Internal Medicine Residency Program, John A. Burns School of Medicine University of Hawai‘i, Honolulu, HI (RJP, JK)
| | | | | | - Jakrin Kewcharoen
- Internal Medicine Residency Program, John A. Burns School of Medicine University of Hawai‘i, Honolulu, HI (RJP, JK)
| | | | - Scott K. Kuwada
- John A. Burns School of Medicine University of Hawai‘i, Honolulu, HI (SKK)
| |
Collapse
|
17
|
Zheng X, Zhao MG, Jiang CH, Sheng XP, Yang HM, Liu Y, Yao XM, Zhang J, Yin ZQ. Triterpenic acids-enriched fraction from Cyclocarya paliurus attenuates insulin resistance and hepatic steatosis via PI3K/Akt/GSK3β pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2020; 66:153130. [PMID: 31790897 DOI: 10.1016/j.phymed.2019.153130] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 10/16/2019] [Accepted: 10/29/2019] [Indexed: 06/10/2023]
Abstract
BACKGROUND Non-alcoholic fatty liver disease (NAFLD) is the most prevalent form of chronic liver diseases. Cyclocarya paliurus (C. paliurus), an edible and medicinal plant in Chinese folk, has been demonstrated to ameliorate diabetes, obesity and lipid metabolism disorders. However, its effects on NAFLD and its potential molecular mechanism have not been clearly expounded. PURPOSE The present study was designed to explore the therapeutic potential of triterpenic acids-enriched fraction from C. paliurus (CPT), as well as its underlying mechanism in vivo and in vitro models of NAFLD. METHODS The metabolic effects and possible molecular mechanism of CPT were examined using HepG2 cells and primary hepatocytes (isolated from C57BL/6 J mice) models of fatty liver induced by palmitic acid (PA) and a high fat diet mouse model. RESULTS In high fat diet-induced C57BL/6 J mice, CPT significantly reduced liver weight index, serum alanine transaminase (ALT), aspartate transaminase (AST), triacylglycerol (TG), total cholesterol (TC) and hepatic TG, TC levels. Moreover, CPT dramatically decreased the contents of blood glucose, insulin, and insulin resistance (HOMA-IR) index. Meanwhile, CPT significantly increased the tyrosine phosphorylation level of IRS and the uptake of 2-deoxyglucose (2DG) in PA-induced HepG2 cells and primary hepatocytes fatty liver models. Furthermore, in PA-induced HepG2 cells and primary hepatocytes, CPT significantly decreased the number of lipid droplets and intracellular TG content. In addition, mechanism investigation showed that CPT increased the phosphorylation of phosphoinositide 3-kinase (PI3K), protein kinase B (Akt) and glycogen synthase-3β (GSK3β) in vivo and in vitro models, which were abrogated by PI3K inhibitor LY294002 in vitro models. CONCLUSION These findings indicate that CPT may exert the therapeutic effects on NAFLD via regulating PI3K/Akt/GSK3β pathway.
Collapse
Affiliation(s)
- Xian Zheng
- Department of TCMs Pharmaceuticals & State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China; Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210009, China; Laboratories of Translational Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210009, China
| | - Meng-Ge Zhao
- Department of TCMs Pharmaceuticals & State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China; Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210009, China; Laboratories of Translational Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210009, China
| | - Cui-Hua Jiang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210009, China; Laboratories of Translational Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210009, China
| | - Xue-Ping Sheng
- Department of TCMs Pharmaceuticals & State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China; Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210009, China; Laboratories of Translational Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210009, China
| | - Hui-Min Yang
- Department of TCMs Pharmaceuticals & State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China; Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210009, China; Laboratories of Translational Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210009, China
| | - Yao Liu
- Department of TCMs Pharmaceuticals & State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China; Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210009, China; Laboratories of Translational Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210009, China
| | - Xiao-Ming Yao
- Clinical Laboratory, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, Jiangsu 210028, China
| | - Jian Zhang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210009, China; Laboratories of Translational Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210009, China.
| | - Zhi-Qi Yin
- Department of TCMs Pharmaceuticals & State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
18
|
Relationship between regional hepatic glucose metabolism and regional distribution of hepatic fat. Nucl Med Commun 2019; 40:212-218. [DOI: 10.1097/mnm.0000000000000968] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
|
19
|
Gurusamy KS, Tsochatzis E, Madden AM. Nutritional supplementation for non-alcohol-related fatty liver disease: a network meta-analysis. Cochrane Database Syst Rev 2018. [DOI: 10.1002/14651858.cd013157] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Kurinchi Selvan Gurusamy
- Royal Free Campus, UCL Medical School; Department of Surgery; Royal Free Hospital Rowland Hill Street London UK NW3 2PF
| | - Emmanuel Tsochatzis
- Royal Free Hospital and the UCL Institute of Liver and Digestive Health; Sheila Sherlock Liver Centre; Pond Street London UK NW3 2QG
| | - Angela M Madden
- University of Hertfordshire; Biological & Environmental Sciences; College Lane Hatfield Hertfordshire UK AL10 9AB
| |
Collapse
|
20
|
The potential association between periodontitis and non-alcoholic fatty liver disease: a systematic review. Clin Oral Investig 2018; 22:2965-2974. [DOI: 10.1007/s00784-018-2726-1] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 10/18/2018] [Indexed: 02/06/2023]
|
21
|
Gurusamy KS, Tsochatzis E, Madden AM. Lifestyle modifications for non-alcohol related fatty liver disease: a network meta-analysis. Hippokratia 2018. [DOI: 10.1002/14651858.cd013156] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Kurinchi Selvan Gurusamy
- Royal Free Campus, UCL Medical School; Department of Surgery; Royal Free Hospital Rowland Hill Street London UK NW3 2PF
| | - Emmanuel Tsochatzis
- Royal Free Hospital and the UCL Institute of Liver and Digestive Health; Sheila Sherlock Liver Centre; Pond Street London UK NW3 2QG
| | - Angela M Madden
- University of Hertfordshire; Biological & Environmental Sciences; College Lane Hatfield Hertfordshire UK AL10 9AB
| |
Collapse
|
22
|
Fujita M, Kuraji R, Ito H, Hashimoto S, Toen T, Fukada T, Numabe Y. Histological effects and pharmacokinetics of lipopolysaccharide derived from Porphyromonas gingivalis on rat maxilla and liver concerning with progression into non-alcoholic steatohepatitis. J Periodontol 2018; 89:1101-1111. [PMID: 29799627 DOI: 10.1002/jper.17-0678] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Revised: 04/10/2018] [Accepted: 04/29/2018] [Indexed: 11/10/2022]
Abstract
BACKGROUND Non-alcoholic steatohepatitis (NASH) is one of the chronic liver diseases that can develop into hepatocirrhosis. The purpose of the present study was to investigate the impact of lipopolysaccharide (LPS) from Porphyromonas gingivalis (P. gingivalis) on NASH onset, and to determine the biodistribution of double-radiolabeled LPS (R-LPS) biosynthesized in P. gingivalis. METHODS Rats fed a basal diet (BD) or a high-fat diet (HD) were injected with P. gingivalis-LPS or R-LPS into the palatine gingiva around the right maxillary first molar, and were classified into the following 4 groups: BD/LPS (-), BD/LPS (+), HD/LPS (-), and HD/LPS (+) or 2 groups: BD/R-LPS and HD/R-LPS. RESULTS Inflammation in the gingiva of the LPS (+) groups progressed significantly more than the LPS (-) groups. Furthermore, in the HD/LPS (+) liver, histologic analysis confirmed the presence of NASH, characterized by large fat droplets, ballooning degeneration, and infiltration of inflammatory cells. When 3 H, 14 C-R-LPS was injected into the palatine gingiva, radioactivity in the right palatal mucosa of HD/R-LPS rats was the highest in comparison with other regions and was significantly elevated after 24 hours compared to BD/R-LPS rats. Autoradiographic analysis of the maxilla showed distributions from the palatal mucosa to the hard palate and the interdental region. Radioactivity in organs of both BD/R-LPS and HD/R-LPS rats were mostly localized to the liver even after 24 hours. CONCLUSION The present study suggests that the transfer of P. gingivalis-LPS from the oral cavity to the liver plays an important role in disease exacerbation of NASH.
Collapse
Affiliation(s)
- Miyako Fujita
- Department of Periodontology, The Nippon Dental University School of Life Dentistry at Tokyo, Tokyo, Japan
| | - Ryutaro Kuraji
- Department of Periodontology, The Nippon Dental University School of Life Dentistry at Tokyo, Tokyo, Japan.,Department of Life Science Dentistry, The Nippon Dental University, Tokyo, Japan
| | - Hiroshi Ito
- Department of Periodontology, The Nippon Dental University School of Life Dentistry at Tokyo, Tokyo, Japan
| | | | - Toshiyuki Toen
- Dental Research Institute, Radio Isotope Center, The Nippon Dental University School of Life Dentistry at Tokyo, Tokyo, Japan
| | - Tetsuya Fukada
- Dental Research Institute, Radio Isotope Center, The Nippon Dental University School of Life Dentistry at Tokyo, Tokyo, Japan
| | - Yukihiro Numabe
- Department of Periodontology, The Nippon Dental University School of Life Dentistry at Tokyo, Tokyo, Japan
| |
Collapse
|
23
|
Bratoeva K, Nikolova S, Merdzhanova A, Stoyanov GS, Dimitrova E, Kashlov J, Conev N, Radanova M. Association Between Serum CK-18 Levels and the Degree of Liver Damage in Fructose-Induced Metabolic Syndrome. Metab Syndr Relat Disord 2018; 16:350-357. [PMID: 29989845 DOI: 10.1089/met.2017.0162] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The pathogenesis of nonalcoholic fatty liver disease as a component of metabolic syndrome (MetS) involves the activation of apoptosis in steatotic hepatocytes. Caspase-generated fragments such as cytokeratin-18 (CK-18) in patients with various hepatic impairments are investigated as markers for diagnosis and assessment of disease severity. The goal of the study was to capture early biomarkers of apoptosis and elucidate their role in assessing the presence and extent of hepatic damage in a MetS model. MATERIALS AND METHODS We used male Wistar rats, divided into two groups (n = 7): control and high-fructose drinking (HFD) (35% fructose corn syrup for 16 weeks). Metabolic disorders and liver damage were studied by histochemistry (hematoxylin and eosin), immunohistochemical, immunological, and biochemical testing. RESULTS Our results showed significant increase in liver and serum levels of CK-18 and pro/antiapoptotic Bax/Bcl2 ratio, and decreased levels of HMGB1 (marker of necrosis) in the HFD group when compared with the control. All HFD rats developed obesity, hyperglycemia, hepatomegaly, microvesicular steatosis, an imbalance in hepatic antioxidative defense by measuring malondialdehyde and sulfhydryl groups (SH) with no inflammation and fibrosis, elevated serum levels of triglycerides, tumor necrosis factor alpha (TNF-α), and C-reactive protein without changes in serum aminotransferase levels relative to the control group. As a result of the applied regression analysis, we have determined that the variables TNF-α (0.92) and SH (0.659) have a strong complex effect on hepatic CK-18 levels with predicted value of the model R = 0.9. CONCLUSION The elevated CK-18 serum levels in the HFD group and their association with the histological changes in the liver and biochemical indicators demonstrate the key role of apoptosis in the pathogenesis of HFD-induced liver damage and the reliability of CK-18 as a biomarker for noninvasive assessment of liver damages in MetS.
Collapse
Affiliation(s)
- Kameliya Bratoeva
- 1 Division of Pathophysiology, Department of Physiology and Pathophysiology, Faculty of Medicine, Medical University of Varna , Varna, Bulgaria
| | - Silviya Nikolova
- 2 Department of Social Medicine and Healthcare Organization, Faculty of Public Health, Medical University of Varna , Varna, Bulgaria
| | - Albena Merdzhanova
- 3 Department of Chemistry, Faculty of Pharmacy, Medical University of Varna , Varna, Bulgaria
| | - George St Stoyanov
- 4 Department of Anatomy and Cell Biology, Faculty of Medicine, Medical University of Varna , Varna, Bulgaria
| | - Eleonora Dimitrova
- 5 Department of Propedeutics of Internal Diseases, Faculty of Medicine, Medical University of Varna , Varna, Bulgaria
| | - Javor Kashlov
- 5 Department of Propedeutics of Internal Diseases, Faculty of Medicine, Medical University of Varna , Varna, Bulgaria
| | - Nikolay Conev
- 5 Department of Propedeutics of Internal Diseases, Faculty of Medicine, Medical University of Varna , Varna, Bulgaria
| | - Mariya Radanova
- 6 Department of Biochemistry, Molecular Medicine and Nutrigenomics, Faculty of Pharmacy, Medical University of Varna , Varna, Bulgaria
| |
Collapse
|
24
|
Bajantri B, Lvovsky D. A Case of Concomitant Obstructive Sleep Apnea and Non-Alcoholic Steatohepatitis Treated With CPAP Therapy. Gastroenterology Res 2018; 11:252-259. [PMID: 29915639 PMCID: PMC5997478 DOI: 10.14740/gr1033w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Accepted: 05/09/2018] [Indexed: 12/12/2022] Open
Abstract
Obstructive sleep apnea syndrome is a disorder of sleep breathing that is a result of recurrent and intermittent hypoxia during sleep induced by the repeated partial or complete collapse of the upper airway, eventually causing chronic intermittent hypoxia. Non-alcoholic fatty liver disease is divided into non-alcoholic fatty liver and non-alcoholic steatohepatitis. Animal and human studies showed that obesity is associated with chronic liver hypoxia, even in the presence of systemic normoxia causing inflammation and release of cytokines. A "two-hit" model has been proposed. The first hit is characterized by insulin resistance and excess hepatic lipid accumulation secondary to abnormal fatty acid metabolism. Oxidative stress and inflammation are thought to comprise the second hit. Gold standard for the diagnosis of non-alcoholic steatohepatitis is a liver biopsy. Many clinical scores and non-invasive tools are used for the diagnosis of non-alcoholic steatohepatitis. Conservative management with lifestyle modifications including diet, exercise and weight loss remains the therapy of choice today. We present a case report of a 39-year-old man who was diagnosed with concomitant non-alcoholic steatohepatitis and severe obstructive sleep apnea. He was started treatment with continuous positive airway pressure and demonstrated excellent adherence to therapy for 6 years, with concomitant obstructive sleep apnea and non-alcoholic steatohepatitis which reversed with prolonged optimal continuous positive airway pressure therapy. Physical examination remained unremarkable except for morbid obesity. His abdominal girth, as well as body mass index, remained unchanged. After 6 years of optimal continuous positive airway pressure therapy, liver enzymes and relevant lipid panel normalized, suggesting reversal of non-alcoholic steatohepatitis.
Collapse
Affiliation(s)
- Bharat Bajantri
- Division of Pulmonary Critical Care, Department of Medicine, Bronx Care Health System, Bronx, NY 10457, USA
| | - Dmitry Lvovsky
- Division of Pulmonary Critical Care, Department of Medicine, Bronx Care Health System, Bronx, NY 10457, USA.,Division of Pulmonary and Critical Care Medicine, Bronx Lebanon Hospital Center, Bronx, NY 10457, USA
| |
Collapse
|
25
|
Alkhalidy H, Wang Y, Liu D. Dietary Flavonoids in the Prevention of T2D: An Overview. Nutrients 2018; 10:nu10040438. [PMID: 29614722 PMCID: PMC5946223 DOI: 10.3390/nu10040438] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 03/15/2018] [Accepted: 03/29/2018] [Indexed: 12/16/2022] Open
Abstract
Type 2 diabetes (T2D) is a progressive metabolic disease that is increasing in prevalence globally. It is well established that insulin resistance (IR) and a progressive decline in functional β-cell mass are hallmarks of developing T2D. Obesity is a leading pathogenic factor for developing IR. Constant IR will progress to T2D when β-cells are unable to secret adequate amounts of insulin to compensate for decreased insulin sensitivity. Recently, a considerable amount of research has been devoted to identifying naturally occurring anti-diabetic compounds that are abundant in certain types of foods. Flavonoids are a group of polyphenols that have drawn great interest for their various health benefits. Results from many clinical and animal studies demonstrate that dietary intake of flavonoids might be helpful in preventing T2D, although cellular and molecular mechanisms underlying these effects are still not completely understood. This review discusses our current understanding of the pathophysiology of T2D and highlights the potential anti-diabetic effects of flavonoids and mechanisms of their actions.
Collapse
Affiliation(s)
- Hana Alkhalidy
- Department of Human Nutrition, Foods and Exercise, College of Agricultural and Life Sciences, Virginia Tech, Blacksburg, VA 24060, USA.
- Department of Nutrition and Food Technology, Faculty of Agriculture, Jordan University of Science and Technology, Irbid 22110, Jordan.
| | - Yao Wang
- Department of Human Nutrition, Foods and Exercise, College of Agricultural and Life Sciences, Virginia Tech, Blacksburg, VA 24060, USA.
| | - Dongmin Liu
- Department of Human Nutrition, Foods and Exercise, College of Agricultural and Life Sciences, Virginia Tech, Blacksburg, VA 24060, USA.
| |
Collapse
|
26
|
Lee SA, Lee SY, Choi YM, Kim H, Kim BJ. Sex disparity in viral load, inflammation and liver damage in transgenic mice carrying full hepatitis B virus genome with the W4P mutation in the preS1 region. World J Gastroenterol 2018; 24:1084-1092. [PMID: 29563753 PMCID: PMC5850128 DOI: 10.3748/wjg.v24.i10.1084] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Revised: 01/31/2018] [Accepted: 02/09/2018] [Indexed: 02/06/2023] Open
Abstract
AIM To study sex disparity in susceptibility to hepatocellular carcinoma (HCC), we created a transgenic mouse model that expressed the full hepatitis B virus (HBV) genome with the W4P mutation. METHODS Transgenic mice were generated by transferring the pHY92-1.1x-HBV-full genome plasmid (genotype A2) into C57Bl/6N mice. We compared serum levels of hepatitis B surface antigen (HBsAg), interleukin (IL)-6, and the liver enzymes alanine aminotransferase (ALT) and aspartate transaminase (AST), as well as liver histopathological features in male and female transgenic (W4P TG) mice and in nontransgenic littermates of 10 mo of age. RESULTS W4P TG males exhibited more pronounced hepatomegaly, significantly increased granule generation in liver tissue, elevated HBsAg expression in the liver and serum, and higher serum ALT and IL-6 levels compared to W4P TG females or littermate control groups. CONCLUSION Together, our data indicate that the W4P mutation in preS1 may contribute to sex disparity in susceptibility to HCC by causing increased HBV virion replication and enhanced IL-6-mediated inflammation in male individuals. Additionally, our transgenic mouse model that expresses full HBV genome with the W4P mutation in preS1 could be effectively used for the studies of the progression of liver diseases, including HCC.
Collapse
Affiliation(s)
- Seoung-Ae Lee
- Department of Microbiology and Immunology, Biomedical Sciences, Liver Research Institute and Cancer Research Institute, Seoul National University, College of Medicine, Seoul 110799, South Korea
| | - So-Young Lee
- Department of Microbiology and Immunology, Biomedical Sciences, Liver Research Institute and Cancer Research Institute, Seoul National University, College of Medicine, Seoul 110799, South Korea
| | - Yu-Min Choi
- Department of Microbiology and Immunology, Biomedical Sciences, Liver Research Institute and Cancer Research Institute, Seoul National University, College of Medicine, Seoul 110799, South Korea
| | - Hong Kim
- Department of Microbiology and Immunology, Biomedical Sciences, Liver Research Institute and Cancer Research Institute, Seoul National University, College of Medicine, Seoul 110799, South Korea
| | - Bum-Joon Kim
- Department of Microbiology and Immunology, Biomedical Sciences, Liver Research Institute and Cancer Research Institute, Seoul National University, College of Medicine, Seoul 110799, South Korea
| |
Collapse
|
27
|
Seko Y, Yamaguchi K, Mizuno N, Okuda K, Takemura M, Taketani H, Hara T, Umemura A, Nishikawa T, Moriguchi M, Yasui K, Kamaguchi M, Nishioji K, Mochizuki N, Kobayashi M, Mori K, Tanaka S, Matsuura K, Tanaka Y, Itoh Y. Combination of PNPLA3 and TLL1 polymorphism can predict advanced fibrosis in Japanese patients with nonalcoholic fatty liver disease. J Gastroenterol 2018; 53:438-448. [PMID: 28744823 DOI: 10.1007/s00535-017-1372-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Accepted: 07/19/2017] [Indexed: 02/04/2023]
Abstract
BACKGROUND Hepatic fibrosis is an independent risk factor for mortality and liver-related events in patients with nonalcoholic fatty liver disease (NAFLD). PNPLA3 rs738409 has been associated with fibrosis in viral and non-viral hepatitis. TLL1 rs17047200 also has been associated with developing hepatocellular carcinoma probably via hepatic fibrogenesis. We estimated the impact of these genetic polymorphisms on hepatic fibrosis in Japanese patients with NAFLD. METHODS We analyzed the association between these genetic variants and the backgrounds of 817 individuals who received health checkups (health check cohort) from 2012 to 2014. Then, we investigated the relationship between genetic variants and liver histology in 258 consecutive patients with biopsy-proven NAFLD in Japan (NAFLD cohort) from 2012 to 2017 (UMIN000027399). RESULTS The prevalence of PNPLA3 CG/GG in the NAFLD cohort was higher than that in the health check cohort (p < 0.001). The prevalence of patients with advanced fibrosis (stages 3-4) was higher for PNPLA3 genotype CG/GG than CC (p = 0.048) and for TLL1 genotype AT/TT than AA (p = 0.044). The high-risk group which had at least two risk alleles of these variants was more likely to have advanced fibrosis (p = 0.004). Multivariate analysis identified body mass index [odds ratio (OR) 1.123, serum AST (OR 1.037, p = 0.004], serum albumin (OR 0.247, p = 0.032), and genetic high risk (OR 2.632, p = 0.026) as predictors of advanced fibrosis. CONCLUSIONS In Japanese patients with NAFLD, individuals with risk alleles of PNPLA3 and TLL1 may have a risk of advanced fibrosis.
Collapse
Affiliation(s)
- Yuya Seko
- Department of Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyou-ku, Kyoto, 602-8566, Japan
| | - Kanji Yamaguchi
- Department of Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyou-ku, Kyoto, 602-8566, Japan
| | - Naoki Mizuno
- Department of Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyou-ku, Kyoto, 602-8566, Japan
| | - Keiichiro Okuda
- Department of Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyou-ku, Kyoto, 602-8566, Japan
| | - Masashi Takemura
- Department of Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyou-ku, Kyoto, 602-8566, Japan
| | - Hiroyoshi Taketani
- Department of Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyou-ku, Kyoto, 602-8566, Japan
| | - Tasuku Hara
- Department of Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyou-ku, Kyoto, 602-8566, Japan
| | - Atsushi Umemura
- Department of Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyou-ku, Kyoto, 602-8566, Japan
| | - Taichiro Nishikawa
- Department of Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyou-ku, Kyoto, 602-8566, Japan
| | - Michihisa Moriguchi
- Department of Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyou-ku, Kyoto, 602-8566, Japan
| | - Kohichiroh Yasui
- Department of Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyou-ku, Kyoto, 602-8566, Japan
| | - Mai Kamaguchi
- Health Care Division, Kyoto Second Red Cross Hospital, Kyoto, Kyoto, Japan
| | - Kenichi Nishioji
- Health Care Division, Kyoto Second Red Cross Hospital, Kyoto, Kyoto, Japan
| | - Naomi Mochizuki
- Health Care Division, Kyoto Second Red Cross Hospital, Kyoto, Kyoto, Japan
| | - Masao Kobayashi
- Health Care Division, Kyoto Second Red Cross Hospital, Kyoto, Kyoto, Japan
| | - Kojiroh Mori
- Center for Digestive and Liver Diseases, Nara City Hospital, Nara, Japan
| | - Saiyu Tanaka
- Center for Digestive and Liver Diseases, Nara City Hospital, Nara, Japan
| | - Kentaro Matsuura
- Department of Virology and Liver Unit, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Yasuhito Tanaka
- Department of Virology and Liver Unit, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Yoshito Itoh
- Department of Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyou-ku, Kyoto, 602-8566, Japan.
| |
Collapse
|
28
|
Nakahara T, Hyogo H, Ono A, Nagaoki Y, Kawaoka T, Miki D, Tsuge M, Hiraga N, Hayes CN, Hiramatsu A, Imamura M, Kawakami Y, Aikata H, Ochi H, Abe-Chayama H, Furusho H, Shintani T, Kurihara H, Miyauchi M, Takata T, Arihiro K, Chayama K. Involvement of Porphyromonas gingivalis in the progression of non-alcoholic fatty liver disease. J Gastroenterol 2018; 53:269-280. [PMID: 28741270 DOI: 10.1007/s00535-017-1368-4] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Accepted: 07/03/2017] [Indexed: 02/04/2023]
Abstract
BACKGROUND AND AIMS The risk factors in the progression of nonalcoholic fatty liver disease (NAFLD) have not been fully clarified. Porphyromonas gingivalis (P.g) has been considered to be a confounding risk factor for systemic diseases. We aimed to evaluate the effect of P.g infection on risk of progression to NASH. METHODS (1) Serum IgG antibody titers against P.g fimbriae (fimA) in 200 biopsy-proven NAFLD patients were measured by ELISA and compared with histological findings. (2) C57BL/6J mice were fed a control diet (CD) or high-fat diet (HFD) with or without P.g-odontogenic infection and analyzed histologically. Mouse livers were analyzed using CE-TOFMS and LC-TOFMS. RESULTS (1) A significant correlation between fibrosis progression and antibody titers against P.g possessing fimA type 4 was identified (P = 0.0081). Multivariate analysis identified older age and type 4 P.g-positivity as risk factors for advanced fibrosis. (2) Fibrosis and steatosis were more severe in HFD P.g(+) mice compared with HFD P.g(-) mice. In metabolome analysis, fatty acid metabolism was significantly disrupted with HFD in P.g-infected mouse livers. Monounsaturated/saturated fatty acid ratios were significantly higher in the HFD P.g(+) group than in the HFD P.g(-) group (P < 0.05). Moreover, expression levels of SCD1 and ELOVL6 were significantly reduced. CONCLUSIONS These results suggest that P.g infection is an important risk factor for pathological progression in NAFLD. Increase in the monounsaturated/saturated fatty acid ratio may be an important change that facilitates progression of NAFLD.
Collapse
Affiliation(s)
- Takashi Nakahara
- Department of Gastroenterology and Metabolism, Division of Frontier Medical Science, Programs for Biomedical Research Graduate School of Biomedical Science, Applied Life Sciences, Institute of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan.,Liver Research Project Center, Hiroshima University, Hiroshima, Japan
| | - Hideyuki Hyogo
- Department of Gastroenterology and Hepatology, JA Hiroshima General Hospital, Hiroshima, Japan.,Liver Research Project Center, Hiroshima University, Hiroshima, Japan
| | - Atsushi Ono
- Department of Gastroenterology and Metabolism, Division of Frontier Medical Science, Programs for Biomedical Research Graduate School of Biomedical Science, Applied Life Sciences, Institute of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan.,Liver Research Project Center, Hiroshima University, Hiroshima, Japan
| | - Yuko Nagaoki
- Department of Gastroenterology and Metabolism, Division of Frontier Medical Science, Programs for Biomedical Research Graduate School of Biomedical Science, Applied Life Sciences, Institute of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan.,Liver Research Project Center, Hiroshima University, Hiroshima, Japan
| | - Tomokazu Kawaoka
- Department of Gastroenterology and Metabolism, Division of Frontier Medical Science, Programs for Biomedical Research Graduate School of Biomedical Science, Applied Life Sciences, Institute of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan.,Liver Research Project Center, Hiroshima University, Hiroshima, Japan
| | - Daiki Miki
- Laboratory for Digestive Diseases, RIKEN Center for Integrative Medical Sciences, Hiroshima, Japan.,Laboratory for Digestive Diseases, Center for Genomic Medicine, RIKEN, Hiroshima, Japan.,Liver Research Project Center, Hiroshima University, Hiroshima, Japan
| | - Masataka Tsuge
- Department of Gastroenterology and Metabolism, Division of Frontier Medical Science, Programs for Biomedical Research Graduate School of Biomedical Science, Applied Life Sciences, Institute of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan.,Liver Research Project Center, Hiroshima University, Hiroshima, Japan
| | - Nobuhiko Hiraga
- Department of Gastroenterology and Metabolism, Division of Frontier Medical Science, Programs for Biomedical Research Graduate School of Biomedical Science, Applied Life Sciences, Institute of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan.,Liver Research Project Center, Hiroshima University, Hiroshima, Japan
| | - Clair Nelson Hayes
- Department of Gastroenterology and Metabolism, Division of Frontier Medical Science, Programs for Biomedical Research Graduate School of Biomedical Science, Applied Life Sciences, Institute of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan.,Liver Research Project Center, Hiroshima University, Hiroshima, Japan
| | - Akira Hiramatsu
- Department of Gastroenterology and Metabolism, Division of Frontier Medical Science, Programs for Biomedical Research Graduate School of Biomedical Science, Applied Life Sciences, Institute of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan.,Liver Research Project Center, Hiroshima University, Hiroshima, Japan
| | - Michio Imamura
- Department of Gastroenterology and Metabolism, Division of Frontier Medical Science, Programs for Biomedical Research Graduate School of Biomedical Science, Applied Life Sciences, Institute of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan.,Liver Research Project Center, Hiroshima University, Hiroshima, Japan
| | - Yoshiiku Kawakami
- Department of Gastroenterology and Metabolism, Division of Frontier Medical Science, Programs for Biomedical Research Graduate School of Biomedical Science, Applied Life Sciences, Institute of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan.,Liver Research Project Center, Hiroshima University, Hiroshima, Japan
| | - Hiroshi Aikata
- Department of Gastroenterology and Metabolism, Division of Frontier Medical Science, Programs for Biomedical Research Graduate School of Biomedical Science, Applied Life Sciences, Institute of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan.,Liver Research Project Center, Hiroshima University, Hiroshima, Japan
| | - Hidenori Ochi
- Department of Gastroenterology and Metabolism, Division of Frontier Medical Science, Programs for Biomedical Research Graduate School of Biomedical Science, Applied Life Sciences, Institute of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan.,Laboratory for Digestive Diseases, Center for Genomic Medicine, RIKEN, Hiroshima, Japan.,Liver Research Project Center, Hiroshima University, Hiroshima, Japan
| | - Hiromi Abe-Chayama
- Liver Research Project Center, Hiroshima University, Hiroshima, Japan.,Center for Medical Specialist Graduate Education and Research, Institute of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Hisako Furusho
- Department of Oral and Maxillofacial Pathobiology, Basic Life Sciences, Institute of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Tomoaki Shintani
- Center of Oral Examination, Hiroshima University Hospital, Hiroshima, Japan
| | - Hidemi Kurihara
- Department of Periodontal Medicine, Division of Applied Life Sciences, Institute of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Mutsumi Miyauchi
- Department of Oral and Maxillofacial Pathobiology, Basic Life Sciences, Institute of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Takashi Takata
- Department of Oral and Maxillofacial Pathobiology, Basic Life Sciences, Institute of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Koji Arihiro
- Department of Anatomical Pathology, Hiroshima University Hospital, Hiroshima, Japan
| | - Kazuaki Chayama
- Department of Gastroenterology and Metabolism, Division of Frontier Medical Science, Programs for Biomedical Research Graduate School of Biomedical Science, Applied Life Sciences, Institute of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan. .,Laboratory for Digestive Diseases, RIKEN Center for Integrative Medical Sciences, Hiroshima, Japan. .,Laboratory for Digestive Diseases, Center for Genomic Medicine, RIKEN, Hiroshima, Japan. .,Liver Research Project Center, Hiroshima University, Hiroshima, Japan.
| |
Collapse
|
29
|
Yilmaz Y, Colak Y, Kurt R, Senates E, Eren F. Linking Nonalcoholic Fatty Liver Disease to Hepatocellular Carcinoma: From Bedside to Bench and Back. TUMORI JOURNAL 2018; 99:10-6. [DOI: 10.1177/030089161309900102] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Aims and background Nonalcoholic fatty liver disease (NAFLD) and hepatocellular carcinoma (HCC) are two major causes of liver disease worldwide. Epidemiological and clinical data have clearly demonstrated that NAFLD and its associated metabolic abnormalities are a risk factor for HCC. Traditionally, the mechanisms whereby NAFLD acts as a risk for HCC are believed to include replicative senescence of steatotic hepatocytes and compensatory hyperplasia of progenitor cells as a reaction to chronic hepatic injury. Recent years have witnessed significant advances in our understanding of the mechanisms underlying the link between NAFLD and HCC. Methods In the present review, we provide an update on the pathophysiological pathways linking NAFLD and its associated metabolic derangements to malignant hepatic transformation, with a special focus on insulin resistance, adipokines, inflammation, and angiogenesis. We will also discuss the potential therapeutic implications that such molecular links carry. Results Although treating NAFLD could reduce the risk of malignant hepatic transformation, no long-term studies focusing on this issue have been conducted thus far. Insulin resistance, inflammation as well as derangements in adipokines and angiogenic factors associated with NAFLD are closely intertwined with the risk of developing HCC. Conclusions Traditional therapeutic approaches in NAFLD including metformin and statins may theoretically reduce the risk of HCC by acting on common pathophysiological pathways shared by NAFLD and HCC.
Collapse
Affiliation(s)
- Yusuf Yilmaz
- Institute of Gastroenterology, School
of Medicine, Marmara University, Istanbul
- Department of Gastroenterology, School
of Medicine, Marmara University, Istanbul
| | - Yasar Colak
- Department of Gastroenterology,
Faculty of Medicine, Istanbul Medeniyet University, Istanbul
| | - Ramazan Kurt
- Department of Gastroenterology, School
of Medicine, Marmara University, Istanbul
| | - Ebubekir Senates
- Department of Gastroenterology, School
of Medicine, Dicle University, Diyarbakir
| | - Fatih Eren
- Institute of Gastroenterology, School
of Medicine, Marmara University, Istanbul
- Department of Medical Biology and
Genetics, School of Medicine, Marmara University, Istanbul, Turkey
| |
Collapse
|
30
|
Kim MJ, An HJ, Kim DH, Lee B, Lee HJ, Ullah S, Kim SJ, Jeong HO, Moon KM, Lee EK, Yang J, Akter J, Chun P, Moon HR, Chung HY. Novel SIRT1 activator MHY2233 improves glucose tolerance and reduces hepatic lipid accumulation in db/db mice. Bioorg Med Chem Lett 2018; 28:684-688. [PMID: 29402742 DOI: 10.1016/j.bmcl.2018.01.021] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Revised: 01/04/2018] [Accepted: 01/12/2018] [Indexed: 02/08/2023]
Abstract
The NAD+-dependent deacetylase SIRT1, which is associated with the improvement of metabolic syndromes, such as type 2 diabetes, is a well-known longevity-related gene. Several in vitro and in vivo studies have shown the known protective effects of SIRT1 activators, such as resveratrol and SRT1720, on diabetes- or obesity-induced fatty liver and insulin resistance. Here, we newly synthesized 18 benzoxazole hydrochloride derivatives based on the structure of resveratrol and SRT1720. We performed an in vitro SIRT1 activity assay to identify the strongest SIRT1 activator. The assay confirmed MHY2233 to be the strongest SIRT1 activator (1.5-fold more potent than resveratrol), and docking simulation showed that the binding affinity of MHY2233 was higher than that of resveratrol and SRT1720. To investigate its beneficial effects, db/db mice were orally administered MHY2233 for 1 month, and various metabolic parameters were assessed in the serum and liver tissues. MHY2233 markedly ameliorated insulin signaling without affecting body weight in db/db mice. In particular, the mRNA expression of lipogenic genes, such as acetyl CoA carboxylase, fatty acid synthase, and sterol regulatory element-binding protein, which increased in db/db mice, decreased following oral treatment with MHY2233. In conclusion, the novel SIRT1 activator MHY2233 reduced lipid accumulation and improved insulin resistance. This finding may contribute toward therapeutic approaches for fatty liver disease and glucose tolerance.
Collapse
Affiliation(s)
- Min Jo Kim
- Molecular Inflammation Research Center for Aging Intervention (MRCA), College of Pharmacy, Pusan National University, Busan 609-735, Republic of Korea
| | - Hye Jin An
- Molecular Inflammation Research Center for Aging Intervention (MRCA), College of Pharmacy, Pusan National University, Busan 609-735, Republic of Korea
| | - Dae Hyun Kim
- Molecular Inflammation Research Center for Aging Intervention (MRCA), College of Pharmacy, Pusan National University, Busan 609-735, Republic of Korea
| | - Bonggi Lee
- Molecular Inflammation Research Center for Aging Intervention (MRCA), College of Pharmacy, Pusan National University, Busan 609-735, Republic of Korea; Korean Medicine (KM)-Application Center, Korea Institute of Oriental Medicine (KIOM), Daegu 41062, Republic of Korea
| | - Hye Jin Lee
- Molecular Inflammation Research Center for Aging Intervention (MRCA), College of Pharmacy, Pusan National University, Busan 609-735, Republic of Korea
| | - Sultan Ullah
- Molecular Inflammation Research Center for Aging Intervention (MRCA), College of Pharmacy, Pusan National University, Busan 609-735, Republic of Korea
| | - Su Jeong Kim
- Molecular Inflammation Research Center for Aging Intervention (MRCA), College of Pharmacy, Pusan National University, Busan 609-735, Republic of Korea
| | - Hyoung Oh Jeong
- Molecular Inflammation Research Center for Aging Intervention (MRCA), College of Pharmacy, Pusan National University, Busan 609-735, Republic of Korea
| | - Kyoung Mi Moon
- Molecular Inflammation Research Center for Aging Intervention (MRCA), College of Pharmacy, Pusan National University, Busan 609-735, Republic of Korea; Korean Medicine (KM)-Application Center, Korea Institute of Oriental Medicine (KIOM), Daegu 41062, Republic of Korea
| | - Eun Kyeong Lee
- Molecular Inflammation Research Center for Aging Intervention (MRCA), College of Pharmacy, Pusan National University, Busan 609-735, Republic of Korea
| | - Jungho Yang
- Molecular Inflammation Research Center for Aging Intervention (MRCA), College of Pharmacy, Pusan National University, Busan 609-735, Republic of Korea
| | - Jinia Akter
- Molecular Inflammation Research Center for Aging Intervention (MRCA), College of Pharmacy, Pusan National University, Busan 609-735, Republic of Korea
| | - Pusoon Chun
- College of Pharmacy, Inje University, Gyeongsangnam-do 50834, Republic of Korea
| | - Hyung Ryong Moon
- Molecular Inflammation Research Center for Aging Intervention (MRCA), College of Pharmacy, Pusan National University, Busan 609-735, Republic of Korea.
| | - Hae Young Chung
- Molecular Inflammation Research Center for Aging Intervention (MRCA), College of Pharmacy, Pusan National University, Busan 609-735, Republic of Korea.
| |
Collapse
|
31
|
Liu X, Miao Y, Wu F, Du T, Zhang Q. Effect of CPAP therapy on liver disease in patients with OSA: a review. Sleep Breath 2018; 22:963-972. [PMID: 29327118 DOI: 10.1007/s11325-018-1622-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Revised: 12/15/2017] [Accepted: 01/05/2018] [Indexed: 12/15/2022]
Abstract
Obstructive sleep apnea (OSA) may play an important role in the progression of nonalcoholic fatty liver disease (NAFLD).The effect of continuous positive airway pressure (CPAP) treatment, the first-line therapy for OSA, on liver disease in OSA patients is still debated. We provide this review of previous studies to summarize the effects of CPAP treatment on liver disease in OSA patients in aspects of liver function, liver steatosis, fibrosis, and incidence of liver disease. CPAP treatment may be beneficial to liver disease in subjects with OSA independent of metabolic risk factors, but a sufficiently long therapeutic duration (perhaps greater than 3 months) may be needed to achieve these positive effects. Though the mechanism of impact of CPAP treatment on liver in OSA patients is unclear, the influence of CPAP treatment on the factors of the "Two-hit" hypothesis (insulin resistance, fatty acids dysregulation, oxidative stress, and inflammation) may be a reasonable explanation.
Collapse
Affiliation(s)
- Xin Liu
- Institute of Gerontology of Tianjin, Tianjin Medical University General Hospital, No.154, Anshan Road, Heping District, Tianjin, China
| | | | - Fan Wu
- Institute of Gerontology of Tianjin, Tianjin Medical University General Hospital, No.154, Anshan Road, Heping District, Tianjin, China
| | - Tingting Du
- Institute of Gerontology of Tianjin, Tianjin Medical University General Hospital, No.154, Anshan Road, Heping District, Tianjin, China
| | - Qiang Zhang
- Institute of Gerontology of Tianjin, Tianjin Medical University General Hospital, No.154, Anshan Road, Heping District, Tianjin, China.
| |
Collapse
|
32
|
Wang C, Batey R, Yamahara J, Li Y. Multiple molecular targets in the liver, adipose tissue and skeletal muscle in ginger-elicited amelioration of nonalcoholic fatty liver disease. J Funct Foods 2017. [DOI: 10.1016/j.jff.2017.06.040] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
|
33
|
Nagy E, Jermendy AL, Merkely B, Maurovich-Horvat P. Clinical importance of epicardial adipose tissue. Arch Med Sci 2017; 13:864-874. [PMID: 28721155 PMCID: PMC5507110 DOI: 10.5114/aoms.2016.63259] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2016] [Accepted: 08/23/2016] [Indexed: 12/12/2022] Open
Abstract
Different visceral fat compartments have several systemic effects and may play a role in the development of both insulin resistance and cardiovascular diseases. In the last couple of years special attention has been paid to the epicardial adipose tissue (EAT), which can be quantified by non-invasive cardiac imaging techniques. The epicardial fat is a unique fat compartment between the myocardium and the visceral pericardium sharing a common embryologic origin with the visceral fat depot. Epicardial adipose tissue has several specific roles, and its local effects on cardiac function are incorporated in the complex pathomechanism of coronary artery disease. Importantly, EAT may produce several adipocytokines and chemokines that may influence - through paracrine and vasocrine effects - the development and progression of coronary atherosclerosis. Epicardial adipose tissue volume has a relatively strong genetic dependence, similarly to other visceral fat depots. In this article, the anatomical and physiological as well as pathophysiological characteristics of the epicardial fat compartment are reviewed.
Collapse
Affiliation(s)
- Eszter Nagy
- MTA-SE Cardiovascular Imaging Research Group, Budapest, Hungary
| | - Adam L Jermendy
- MTA-SE Cardiovascular Imaging Research Group, Budapest, Hungary
| | - Bela Merkely
- MTA-SE Cardiovascular Imaging Research Group, Budapest, Hungary
| | | |
Collapse
|
34
|
Lombardi R, Onali S, Thorburn D, Davidson BR, Gurusamy KS, Tsochatzis E, Cochrane Hepato‐Biliary Group. Pharmacological interventions for non-alcohol related fatty liver disease (NAFLD): an attempted network meta-analysis. Cochrane Database Syst Rev 2017; 3:CD011640. [PMID: 28358980 PMCID: PMC6464620 DOI: 10.1002/14651858.cd011640.pub2] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND Non-alcohol related fatty liver disease (commonly called non-alcoholic fatty liver disease (NAFLD)) is liver steatosis in the absence of significant alcohol consumption, use of hepatotoxic medication, or other disorders affecting the liver such as hepatitis C virus infection, Wilson's disease, and starvation. NAFLD embraces the full spectrum of disease from pure steatosis (i.e. uncomplicated fatty liver) to non-alcoholic steatohepatitis (NASH), via NASH-cirrhosis to cirrhosis. The optimal pharmacological treatment for people with NAFLD remains uncertain. OBJECTIVES To assess the comparative benefits and harms of different pharmacological interventions in the treatment of NAFLD through a network meta-analysis and to generate rankings of the available pharmacological treatments according to their safety and efficacy. However, it was not possible to assess whether the potential effect modifiers were similar across different comparisons. Therefore, we did not perform the network meta-analysis, and instead, assessed the comparative benefits and harms of different interventions using standard Cochrane methodology. SEARCH METHODS We searched the Cochrane Central Register of Controlled Trials (CENTRAL), MEDLINE, Embase, Science Citation Index Expanded, the World Health Organization International Clinical Trials Registry Platform, and ClinicalTrials.com to August 2016. SELECTION CRITERIA We included only randomised clinical trials (irrespective of language, blinding, or publication status) in participants with NAFLD. We excluded trials which included participants who had previously undergone liver transplantation. We considered any of the various pharmacological interventions compared with each other or with placebo or no intervention. DATA COLLECTION AND ANALYSIS We calculated the odds ratio (OR) and rate ratio with 95% confidence intervals (CI) using both fixed-effect and random-effects models based on an available participant analysis with Review Manager. We assessed risk of bias according to the Cochrane risk of bias tool, controlled risk of random errors with Trial Sequential Analysis, and assessed the quality of the evidence using GRADE. MAIN RESULTS We identified 77 trials including 6287 participants that met the inclusion criteria of this review. Forty-one trials (3829 participants) provided information for one or more outcomes. Only one trial was at low risk of bias in all domains. All other trials were at high risk of bias in one or more domains. Overall, all the evidence was very low quality. Thirty-five trials included only participants with non-alcohol related steatohepatitis (NASH) (based on biopsy confirmation). Five trials included only participants with diabetes mellitus; 14 trials included only participants without diabetes mellitus. The follow-up in the trials ranged from one month to 24 months.We present here only the comparisons of active intervention versus no intervention in which two or more trials reported at least one of the following outcomes: mortality at maximal follow-up, serious adverse events, and health-related quality of life, the outcomes that determine whether a treatment should be used. Antioxidants versus no interventionThere was no mortality in either group (87 participants; 1 trial; very low quality evidence). None of the participants developed serious adverse events in the trial which reported the proportion of people with serious adverse events (87 participants; 1 trial; very low quality evidence). There was no evidence of difference in the number of serious adverse events between antioxidants and no intervention (rate ratio 0.89, 95% CI 0.36 to 2.19; 254 participants; 2 trials; very low quality evidence). None of the trials reported health-related quality of life. Bile acids versus no interventionThere was no evidence of difference in mortality at maximal follow-up (OR 5.11, 95% CI 0.24 to 107.34; 659 participants; 4 trials; very low quality evidence), proportion of people with serious adverse events (OR 1.56, 95% CI 0.84 to 2.88; 404 participants; 3 trials; very low quality evidence), or the number of serious adverse events (rate ratio 1.01, 95% CI 0.66 to 1.54; 404 participants; 3 trials; very low quality evidence) between bile acids and no intervention. None of the trials reported health-related quality of life. Thiazolidinediones versus no interventionThere was no mortality in either group (74 participants; 1 trial; very low quality evidence). None of the participants developed serious adverse events in the two trials which reported the proportion of people with serious adverse events (194 participants; 2 trials; very low quality evidence). There was no evidence of difference in the number of serious adverse events between thiazolidinediones and no intervention (rate ratio 0.25, 95% CI 0.06 to 1.05; 357 participants; 3 trials; very low quality evidence). None of the trials reported health-related quality of life. Source of fundingTwenty-six trials were partially- or fully-funded by pharmaceutical companies that would benefit, based on the results of the trial. Twelve trials did not receive any additional funding or were funded by parties with no vested interest in the results. The source of funding was not provided in 39 trials. AUTHORS' CONCLUSIONS Due to the very low quality evidence, we are very uncertain about the effectiveness of pharmacological treatments for people with NAFLD including those with steatohepatitis. Further well-designed randomised clinical trials with sufficiently large sample sizes are necessary.
Collapse
Affiliation(s)
- Rosa Lombardi
- Royal Free Hospital and the UCL Institute of Liver and Digestive HealthSheila Sherlock Liver CentreLondonUKNW3 2QG
| | - Simona Onali
- Royal Free Hospital and the UCL Institute of Liver and Digestive HealthSheila Sherlock Liver CentreLondonUKNW3 2QG
| | - Douglas Thorburn
- Royal Free Hospital and the UCL Institute of Liver and Digestive HealthSheila Sherlock Liver CentreLondonUKNW3 2QG
| | - Brian R Davidson
- Royal Free Campus, UCL Medical SchoolDepartment of SurgeryPond StreetLondonUKNW3 2QG
| | | | - Emmanuel Tsochatzis
- Royal Free Hospital and the UCL Institute of Liver and Digestive HealthSheila Sherlock Liver CentreLondonUKNW3 2QG
| | | |
Collapse
|
35
|
Seth RK, Das S, Dattaroy D, Chandrashekaran V, Alhasson F, Michelotti G, Nagarkatti M, Nagarkatti P, Diehl AM, Bell PD, Liedtke W, Chatterjee S. TRPV4 activation of endothelial nitric oxide synthase resists nonalcoholic fatty liver disease by blocking CYP2E1-mediated redox toxicity. Free Radic Biol Med 2017; 102:260-273. [PMID: 27913210 PMCID: PMC5989309 DOI: 10.1016/j.freeradbiomed.2016.11.047] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Revised: 11/24/2016] [Accepted: 11/28/2016] [Indexed: 01/11/2023]
Abstract
NAFLD is a clinically progressive disease with steatosis, inflammation, endothelial dysfunction and fibrosis being the stages where clinical intervention becomes necessary. Lack of early biomarkers and absence of a FDA approved drug obstructs efforts for effective treatment. NAFLD progression is strongly linked to a balance between liver injury, tissue regeneration and the functioning of endogenous defense mechanisms. The failure of the defense pathways to resist the tissue damage arising from redox stress, one of the "multiple hits" in disease progression, give rise to heightened inflammation and occasional fibrosis. We introduce an endogenous defense mechanism in the liver that is mediated by TRPV4, a transient receptor potential calcium-permeable ion channel that responds to the cytotoxic liver environment and negatively regulates CYP2E1, a cytochrome p450 enzyme. Using Trpv4-/- mice and cultured primary cells, we show that TRPV4 is activated both by damage associated molecular pattern HMGB1 and collagen in diseased Kupffer cells that in turn activate the endothelial NOS (NOS3) to release nitric oxide (NO). The diffusible NO acts in a paracrine fashion in neighboring hepatocytes to deactivate the redox toxicity induced by CYP2E1. We also find that CYP2E1-mediated TRPV4 repression in late stages causes an unrestricted progression of disease. Thus, TRPV4 functions as a sensor of cell stress in the diseased fatty liver and constitutes an endogenous defense molecule, a novel concept with potential for therapeutic approaches against NAFLD, perhaps also against hepatic drug toxicity in general.
Collapse
Affiliation(s)
- Ratanesh K Seth
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, SC 29208, USA
| | - Suvarthi Das
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, SC 29208, USA
| | - Diptadip Dattaroy
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, SC 29208, USA
| | - Varun Chandrashekaran
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, SC 29208, USA
| | - Firas Alhasson
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, SC 29208, USA
| | | | - Mitzi Nagarkatti
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, SC 29208, USA
| | - Prakash Nagarkatti
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, SC 29208, USA
| | - Anna Mae Diehl
- Division of Gastroenterology, Duke University, Durham, NC 27707, USA
| | - P Darwin Bell
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, AL 35294, USA
| | - Wolfgang Liedtke
- Department of Neurology, Duke University School of Medicine, Durham, NC 27707, USA.
| | - Saurabh Chatterjee
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, SC 29208, USA.
| |
Collapse
|
36
|
Kuraji R, Ito H, Fujita M, Ishiguro H, Hashimoto S, Numabe Y. Porphyromonas gingivalis induced periodontitis exacerbates progression of non-alcoholic steatohepatitis in rats. Clin Exp Dent Res 2016; 2:216-225. [PMID: 29744170 PMCID: PMC5839206 DOI: 10.1002/cre2.41] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Revised: 06/22/2016] [Accepted: 07/01/2016] [Indexed: 12/12/2022] Open
Abstract
Non-alcoholic steatohepatitis (NASH) is a chronic liver disease that can develop into hepatocirrhosis and hepatic carcinoma. In recent years, epidemiological and animal studies have reported that Porphyromonas gingivalis (P. gingivalis), a known periodontopathic bacteria, is closely related to NASH. However, previous studies could not demonstrate a direct relationship between periodontitis, P. gingivalis infection, and NASH. The purpose of the present study was to examine the impact of P. gingivalis-associated periodontitis on the onset and progression of NASH. Forty-two male Wistar rats were used in this study. Rats were fed a high-fat diet (HFD) for 12 weeks in order to induce fatty liver. At 4 weeks from the start of feeding, the animals were performed ligature placement around the maxillary first molar tooth in order to induce experimental periodontitis, and then a P. gingivalis slurry was applied around the ligature twice in a week for 8 weeks (HFD/Pg(+) group). Controls were given the slurry without P. gingivalis after ligature placement using the same protocol (HFD/Pg(-) group). Significant increases in alveolar bone resorption and inflammation in periodontal tissue around the molar tooth in the HFD/Pg(+) group were observed when compared with the HFD/Pg(-) group. Moreover, histological images showing NASH characterized by perivenular lipid deposition including big fatty drops, ballooning degeneration, and focal necrosis with inflammatory cells were confirmed in the liver of rats in the HFD/Pg(+) group. Significant increases in alanine aminotransaminase, aspartate aminotransferase, and C-reactive protein levels were observed in the HFD/Pg(+) group. Furthermore, endotoxin levels in serum in the HFD/Pg(+) group were significantly higher than those in the HFD/Pg(-) group. The present study demonstrated that experimental periodontitis induced by P. gingivalis led to the progression of NASH in rats with fatty liver. Increased levels of endotoxin derived from P. gingivalis infection appear to play a considerable role in the progression of NASH.
Collapse
Affiliation(s)
- Ryutaro Kuraji
- Department of PeriodontologySchool of Life Dentistry at Tokyo, Nippon Dental University1‐9‐20 FujimiChiyoda‐kuTokyo102‐8159Japan
| | - Hiroshi Ito
- Department of PeriodontologySchool of Life Dentistry at Tokyo, Nippon Dental University1‐9‐20 FujimiChiyoda‐kuTokyo102‐8159Japan
| | - Miyako Fujita
- Department of PeriodontologySchool of Life Dentistry at Tokyo, Nippon Dental University1‐9‐20 FujimiChiyoda‐kuTokyo102‐8159Japan
| | - Hitomi Ishiguro
- Department of PeriodontologySchool of Life Dentistry at Tokyo, Nippon Dental University1‐9‐20 FujimiChiyoda‐kuTokyo102‐8159Japan
| | | | - Yukihiro Numabe
- Department of PeriodontologySchool of Life Dentistry at Tokyo, Nippon Dental University1‐9‐20 FujimiChiyoda‐kuTokyo102‐8159Japan
| |
Collapse
|
37
|
Hepatic glucose utilization in hepatic steatosis and obesity. Biosci Rep 2016; 36:BSR20160381. [PMID: 27653524 PMCID: PMC5293565 DOI: 10.1042/bsr20160381] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Revised: 09/15/2016] [Accepted: 09/21/2016] [Indexed: 12/21/2022] Open
Abstract
Hepatic steatosis is associated with obesity and insulin resistance. Whether hepatic glucose utilization rate (glucose phosphorylation rate; MRglu) is increased in steatosis and/or obesity is uncertain. Our aim was to determine the separate relationships of steatosis and obesity with MRglu. Sixty patients referred for routine PET/CT had dynamic PET imaging over the abdomen for 30 min post-injection of F-18-fluorodeoxyglucose (FDG), followed by Patlak-Rutland graphical analysis of the liver using abdominal aorta for arterial input signal. The plot gradient was divided by the intercept to give hepatic FDG clearance normalized to hepatic FDG distribution volume (ml/min per 100 ml) and multiplied by blood glucose to give hepatic MRglu (μmol/min per 100 ml). Hepatic steatosis was defined as CT density of ≤40 HU measured from the 60 min whole body routine PET/CT and obesity as body mass index of ≥30 kg/m2 Hepatic MRglu was higher in patients with steatosis (3.3±1.3 μmol/min per 100 ml) than those without (1.7±1.2 μmol/min per 100 ml; P<0.001) but there was no significant difference between obese (2.5±1.6 μmol/min per 100 ml) and non-obese patients (2.1±1.3 μmol/min per 100 ml). MRglu was increased in obese patients only if they had steatosis. Non-obese patients with steatosis still had increased MRglu. There was no association between MRglu and chemotherapy history. We conclude that MRglu is increased in hepatic steatosis probably through insulin resistance, hyperinsulinaemia and up-regulation of hepatic hexokinase, irrespective of obesity.
Collapse
|
38
|
Ashworth WB, Davies NA, Bogle IDL. A Computational Model of Hepatic Energy Metabolism: Understanding Zonated Damage and Steatosis in NAFLD. PLoS Comput Biol 2016; 12:e1005105. [PMID: 27632189 PMCID: PMC5025084 DOI: 10.1371/journal.pcbi.1005105] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2015] [Accepted: 08/12/2016] [Indexed: 12/20/2022] Open
Abstract
In non-alcoholic fatty liver disease (NAFLD), lipid build-up and the resulting damage is known to occur more severely in pericentral cells. Due to the complexity of studying individual regions of the sinusoid, the causes of this zone specificity and its implications on treatment are largely ignored. In this study, a computational model of liver glucose and lipid metabolism is presented which treats the sinusoid as the repeating unit of the liver rather than the single hepatocyte. This allows for inclusion of zonated enzyme expression by splitting the sinusoid into periportal to pericentral compartments. By simulating insulin resistance (IR) and high intake diets leading to the development of steatosis in the model, we identify key differences between periportal and pericentral cells accounting for higher susceptibility to pericentral steatosis. Secondly, variation between individuals is seen in both susceptibility to steatosis and in its development across the sinusoid. Around 25% of obese individuals do not show excess liver fat, whilst 16% of lean individuals develop NAFLD. Furthermore, whilst pericentral cells tend to show higher lipid levels, variation is seen in the predominant location of steatosis from pericentral to pan-sinusoidal or azonal. Sensitivity analysis was used to identify the processes which have the largest effect on both total hepatic triglyceride levels and on the sinusoidal location of steatosis. As is seen in vivo, steatosis occurs when simulating IR in the model, predominantly due to increased uptake, along with an increase in de novo lipogenesis. Additionally, concentrations of glucose intermediates including glycerol-3-phosphate increased when simulating IR due to inhibited glycogen synthesis. Several differences between zones contributed to a higher susceptibility to steatosis in pericentral cells in the model simulations. Firstly, the periportal zonation of both glycogen synthase and the oxidative phosphorylation enzymes meant that the build-up of glucose intermediates was less severe in the periportal hepatocyte compartments. Secondly, the periportal zonation of the enzymes mediating β-oxidation and oxidative phosphorylation resulted in excess fats being metabolised more rapidly in the periportal hepatocyte compartments. Finally, the pericentral expression of de novo lipogenesis contributed to pericentral steatosis when additionally simulating the increase in sterol-regulatory element binding protein 1c (SREBP-1c) seen in NAFLD patients in vivo. The hepatic triglyceride concentration was predicted to be most sensitive to inter-individual variation in the activity of enzymes which, either directly or indirectly, determine the rate of free fatty acid (FFA) oxidation. The concentration was most strongly dependent on the rate constants for β-oxidation and oxidative phosphorylation. It also showed moderate sensitivity to the rate constants for processes which alter the allosteric inhibition of β-oxidation by acetyl-CoA. The predominant sinusoidal location of steatosis meanwhile was most sensitive variations in the zonation of proteins mediating FFA uptake or triglyceride release as very low density lipoproteins (VLDL). Neither the total hepatic concentration nor the location of steatosis showed strong sensitivity to variations in the lipogenic rate constants.
Collapse
Affiliation(s)
- William B. Ashworth
- Institute of Liver and Digestive Health, University College London, London, United Kingdom
- Department of Chemical Engineering, University College London, London, United Kingdom
- CoMPLEX, University College London, London, United Kingdom
| | - Nathan A. Davies
- Institute of Liver and Digestive Health, University College London, London, United Kingdom
| | - I. David L. Bogle
- Department of Chemical Engineering, University College London, London, United Kingdom
- * E-mail:
| |
Collapse
|
39
|
Corey KE, Klebanoff MJ, Tramontano AC, Chung RT, Hur C. Screening for Nonalcoholic Steatohepatitis in Individuals with Type 2 Diabetes: A Cost-Effectiveness Analysis. Dig Dis Sci 2016; 61:2108-17. [PMID: 26825843 PMCID: PMC4920690 DOI: 10.1007/s10620-016-4044-2] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Accepted: 01/16/2016] [Indexed: 12/22/2022]
Abstract
BACKGROUND AND AIM Individuals with type 2 diabetes are at heightened risk for nonalcoholic fatty liver disease, which gives rise to nonalcoholic steatohepatitis (NASH) and cirrhosis. Yet, current guidelines do not recommend screening for NASH among these high-risk patients. Using a simulation model, we assessed the effectiveness and cost-effectiveness of screening diabetic patients for NASH. METHODS A Markov model was constructed to compare two management strategies for 50-year-olds with diabetes. In the No Screening strategy, patients do not undergo screening, although NASH may be diagnosed incidentally over their lifetime. In the NASH Screening strategy, all patients receive a one-time screening ultrasound. Individuals with fatty infiltration on ultrasound then have a liver biopsy, and those found to have NASH receive medical therapy, which decreases progression to cirrhosis. Endpoints evaluated included quality-adjusted life years (QALYs) gained, costs, and incremental cost-effectiveness ratios (ICERs). RESULTS Screening for NASH decreased the number of individuals who developed cirrhosis by 12.9 % and resulted in an 11.9 % decrease in liver-related deaths. However, screening resulted in 0.02 fewer QALYs, due to the disutility associated with treatment, and was therefore dominated by the No Screening strategy. When the model excluded this quality-of-life decrement, screening became cost-effective, at an ICER of $42,134 per QALY. CONCLUSIONS Screening for NASH may improve liver-related outcomes, but is not cost-effective at present, due to side effects of therapy. As better tolerated treatments for NASH become available, even with modest efficacy, screening for NASH will become cost-effective.
Collapse
Affiliation(s)
- Kathleen E. Corey
- Gastrointestinal Unit, Massachusetts General Hospital, Boston, MA, USA,Harvard Medical School, Boston, MA, USA
| | - Matthew J. Klebanoff
- Gastrointestinal Unit, Massachusetts General Hospital, Boston, MA, USA,Institute for Technology Assessment, Massachusetts General Hospital, Boston, MA, USA
| | - Angela C. Tramontano
- Institute for Technology Assessment, Massachusetts General Hospital, Boston, MA, USA
| | - Raymond T. Chung
- Gastrointestinal Unit, Massachusetts General Hospital, Boston, MA, USA,Harvard Medical School, Boston, MA, USA
| | - Chin Hur
- Gastrointestinal Unit, Massachusetts General Hospital, Boston, MA, USA,Harvard Medical School, Boston, MA, USA,Institute for Technology Assessment, Massachusetts General Hospital, Boston, MA, USA
| |
Collapse
|
40
|
Ashworth W, Perez-Galvan C, Davies N, Bogle IDL. Liver function as an engineering system. AIChE J 2016. [DOI: 10.1002/aic.15292] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- William Ashworth
- Centre for Process Systems Engineering, Dept. of Chemical Engineering; University College London, London WC1E 7JE, U.K
- Institute for Liver and Digestive Health, Division of Medicine, University College London, Royal Free Campus, London NW3 2PF, U.K
- COMPLEX (Centre for Mathematics and Physics in the Life Sciences and Experimental Biology); University College London, London WC1E 6BT, U.K
| | - Carlos Perez-Galvan
- Centre for Process Systems Engineering, Dept. of Chemical Engineering; University College London, London WC1E 6BT, U.K
| | - Nathan Davies
- Institute for Liver and Digestive Health, Division of Medicine, University College London, Royal Free Campus, London NW3 2PF, U.K
| | - Ian David Lockhart Bogle
- Centre for Process Systems Engineering, Dept. of Chemical Engineering; University College London, London WC1E 7JE, U.K
- COMPLEX (Centre for Mathematics and Physics in the Life Sciences and Experimental Biology); University College London, London WC1E 6BT, U.K
| |
Collapse
|
41
|
Liu L, Zhao J, Li Y, Wan Y, Lin J, Shen A, Xu W, Li H, Zhang Y, Xu J, Peng J, Hong Z. Artemisia capillaris formula inhibits hepatic steatosis via an miR‑122‑induced decrease in fatty acid synthase expression in vivo and in vitro. Mol Med Rep 2016; 13:4751-4758. [PMID: 27081834 DOI: 10.3892/mmr.2016.5131] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Accepted: 03/05/2015] [Indexed: 11/06/2022] Open
Abstract
Non‑alcoholic fatty liver disease (NAFLD) is a widespread health concern, and there is currently insufficient understanding regarding its pathogenesis and treatment. The present study aimed to explore the effects of Artemisia capillaris formula (ACF) on high‑fat diet‑induced hepatic steatosis and fatty acid‑induced intracellular lipid accumulation, by micro (mi)RNA regulation. A total of 72 Sprague‑Dawley rats were divided into six groups (n=12/group). One group was designated as the control group and fed a normal diet, and the remaining five groups were allowed ad libitum access to a high‑fat diet for eight weeks, in order to establish an NAFLD rat model. The rats were subsequently administered polyene phosphatidylcholine (PP; 0.076 g/kg body weight/day), low dose of ACF (0.462 g/kg body weight/day), middle dose of ACF (0.924 g/kg body weight/day) or high dose of ACF (1.848 g/kg body weight/day) intragastrically for four weeks. HepG2 human hepatocellular carcinoma cells were treated with oleic acid and palm acid, followed by treatment with various concentrations of ACF. Serum alanine transaminase (ALT), aspartate aminotransferase (AST), triglycerides (TG), total cholesterol (TC), high‑density lipoprotein cholesterol (HDL‑C), low‑density lipoprotein cholesterol (LDL‑C), and steatotic HepG2 human liver carcinoma cell TC and TG levels were measured. ACF and PP treatments attenuated high‑fat diet‑induced hepatic steatosis and fatty acid‑induced intracellular lipid accumulation. A modified high‑fat diet significantly increased ALT, AST, TG, TC, LDL‑C levels and decreased HDL‑C levels. Treatment with ACF and PP abrogated the increase in liver enzymes and TG, TC and LDL‑C levels, but did not influence HDL‑C levels in a high‑fat diet induced rat model of steotosis. Steatotic HepG2 cells exhibited significantly increased levels of both TG and TC. Treatment with ACF significantly decreased TC and TG levels in vivo, and ACF and PP treatment decreased the expression levels of fatty acid synthase (FASN) and increased miR‑122 in vivo and in vitro. In conclusion, these results suggested that ACF may inhibit hepatic steatosis via miR‑122‑induced downregulation of FASN in vivo and in vitro.
Collapse
Affiliation(s)
- Liya Liu
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Jinyan Zhao
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Ying Li
- Department of Medicine, Xiamen Hospital of Traditional Chinese Medicine, Jinshan Street Community Health Service, Xiamen, Fujian 361000, P.R. China
| | - Yun Wan
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Jiumao Lin
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Aling Shen
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Wei Xu
- Department of Pharmacology, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Huang Li
- Department of Pharmacology, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Yuchen Zhang
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Jianfeng Xu
- Fujian Guizhentang Pharmaceutical Co., Ltd., Quanzhou, Fujian 362142, P.R. China
| | - Jun Peng
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Zhenfeng Hong
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| |
Collapse
|
42
|
Kim KE, Ko KH, Heo RW, Yi CO, Shin HJ, Kim JY, Park JH, Nam S, Kim H, Roh GS. Artemisia annua Leaf Extract Attenuates Hepatic Steatosis and Inflammation in High-Fat Diet-Fed Mice. J Med Food 2016; 19:290-9. [PMID: 26741655 PMCID: PMC4799707 DOI: 10.1089/jmf.2015.3527] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Artemisia annua L. (AA) is a well-known source of the antimalarial drug artemisinin. AA also has an antibacterial and antioxidant activity. However, the effect of AA extract on hepatic steatosis induced by obesity is unclear. We investigated whether AA extract prevents obesity-induced insulin resistance and hepatic steatosis in high-fat diet (HFD)-fed mice. Mice were randomly divided into groups that received a normal chow diet or HFD with or without AA for 12 weeks. We found that AA extract reduced insulin resistance and hepatic steatosis in HFD-fed mice. Western blot analysis showed that HFD-induced expression of nuclear sterol regulatory element-binding protein 1 and carbohydrate-responsive element-binding protein in the livers was decreased by AA extract. In particular, dietary administration of AA extract decreased hepatic high-mobility group box 1 and cyclooxygenase-2 expression in HFD-fed mice. AA extract also attenuated HFD-induced collagen deposition and fibrosis-related transforming growth factor-β1 and connective tissue growth factor. These data indicate that dietary AA extract has beneficial effects on hepatic steatosis and inflammation in HFD-fed mice.
Collapse
Affiliation(s)
- Kyung Eun Kim
- 1 Department of Anatomy and Convergence Medical Science, Institute of Health Sciences, Gyeongsang National University School of Medicine , Gyeongnam, Republic of Korea
| | - Keon-Hee Ko
- 2 Gyeongnam Oriental Medicinal Herb Institute , Gyeongnam, Republic of Korea.,3 Division of Food Science, Gyeongnam National University of Science and Technology , Gyeongnam, Republic of Korea
| | - Rok Won Heo
- 1 Department of Anatomy and Convergence Medical Science, Institute of Health Sciences, Gyeongsang National University School of Medicine , Gyeongnam, Republic of Korea
| | - Chin-ok Yi
- 1 Department of Anatomy and Convergence Medical Science, Institute of Health Sciences, Gyeongsang National University School of Medicine , Gyeongnam, Republic of Korea
| | - Hyun Joo Shin
- 1 Department of Anatomy and Convergence Medical Science, Institute of Health Sciences, Gyeongsang National University School of Medicine , Gyeongnam, Republic of Korea
| | - Jun Young Kim
- 4 Busan Regional Korea Food and Drug Administration , Ministry of Food and Drug Safety, Busan, Republic of Korea
| | - Jae-Ho Park
- 5 Shinseon F&V Co., Ltd. , Gyeongnam, Republic of Korea
| | - Sanghae Nam
- 3 Division of Food Science, Gyeongnam National University of Science and Technology , Gyeongnam, Republic of Korea
| | - Hwajin Kim
- 1 Department of Anatomy and Convergence Medical Science, Institute of Health Sciences, Gyeongsang National University School of Medicine , Gyeongnam, Republic of Korea
| | - Gu Seob Roh
- 1 Department of Anatomy and Convergence Medical Science, Institute of Health Sciences, Gyeongsang National University School of Medicine , Gyeongnam, Republic of Korea
| |
Collapse
|
43
|
Briata P, Bordo D, Puppo M, Gorlero F, Rossi M, Perrone-Bizzozero N, Gherzi R. Diverse roles of the nucleic acid-binding protein KHSRP in cell differentiation and disease. WILEY INTERDISCIPLINARY REVIEWS-RNA 2015; 7:227-40. [PMID: 26708421 DOI: 10.1002/wrna.1327] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Revised: 11/16/2015] [Accepted: 11/17/2015] [Indexed: 12/15/2022]
Abstract
The single-stranded nucleic acid-binding protein KHSRP (KH-type splicing regulatory protein) modulates RNA life and gene expression at various levels. KHSRP controls important cellular functions as different as proliferation, differentiation, metabolism, and response to infectious agents. We summarize and discuss experimental evidence providing a potential link between changes in KHSRP expression/function and human diseases including neuromuscular disorders, obesity, type II diabetes, and cancer.
Collapse
Affiliation(s)
- Paola Briata
- Gene Expression Regulation Laboratory, IRCCS AOU San Martino-IST, Genova, Italy
| | - Domenico Bordo
- Gene Expression Regulation Laboratory, IRCCS AOU San Martino-IST, Genova, Italy
| | - Margherita Puppo
- Gene Expression Regulation Laboratory, IRCCS AOU San Martino-IST, Genova, Italy
| | - Franco Gorlero
- S.C. Ginecologia e Ostetricia Galliera Hospital, Genova, Italy.,School of Medicine, DINOGMI, University of Genova, Genova, Italy
| | - Martina Rossi
- Gene Expression Regulation Laboratory, IRCCS AOU San Martino-IST, Genova, Italy
| | - Nora Perrone-Bizzozero
- Department of Neurosciences, School of Medicine, University of New Mexico, Albuquerque, NM, USA
| | - Roberto Gherzi
- Gene Expression Regulation Laboratory, IRCCS AOU San Martino-IST, Genova, Italy
| |
Collapse
|
44
|
Bitter A, Rümmele P, Klein K, Kandel BA, Rieger JK, Nüssler AK, Zanger UM, Trauner M, Schwab M, Burk O. Pregnane X receptor activation and silencing promote steatosis of human hepatic cells by distinct lipogenic mechanisms. Arch Toxicol 2015; 89:2089-2103. [PMID: 25182422 DOI: 10.1007/s00204-014-1348-x] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2014] [Accepted: 08/25/2014] [Indexed: 12/20/2022]
Abstract
In addition to its well-characterized role in the regulation of drug metabolism and transport by xenobiotics, pregnane X receptor (PXR) critically impacts on lipid homeostasis. In mice, both ligand-dependent activation and knockout of PXR were previously shown to promote hepatic steatosis. To elucidate the respective pathways in human liver, we generated clones of human hepatoma HepG2 cells exhibiting different PXR protein levels, and analyzed effects of PXR activation and knockdown on steatosis and expression of lipogenic genes. Ligand-dependent activation as well as knockdown of PXR resulted in increased steatosis in HepG2 cells. Activation of PXR induced the sterol regulatory element-binding protein (SREBP) 1-dependent lipogenic pathway via PXR-dependent induction of SREBP1a, which was confirmed in primary human hepatocytes. Inhibiting SREBP1 activity by blocking the cleavage-dependent maturation of SREBP1 protein impaired the induction of lipogenic SREBP1 target genes and triglyceride accumulation by PXR activation. On the other hand, PXR knockdown resulted in up-regulation of aldo-keto reductase (AKR) 1B10, which enhanced the acetyl-CoA carboxylase (ACC)-catalyzed reaction step of de novo lipogenesis. In a cohort of human liver samples histologically classified for non-alcoholic fatty liver disease, AKR1B10, SREBP1a and SREBP1 lipogenic target genes proved to be up-regulated in steatohepatitis, while PXR protein was reduced. In summary, our data suggest that activation and knockdown of PXR in human hepatic cells promote de novo lipogenesis and steatosis by induction of the SREBP1 pathway and AKR1B10-mediated increase of ACC activity, respectively, thus providing mechanistic explanations for a putative dual role of PXR in the pathogenesis of steatohepatitis.
Collapse
Affiliation(s)
- Andreas Bitter
- Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Auerbachstrasse 112, 70376, Stuttgart, Germany
| | - Petra Rümmele
- Institute of Pathology, University of Regensburg, Regensburg, Germany
| | - Kathrin Klein
- Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Auerbachstrasse 112, 70376, Stuttgart, Germany
| | - Benjamin A Kandel
- Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Auerbachstrasse 112, 70376, Stuttgart, Germany
| | - Jessica K Rieger
- Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Auerbachstrasse 112, 70376, Stuttgart, Germany
| | - Andreas K Nüssler
- Department of Traumatology, University of Tübingen, Tübingen, Germany
| | - Ulrich M Zanger
- Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Auerbachstrasse 112, 70376, Stuttgart, Germany
| | - Michael Trauner
- Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Matthias Schwab
- Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Auerbachstrasse 112, 70376, Stuttgart, Germany
- Department of Clinical Pharmacology, Institute of Experimental and Clinical Pharmacology and Toxicology, University Hospital, Tübingen, Germany
| | - Oliver Burk
- Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Auerbachstrasse 112, 70376, Stuttgart, Germany.
| |
Collapse
|
45
|
Abstract
In insulin-resistant states (obesity, pre-diabetes, and type 2 diabetes), hepatic production of glucose and lipid synthesis are heightened in concert, implying that insulin deficiency and insulin excess coexists in this setting. The fact that insulin may be inadequate or excessive at any one point in differing organs and tissues has many biologic ramifications. In this context the concept of metabolic compartmentalization in the liver is offered herein as one perspective of this paradox. In particular, we focus on the hypothesis that insulin resistance accentuates differences in periportal and perivenous hepatocytes, namely periportal glucose production and perivenous lipid synthesis. Subsequently, excessive production of glucose and accumulation of lipids could be expected in the livers of patients with obesity and insulin resistance. Overall, in this review, we provide our integrative perspective regarding how excessive production of glucose in periportal hepatocytes and accumulation of lipids in perivenous hepatocytes interact in insulin resistant states.
Collapse
Affiliation(s)
- Roberto B Bazotte
- a Department of Pharmacology and Therapeutics ; State University of Maringá ; Maringá , Paraná , PR Brazil
| | | | | |
Collapse
|
46
|
Antioxidant and Anti-Inflammatory Activities of Kenyan Leafy Green Vegetables, Wild Fruits, and Medicinal Plants with Potential Relevance for Kwashiorkor. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2015; 2015:807158. [PMID: 26236384 PMCID: PMC4510108 DOI: 10.1155/2015/807158] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Accepted: 06/14/2015] [Indexed: 11/25/2022]
Abstract
Background. Inflammation, together with related oxidative stress, is linked with the etiology of kwashiorkor, a form of severe acute malnutrition in children. A diet rich in anti-inflammatory and antioxidant phytochemicals may offer potential for the prevention and treatment of kwashiorkor. We selected and assayed five leafy green vegetables, two wild fruits, and six medicinal plants from Kenya for their antioxidant and anti-inflammatory properties. Consensus regarding medicinal plant use was established from ethnobotanical data. Methods. Antioxidant activity and phenolic content were determined using the oxygen radical absorbance capacity (ORAC) assay and Folin-Ciocalteu procedure, respectively. Anti-inflammatory activity was assessed in vitro targeting the inflammatory mediator tumour necrosis factor-alpha (TNF-α). Results. Mangifera indica (leaves used medicinally) showed the greatest antioxidant activity (5940 ± 632 µM TE/µg) and total phenolic content (337 ± 3 mg GAE/g) but Amaranthus dubius (leafy vegetable) showed the greatest inhibition of TNF-α (IC50 = 9 ± 1 μg/mL), followed by Ocimum americanum (medicinal plant) (IC50 = 16 ± 1 μg/mL). Informant consensus was significantly correlated with anti-inflammatory effects among active medicinal plants (r2 = 0.7639, P = 0.0228). Conclusions. Several plant species commonly consumed by Kenyan children possess activity profiles relevant to the prevention and treatment of kwashiorkor and warrant further investigation.
Collapse
|
47
|
Cui XB, Luan JN, Chen SY. RGC-32 Deficiency Protects against Hepatic Steatosis by Reducing Lipogenesis. J Biol Chem 2015; 290:20387-95. [PMID: 26134570 DOI: 10.1074/jbc.m114.630186] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Indexed: 12/20/2022] Open
Abstract
Hepatic steatosis is associated with insulin resistance and metabolic syndrome because of increased hepatic triglyceride content. We have reported previously that deficiency of response gene to complement 32 (RGC-32) prevents high-fat diet (HFD)-induced obesity and insulin resistance in mice. This study was conducted to determine the role of RGC-32 in the regulation of hepatic steatosis. We observed that hepatic RGC-32 was induced dramatically by both HFD challenge and ethanol administration. RGC-32 knockout (RGC32(-/-)) mice were resistant to HFD- and ethanol-induced hepatic steatosis. The hepatic triglyceride content of RGC32(-/-) mice was decreased significantly compared with WT controls even under normal chow conditions. Moreover, RGC-32 deficiency decreased the expression of lipogenesis-related genes, sterol regulatory element binding protein 1c (SREBP-1c), fatty acid synthase, and stearoyl-CoA desaturase 1 (SCD1). RGC-32 deficiency also decreased SCD1 activity, as indicated by decreased desaturase indices of the liver and serum. Mechanistically, insulin and ethanol induced RGC-32 expression through the NF-κB signaling pathway, which, in turn, increased SCD1 expression in a SREBP-1c-dependent manner. RGC-32 also promoted SREBP-1c expression through activating liver X receptor. These results demonstrate that RGC-32 contributes to the development of hepatic steatosis by facilitating de novo lipogenesis through activating liver X receptor, leading to the induction of SREBP-1c and its target genes. Therefore, RGC-32 may be a potential novel drug target for the treatment of hepatic steatosis and its related diseases.
Collapse
Affiliation(s)
- Xiao-Bing Cui
- From the Department of Physiology and Pharmacology, University of Georgia, Athens, Georgia 30602 and
| | - Jun-Na Luan
- From the Department of Physiology and Pharmacology, University of Georgia, Athens, Georgia 30602 and
| | - Shi-You Chen
- From the Department of Physiology and Pharmacology, University of Georgia, Athens, Georgia 30602 and the Institute of Clinical Medicine and Department of Cardiology, Renmin Hospital, Hubei Medical University, Shiyan, 442000 Hubei, China
| |
Collapse
|
48
|
Das S, Alhasson F, Dattaroy D, Pourhoseini S, Seth RK, Nagarkatti M, Nagarkatti PS, Michelotti GA, Diehl AM, Kalyanaraman B, Chatterjee S. NADPH Oxidase-Derived Peroxynitrite Drives Inflammation in Mice and Human Nonalcoholic Steatohepatitis via TLR4-Lipid Raft Recruitment. THE AMERICAN JOURNAL OF PATHOLOGY 2015; 185:1944-57. [PMID: 25989356 DOI: 10.1016/j.ajpath.2015.03.024] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Revised: 03/04/2015] [Accepted: 03/26/2015] [Indexed: 12/12/2022]
Abstract
The molecular events that link NADPH oxidase activation and the induction of Toll-like receptor (TLR)-4 recruitment into hepatic lipid rafts in nonalcoholic steatohepatitis (NASH) are unclear. We hypothesized that in liver, NADPH oxidase activation is key in TLR4 recruitment into lipid rafts, which in turn up-regulates NF-κB translocation to the nucleus and subsequent DNA binding, leading to NASH progression. Results from confocal microscopy showed that liver from murine and human NASH had NADPH oxidase activation, which led to the formation of highly reactive peroxynitrite, as shown by 3-nitrotyrosine formation in diseased liver. Expression and recruitment of TLR4 into the lipid rafts were significantly greater in rodent and human NASH. The described phenomenon was NADPH oxidase, p47phox, and peroxynitrite dependent, as liver from p47phox-deficient mice and from mice treated with a peroxynitrite decomposition catalyst [iron(III) tetrakis(p-sulfonatophenyl)porphyrin] or a peroxynitrite scavenger (phenylboronic acid) had markedly less Tlr4 recruitment into lipid rafts. Mechanistically, peroxynitrite-induced TLR4 recruitment was linked to increased IL-1β, sinusoidal injury, and Kupffer cell activation while blocking peroxynitrite-attenuated NASH symptoms. The results strongly suggest that NADPH oxidase-mediated peroxynitrite drove TLR4 recruitment into hepatic lipid rafts and inflammation, whereas the in vivo use of the peroxynitrite scavenger phenylboronic acid, a novel synthetic molecule having high reactivity with peroxynitrite, attenuates inflammatory pathogenesis in NASH.
Collapse
Affiliation(s)
- Suvarthi Das
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, University of South Carolina, Columbia, South Carolina
| | - Firas Alhasson
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, University of South Carolina, Columbia, South Carolina
| | - Diptadip Dattaroy
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, University of South Carolina, Columbia, South Carolina
| | - Sahar Pourhoseini
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, University of South Carolina, Columbia, South Carolina
| | - Ratanesh Kumar Seth
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, University of South Carolina, Columbia, South Carolina
| | - Mitzi Nagarkatti
- Department of Pathology, Microbiology and Immunology, University of South Carolina, Columbia, South Carolina
| | - Prakash S Nagarkatti
- Department of Pathology, Microbiology and Immunology, University of South Carolina, Columbia, South Carolina
| | - Gregory A Michelotti
- Division of Gastroenterology, Department of Medicine, Duke University, Durham, North Carolina
| | - Anna Mae Diehl
- Division of Gastroenterology, Department of Medicine, Duke University, Durham, North Carolina
| | - Balaraman Kalyanaraman
- Department of Biophysics, Free Radical Research Center, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Saurabh Chatterjee
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, University of South Carolina, Columbia, South Carolina.
| |
Collapse
|
49
|
Chou TC, Liang WM, Wang CB, Wu TN, Hang LW. Obstructive sleep apnea is associated with liver disease: a population-based cohort study. Sleep Med 2015; 16:955-60. [PMID: 26116463 DOI: 10.1016/j.sleep.2015.02.542] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Revised: 02/12/2015] [Accepted: 02/25/2015] [Indexed: 12/21/2022]
Abstract
BACKGROUND The association between obstructive sleep apnea (OSA) and the risk of liver disease is unclear. Moreover, population-based studies on the risk of liver disease among people with OSA have not yet been conducted. This study aimed to investigate the risk of subsequent development of liver disease among people with OSA. METHODS Using Taiwan National Health Insurance claims data, this study collected subjects from a cohort of 17,374 people with OSA who were diagnosed between 2000 and 2008. A control group of 69,496 people was selected from the same database and matched by age, gender, urbanization, income, and date of initial admission. All subjects were followed up until 2010. Liver disease incidence and risk were calculated. RESULTS The overall risk of liver disease among people with OSA was significantly higher than in the control group (aHR = 5.52, p <0.001). Non-alcoholic fatty liver disease, cirrhosis, and hepatitis C had significant aHRs of 5.29, 7.50, and 7.19 (all at p <0.001), respectively. In contrast, hepatitis B had the smallest aHR of 3.71. CONCLUSIONS The risk of liver disease was more than five times higher among people with OSA compared with the control group; this was particularly for cirrhosis and hepatitis C. Liver disease is thus a very important health issue among people with OSA.
Collapse
Affiliation(s)
- Tzu-Chieh Chou
- Department of Public Health, College of Public Health, China Medical University, Taichung, Taiwan; Department of Health Risk Management, College of Management, China Medical University, Taichung, Taiwan
| | - Wen-Miin Liang
- Department of Public Health, College of Public Health, China Medical University, Taichung, Taiwan; Graduate Institute of Biostatistics, Biostatistics Center, College of Management, China Medical University, Taichung, Taiwan
| | - Chang-Bi Wang
- Department of Public Health, College of Public Health, China Medical University, Taichung, Taiwan
| | - Trong-Neng Wu
- Department of Public Health, College of Public Health, China Medical University, Taichung, Taiwan; Department of Nursing, College of Medicine and Nursing, Hunguang University, Taichung, Taiwan.
| | - Liang-Wen Hang
- Department of Respiratory Therapy, College of Health Care, China Medical University, Taichung, Taiwan; Sleep Medicine Center, Department of Pulmonary and Critical Care Medicine, China Medical University Hospital, Taichung, Taiwan; Department of Healthcare Administration, Asia University, Taichung, Taiwan.
| |
Collapse
|
50
|
Kabel AM, Abd Elmaaboud MA, Albarraq AA. Ameliorative potential of omega 3 fatty acids and HMG-CoA reductase inhibitors on experimentally-induced non-alcoholic steatohepatitis. Prostaglandins Leukot Essent Fatty Acids 2015; 96:1-9. [PMID: 25541279 DOI: 10.1016/j.plefa.2014.12.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Revised: 11/18/2014] [Accepted: 12/04/2014] [Indexed: 12/13/2022]
Abstract
Non-alcoholic steatohepatitis (NASH) has a relation to obesity. It may lead to hepatocellular carcinoma. To date, the therapeutic options are limited due to complex pathogenesis. This study aimed to investigate the effect of atorvastatin and omega 3 fatty acids on experimentally-induced NASH. Sixty male albino rats were divided into 6 equal groups; control group, high fat emulsion/sucrose (HFE/S) diet, HFE/S+carboxymethyl cellulose, HFE/S +Atorvastatin, HFE/S+Fish oil and HFE/S+Atorvastatin+Fish oil. Serum alanine aminotransferase, total cholesterol (TC), triglycerides (TG), high density lipoproteins, insulin, glucose, C-reactive protein and quantitative insulin sensitivity check index were measured. Also, hepatic TC, TG, malondialdehyde, tumor necrosis factor alpha (TNF-α), interleukin-6 (IL-6) and transforming growth factor beta 1 (TGF-β1) were determined. Liver sections were examined histopathologically. Atorvastatin improved lipid profile, inflammation and oxidative stress but did not improve insulin resistance, hepatic TGF-β1 or body weight while fish oil improved lipid profile, decreased inflammation and oxidative stress, improved insulin resistance, hepatic TGF-β1 and body weight compared to HFE/S group. Atorvastatin/fish oil combination produced significant improvement in the lipid profile, inflammation, oxidative stress, insulin resistance, hepatic TGF-β1 and body weight compared to the use of each of these drugs alone. This might be attributed to the effect of fish oil on the lipid profile, inflammatory cytokines, insulin resistance and TGF-β1 which potentiates the effect of atorvastatin on NASH.
Collapse
Affiliation(s)
- Ahmed M Kabel
- Department of Clinical Pharmacy, Faculty of Pharmacy, Taif University, Taif, Saudi Arabia; Pharmacology department, Faculty of Medicine, Tanta University, Tanta, Egypt.
| | | | - Ahmed A Albarraq
- Department of Clinical Pharmacy, Faculty of Pharmacy, Taif University, Taif, Saudi Arabia
| |
Collapse
|