1
|
Alshuweishi Y, Alfayez D, Almufarrih AA, Abudawood A, Alyami H, Alshuweishi FA, Al-Sheikh YA, Alfhili MA. Elevated Alanine Transaminase-to-Platelet Index (APRI) Is Associated with Obesity and Distinct Forms of Dyslipidemia: A Retrospective Cross-Sectional Study. J Clin Med 2024; 13:5650. [PMID: 39337137 PMCID: PMC11432626 DOI: 10.3390/jcm13185650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 09/17/2024] [Accepted: 09/22/2024] [Indexed: 09/30/2024] Open
Abstract
Background: Obesity is a pathological condition and a major risk factor for dyslipidemia, type 2 diabetes, and non-alcoholic fatty liver disease. Recent research highlighted the association of non-invasive serum markers with these conditions but the clinical utility of ALT APRI in obesity and its relationship with dyslipidemia remain unexplored. Methods: We examined the association of ALT APRI in 165 non-diabetic adults stratified by BMI and serum lipid parameters. Results: Obese subjects had significantly higher APRI than lean subjects, with an area under the curve (AUC) of 0.65 (p = 0.019). Medians of APRI were significantly increased in subjects with high TG, TG/HDL, TC/HDL, and LDL/HDL and low HDL. Notably, all lipid parameters and ratios were significantly elevated in the highest APRI tertile, compared with patients in the lowest tertile. APRI was weakly yet significantly correlated with BMI (R2 = 0.032, p = 0.022), HDL (R2 = 0.071), TG/HDL (R2 = 0.031), TC/HDL (R2 = 0.063), LDL/HDL (R2 = 0.072), and TyG index (R2 = 0.081). While APRI only showed a discriminating capacity for HDL (AUC: 0.69, p = 0.003), TG/HDL (AUC: 0.63, p = 0.020), LDL/HDL (AUC: 0.68, p < 0.001), and TyG index (AUC: 0.65, p = 0.037), the highest diagnostic performance of APRI was observed with TC/HDL (AUC: 0.74, p < 0.001). Additionally, APRI was a risk factor for high TG (OR: 1.6, p = 0.028), low HDL (OR: 2.7, p = 0.0002), high TG/HDL (OR: 1.94, p = 0.0011), high TC/HDL (OR: 2.3, p < 0.0001), high LDL/HDL (OR: 2.2, p = 0.0001), and high TyG index (OR: 2.1, p = 0.008). Conclusions: Our findings argue for the role of APRI as a potential marker for obesity and dyslipidemia, which requires further confirmation in longitudinal studies.
Collapse
Affiliation(s)
- Yazeed Alshuweishi
- Chair of Medical and Molecular Genetics Research, Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, Riyadh 12372, Saudi Arabia
| | - Dalal Alfayez
- Department of Family and Community Medicine, Prince Sultan Military Medical City, Riyadh 11159, Saudi Arabia
| | - Abdulmalik A Almufarrih
- Department of Family and Community Medicine, Prince Sultan Military Medical City, Riyadh 11159, Saudi Arabia
| | - Arwa Abudawood
- Department of Family and Community Medicine, Prince Sultan Military Medical City, Riyadh 11159, Saudi Arabia
| | - Hanan Alyami
- Department of Medical and Surgical Nursing, College of Nursing, Princess Norah bint Abdurrahman University, Riyadh 11564, Saudi Arabia
| | - Faisal A Alshuweishi
- Department of Pathology and Laboratory Medicine, King Khalid University Hospital, King Saud University Medical City, Riyadh 12372, Saudi Arabia
| | - Yazeed A Al-Sheikh
- Chair of Medical and Molecular Genetics Research, Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, Riyadh 12372, Saudi Arabia
| | - Mohammad A Alfhili
- Chair of Medical and Molecular Genetics Research, Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, Riyadh 12372, Saudi Arabia
| |
Collapse
|
2
|
Guimarães JSF, Mesquita JA, Kimura TY, Oliveira ALM, Leite MF, Oliveira AG. Burden of liver disease in Brazil, 1996-2022: a retrospective descriptive study of the epidemiology and impact on public healthcare. LANCET REGIONAL HEALTH. AMERICAS 2024; 33:100731. [PMID: 38800645 PMCID: PMC11117060 DOI: 10.1016/j.lana.2024.100731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 03/15/2024] [Accepted: 03/20/2024] [Indexed: 05/29/2024]
Abstract
Background Liver disease is a major cause of mortality and morbidity worldwide and its epidemiology depends on the genetic background, exposure to risk factors, access to healthcare and other sociodemographic characteristics. Brazil is a large country with diverse multicultural and ethnic heritages and important socioeconomic inequalities. The burden of liver disease in Brazil, its regions and population is unknown. Methods We retrieved data from the Unified Health System regarding liver diseases and analyzed the mortality and morbidity from 1996 to 2022 by gender, race/ethnicity, age, region and overall. We calculated the age-specific risk of deaths by liver disease, age-standardization of the data, mean hospitalization and liver transplant-associated costs. Findings Malignant neoplasm of the liver and intrahepatic bile ducts, alcohol-associated liver disease, fibrosis, and cirrhosis of the liver, other diseases of the liver, hepatic failure, chronic viral hepatitis were identified as the major causes of death and morbidity in Brazil in the period analyzed. The epidemiology of these diseases was diverse, with variations according to geographic regions, gender and race/ethnicity. The major economic burden of liver disease is related to liver transplants, a common outcome of the progression of these diseases. Interpretation Liver disease in Brazil is a serious issue for the public health system due to the high number of deaths and increasing mortality rate. Our study contributes as a necessary prerequisite for the development of tailored public health policies aimed at mitigating the increasing burden of liver diseases in specific populations and regions. Funding CNPq, INCT, CAPES, FAPEMIG.
Collapse
Affiliation(s)
- João Sérgio Fonseca Guimarães
- Department of Physiology and Biophysics, Institute of Biological Science, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Jordana Almeida Mesquita
- Department of Physiology and Biophysics, Institute of Biological Science, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
- School of Medicine, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Thais Yuki Kimura
- Department of Physiology and Biophysics, Institute of Biological Science, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
- School of Medicine, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Ana Luíza Matos Oliveira
- Economic Development Unit, Economic Commission for Latin America and the Caribbean (ECLAC), Mexico City, Mexico
| | - M. Fatima Leite
- Department of Physiology and Biophysics, Institute of Biological Science, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - André Gustavo Oliveira
- Department of Physiology and Biophysics, Institute of Biological Science, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| |
Collapse
|
3
|
Li X, Chen W, Ren J, Gao X, Zhao Y, Song T, Fu K, Zheng Y, Yang J. Effects of curcumin on non-alcoholic fatty liver disease: A scientific metrogy study. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 123:155241. [PMID: 38128395 DOI: 10.1016/j.phymed.2023.155241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Revised: 10/26/2023] [Accepted: 11/23/2023] [Indexed: 12/23/2023]
Abstract
BACKGROUND Non-alcoholic fatty liver disease (NAFLD) is one of the most common chronic liver diseases encountered in clinical practice. Curcumin can alleviate insulin resistance, inhibit oxidative stress response, reduce inflammation, reduce liver fat deposition, and effectively improve NAFLD through various modalities, inhibiting the progression into cirrhosis and fibrosis. PURPOSE To explore the current status, hot spots, and developing trends of curcumin in NAFLD treatment through quantitative scientific analysis to serve as a reference for subsequent studies. STUDY DESIGN A comprehensive analysis of the mechanism of action of curcumin in the treatment of NAFLD and methods to increase curcumin bioavailability using bibliometric analysis and literature review. METHODS This study used VOSviewer software to analyze the literature related to curcumin treatment of NAFLD in the Web of Science (WOS) core set database. A comprehensive and in-depth review was conducted based on the results of scientific econometric research and literature review. RESULTS The review observed that curcumin can activate various signaling pathways such as AMPK and NF-κB to inhibit oxidative stress and apoptosis, thereby reflecting its pharmacological effects: lowering lipid, anti-inflammatory, reducing insulin resistance, and anti-fibrosis. These mechanisms improve or even reverse the complex pathological features of lipid metabolism disorders associated with NAFLD. Curcumin also can potentially serve as a primary regulatory target for treating hepatic steatosis using gut microbiota. However, these pharmacological effects of curcumin were limited owing to its low bioavailability. CONCLUSION This review discusses NAFLD treatment with curcumin, analyzes the reasons for its low bioavailability, and introduces models for studying and methods for improving curcumin bioavailability. As research on NAFLD grows, future research should capture the trend of basic research, pay attention to clinical research, and continuously explore the therapeutic potential of curcumin.
Collapse
Affiliation(s)
- Xiankuan Li
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; State Key Lab of New Ceramics and Fine Processing, School of Materials Science and Engineering, Tsinghua University, Beijing 100084, China; Tianjin Key Laboratory of Therapeutic Substance of Traditional Chinese Medicine, Tianjin 301617, China
| | - Weisan Chen
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Tianjin Key Laboratory of Therapeutic Substance of Traditional Chinese Medicine, Tianjin 301617, China
| | - Jiali Ren
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Tianjin Key Laboratory of Therapeutic Substance of Traditional Chinese Medicine, Tianjin 301617, China
| | - Xinchen Gao
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Tianjin Key Laboratory of Therapeutic Substance of Traditional Chinese Medicine, Tianjin 301617, China
| | - Ying Zhao
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Tianjin Key Laboratory of Therapeutic Substance of Traditional Chinese Medicine, Tianjin 301617, China
| | - Tianbao Song
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Tianjin Key Laboratory of Therapeutic Substance of Traditional Chinese Medicine, Tianjin 301617, China
| | - Kun Fu
- Second Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300120, China
| | - Yanchao Zheng
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Jinlong Yang
- State Key Lab of New Ceramics and Fine Processing, School of Materials Science and Engineering, Tsinghua University, Beijing 100084, China.
| |
Collapse
|
4
|
Aboujassoum HM, Mohamed-Ali V, Abraham D, Clapp LH, Al-Naemi HA. Relative Recovery of Non-Alcoholic Fatty Liver Disease (NAFLD) in Diet-Induced Obese Rats. Nutrients 2023; 16:115. [PMID: 38201945 PMCID: PMC10780646 DOI: 10.3390/nu16010115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 11/06/2023] [Accepted: 11/09/2023] [Indexed: 01/12/2024] Open
Abstract
Consumption of a high-carbohydrate diet has a critical role in the induction of weight gain and obesity-related pathologies. This study tested the hypothesis that a carbohydrate-rich diet induces weight gain, ectopic fat deposition, associated metabolic risks and development of non-alcoholic fatty liver disease (NAFLD), which are partially reversible following carbohydrate reduction. Sprague Dawley (SD) rats were fed a carbohydrate-enriched cafeteria diet (CAF) or normal chow (NC) ad libitum for 16-18 weeks. In the reversible group (REV), the CAF was replaced with NC for a further 3 weeks (18-21 weeks). Animals fed the CAF diet showed significantly increased body weight compared to those fed NC, accompanied by abnormal changes in their systemic insulin and triglycerides, elevation of hepatic triglyceride and hepatic steatosis. In the REV group, when the CAF diet was stopped, a modest, non-significant weight loss was associated with improvement in systemic insulin and appearance of the liver, with lower gross fatty deposits and hepatic triglyceride. In conclusion, a carbohydrate-enriched diet led to many features of metabolic syndrome, including hyperinsulinemia, while a dietary reduction in this macronutrient, even for a short period, was able to restore normoinsulinemia, and reversed some of the obesity-related hepatic abnormalities, without significant weight loss.
Collapse
Affiliation(s)
| | - Vidya Mohamed-Ali
- Anti-Doping Laboratory Qatar, Sports City Road, Doha P.O. Box 2713, Qatar;
- Centre of Metabolism and Inflammation, Division of Medicine, Royal Free Campus, University College London, Rowland Hill Street, London NW3 2PF, UK
| | - David Abraham
- Centre of Rheumatology and Connective Tissue Disorders, Division of Medicine, Royal Free Campus, University College London, Rowland Hill Street, London NW3 2PF, UK;
| | - Lucie H. Clapp
- Institute of Cardiovascular Science, University College London, Rayne Building, 5 University Street, London WC1E 6JF, UK;
| | - Hamda A. Al-Naemi
- Laboratory Animal Research Center, Qatar University, Doha P.O. Box 2713, Qatar;
- Department of Biological and Environmental Sciences, Qatar University, Doha P.O. Box 2713, Qatar
| |
Collapse
|
5
|
Bahijri S, Eldakhakhny B, Enani S, Ajabnoor G, Al-Mowallad AS, Alsheikh L, Alhozali A, Alamoudi AA, Borai A, Tuomilehto J. Fibroblast Growth Factor 21: A More Effective Biomarker Than Free Fatty Acids and Other Insulin Sensitivity Measures for Predicting Non-alcoholic Fatty Liver Disease in Saudi Arabian Type 2 Diabetes Patients. Cureus 2023; 15:e50524. [PMID: 38222178 PMCID: PMC10787595 DOI: 10.7759/cureus.50524] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/14/2023] [Indexed: 01/16/2024] Open
Abstract
Background Non-alcoholic fatty liver disease (NAFLD) is more prevalent among individuals with type 2 diabetes (T2DM), elevating their risk of cardiovascular diseases (CVDs) and premature mortality. There is a need to modify treatment strategies to prevent or delay these adverse outcomes. Currently, there are no sensitive or specific biomarkers for predicting NAFLD in Saudi T2DM patients. Therefore, we aimed to explore the possibility of using fibroblast growth factor 21 (FGF-21), free fatty acids (FFAs), homeostatic model assessment for insulin resistance (HOMA-IR), and quantitative insulin sensitivity check index (QUICKI) as possible markers. Methodology In this study, a total of 67 T2DM patients were recruited. NAFLD was detected by ultrasonography in 28 patients. Plasma glucose, FFAs, FGF-21, and serum insulin were measured in fasting blood samples. HOMA-IR and QUICKI were calculated. The means of the two groups with and without NAFLD were statistically compared. The receiver operating characteristics (ROC) curve and the area under the curve (AUC) were used to assess the ability to identify NAFLD. Results The mean levels of FGF-21 and HOMA-IR were significantly higher and that of QUICKI was significantly lower in patients with NAFLD than in those without (p < 0.001, p = 0.023, and p = 0.018, respectively). FGF-21 had the highest AUC to identify NAFLD (AUC = 0.981, 95% confidence interval = 0.954-1, P < 0.001). The AUCs for HOMA-IR, QUICKI, and FFA were <0.7. The highest sensitivity, specificity, positive likelihood ratio, and the lowest negative likelihood ratio were found when FGF-21 was used to predict NAFLD. Conclusions FGF-21 may be used as a biomarker to predict NAFLD in people with T2DM due to its high sensitivity and specificity compared to the other markers.
Collapse
Affiliation(s)
- Suhad Bahijri
- Department of Clinical Biochemistry, King Abdulaziz University Faculty of Medicine, Jeddah, SAU
- Saudi Diabetes Research Group, Deanship of Scientific Research, King Abdulaziz University, Jeddah, SAU
- Food, Nutrition and Lifestyle Unit, King Fahd Medical Research Centre, King Abdulaziz University, Jeddah, SAU
| | - Basmah Eldakhakhny
- Department of Clinical Biochemistry, King Abdulaziz University Faculty of Medicine, Jeddah, SAU
- Saudi Diabetes Research Group, Deanship of Scientific Research, King Abdulaziz University, Jeddah, SAU
- Food, Nutrition and Lifestyle Unit, King Fahd Medical Research Centre, King Abdulaziz University, Jeddah, SAU
| | - Sumia Enani
- Department of Food and Nutrition, Faculty of Human Sciences and Design, King Abdulaziz University, Jeddah, SAU
- Saudi Diabetes Research Group, Deanship of Scientific Research, King Abdulaziz University, Jeddah, SAU
- Food, Nutrition and Lifestyle Unit, King Fahd Medical Research Centre, King Abdulaziz University, Jeddah, SAU
| | - Ghada Ajabnoor
- Department of Clinical Biochemistry, King Abdulaziz University Faculty of Medicine, Jeddah, SAU
- Saudi Diabetes Research Group, Deanship of Scientific Research, King Abdulaziz University, Jeddah, SAU
- Food, Nutrition and Lifestyle Unit, King Fahd Medical Research Centre, King Abdulaziz University, Jeddah, SAU
| | - Alaa S Al-Mowallad
- Department of Clinical Biochemistry, King Abdulaziz University Faculty of Medicine, Jeddah, SAU
| | - Lubna Alsheikh
- Department of Biochemistry, King Abdulaziz University, Jeddah, SAU
| | - Amani Alhozali
- Department of Internal Medicine, King Abdulaziz University Hospital, Jeddah, SAU
| | - Aliaa A Alamoudi
- Department of Clinical Biochemistry, King Abdulaziz University Faculty of Medicine, Jeddah, SAU
- Saudi Diabetes Research Group, Deanship of Scientific Research, King Abdulaziz University, Jeddah, SAU
| | - Anwar Borai
- King Abdullah International Medical Research Center (KAIMRC), King Saud Bin Abdulaziz University for Health Sciences, Jeddah, SAU
- Saudi Diabetes Research Group, Deanship of Scientific Research, King Abdulaziz University, Jeddah, SAU
| | - Jaakko Tuomilehto
- Department of Public Health, University of Helsinki, Helsinki, FIN
- Public Health Promotion Unit, Finnish Institute for Health and Welfare, Helsinki, FIN
| |
Collapse
|
6
|
Eskridge W, Cryer DR, Schattenberg JM, Gastaldelli A, Malhi H, Allen AM, Noureddin M, Sanyal AJ. Metabolic Dysfunction-Associated Steatotic Liver Disease and Metabolic Dysfunction-Associated Steatohepatitis: The Patient and Physician Perspective. J Clin Med 2023; 12:6216. [PMID: 37834859 PMCID: PMC10573476 DOI: 10.3390/jcm12196216] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 09/19/2023] [Accepted: 09/19/2023] [Indexed: 10/15/2023] Open
Abstract
Diagnosing and managing metabolic dysfunction-associated steatotic liver disease (MASLD) remains a major challenge in primary care due to lack of agreement on diagnostic tools, difficulty in identifying symptoms and determining their cause, absence of approved pharmacological treatments, and limited awareness of the disease. However, prompt diagnosis and management are critical to preventing MASLD from progressing to more severe forms of liver disease. This highlights the need to raise awareness and improve understanding of MASLD among both patients and physicians. The patient perspective is invaluable to advancing our knowledge of this disease and how to manage it, as their perspectives have led to the growing recognition that patients experience subtle symptoms and that patient-reported outcomes should be incorporated into drug development. This review and expert opinion examine MASLD and metabolic dysfunction-associated steatohepatitis from the patient and physician perspective from pre-diagnosis to diagnosis and early care, through to progression to advanced liver damage. Specifically, the paper dives into the issues patients and physicians experience, and, in turn, what is required to improve diagnosis and management, including tips and tools to empower patients and physicians dealing with MASLD.
Collapse
Affiliation(s)
| | | | - Jörn M. Schattenberg
- Metabolic Liver Research Program, Department of Medicine, University Medical Center of the Johannes Gutenberg University, 155131 Mainz, Germany
| | - Amalia Gastaldelli
- Cardiometabolic Risk Laboratory, Institute of Clinical Physiology, Italian National Research Council CNR, 00133 Pisa, Italy
| | - Harmeet Malhi
- Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN 55901, USA
| | - Alina M. Allen
- Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN 55901, USA
| | - Mazen Noureddin
- Fatty Liver Program, Karsh Division of Gastroenterology and Hepatology, Cedar Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Arun J. Sanyal
- Stravitz-Sanyal Institute of Liver Disease and Metabolic Health, VCU School of Medicine and Health System and Division of Gastroenterology, Department of Internal Medicine, VCU School of Medicine, Richmond, VA 23298, USA
| |
Collapse
|
7
|
Zeng J, Acin-Perez R, Assali EA, Martin A, Brownstein AJ, Petcherski A, Fernández-Del-Rio L, Xiao R, Lo CH, Shum M, Liesa M, Han X, Shirihai OS, Grinstaff MW. Restoration of lysosomal acidification rescues autophagy and metabolic dysfunction in non-alcoholic fatty liver disease. Nat Commun 2023; 14:2573. [PMID: 37142604 PMCID: PMC10160018 DOI: 10.1038/s41467-023-38165-6] [Citation(s) in RCA: 48] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 04/18/2023] [Indexed: 05/06/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is the most common liver disease in the world. High levels of free fatty acids in the liver impair hepatic lysosomal acidification and reduce autophagic flux. We investigate whether restoration of lysosomal function in NAFLD recovers autophagic flux, mitochondrial function, and insulin sensitivity. Here, we report the synthesis of novel biodegradable acid-activated acidifying nanoparticles (acNPs) as a lysosome targeting treatment to restore lysosomal acidity and autophagy. The acNPs, composed of fluorinated polyesters, remain inactive at plasma pH, and only become activated in lysosomes after endocytosis. Specifically, they degrade at pH of ~6 characteristic of dysfunctional lysosomes, to further acidify and enhance the function of lysosomes. In established in vivo high fat diet mouse models of NAFLD, re-acidification of lysosomes via acNP treatment restores autophagy and mitochondria function to lean, healthy levels. This restoration, concurrent with reversal of fasting hyperglycemia and hepatic steatosis, indicates the potential use of acNPs as a first-in-kind therapeutic for NAFLD.
Collapse
Affiliation(s)
- Jialiu Zeng
- Department of Biomedical Engineering, Boston University, Boston, MA, 02215, USA.
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 308232, Singapore, Singapore.
| | - Rebeca Acin-Perez
- Division of Endocrinology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90045, USA
| | - Essam A Assali
- Division of Endocrinology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90045, USA
| | - Andrew Martin
- Department of Biomedical Engineering, Boston University, Boston, MA, 02215, USA
| | - Alexandra J Brownstein
- Division of Endocrinology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90045, USA
| | - Anton Petcherski
- Division of Endocrinology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90045, USA
| | - Lucía Fernández-Del-Rio
- Division of Endocrinology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90045, USA
| | - Ruiqing Xiao
- Department of Chemistry, Boston University, Boston, MA, 02215, USA
- Shenzhen Middle School, Shenzhen, Guangdong, 518001, China
| | - Chih Hung Lo
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 308232, Singapore, Singapore
| | - Michaël Shum
- Division of Endocrinology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90045, USA
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Marc Liesa
- Division of Endocrinology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90045, USA
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Molecular Biology Institute at University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Institut de Biologia Molecular de Barcelona, IBMB, CSIC, Barcelona, Catalonia, 08028, Spain
| | - Xue Han
- Department of Biomedical Engineering, Boston University, Boston, MA, 02215, USA
| | - Orian S Shirihai
- Division of Endocrinology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90045, USA.
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
- Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, 02118, USA.
| | - Mark W Grinstaff
- Department of Biomedical Engineering, Boston University, Boston, MA, 02215, USA.
- Department of Chemistry, Boston University, Boston, MA, 02215, USA.
- Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, 02118, USA.
| |
Collapse
|
8
|
Stepanova M, Henry L, Younossi ZM. Economic Burden and Patient-Reported Outcomes of Nonalcoholic Fatty Liver Disease. Clin Liver Dis 2023; 27:483-513. [PMID: 37024220 DOI: 10.1016/j.cld.2023.01.007] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/08/2023]
Abstract
In addition to adverse clinical outcomes such as liver-related morbidity and mortality, nonalcoholic fatty liver disease (NAFLD) is associated with a substantial public health and economic burden and could also potentially impair health-related quality of life and other patient-reported outcomes. The disease also affects multiple aspects of patients' quality of life which are the most pronounced in physical health-related and fatigue domains as well as work productivity, and get more severe in patients with advanced liver disease or with non-hepatic comorbidities. The economic burden of NAFLD is substantial and is increasing, with the highest costs in those with advanced disease.
Collapse
Affiliation(s)
- Maria Stepanova
- Department of Medicine, Center for Liver Diseases, Inova Fairfax Medical Campus, Falls Church, VA, USA; Betty and Guy Beatty Center for Integrated Research, Inova Health System, Falls Church, VA, USA; Center for Outcomes Research in Liver Diseases, Washington, DC, USA
| | - Linda Henry
- Department of Medicine, Center for Liver Diseases, Inova Fairfax Medical Campus, Falls Church, VA, USA; Betty and Guy Beatty Center for Integrated Research, Inova Health System, Falls Church, VA, USA; Center for Outcomes Research in Liver Diseases, Washington, DC, USA
| | - Zobair M Younossi
- Department of Medicine, Center for Liver Diseases, Inova Fairfax Medical Campus, Falls Church, VA, USA; Betty and Guy Beatty Center for Integrated Research, Inova Health System, Falls Church, VA, USA.
| |
Collapse
|
9
|
Xie R, Chen S, Li F, Yang L, Yu B. Pirfenidone attenuates nonalcoholic fatty liver disease through activation of the nuclear factor erythroid 2-related factor 2 (Nrf2) signaling pathway. J Biochem Mol Toxicol 2023; 37:e23251. [PMID: 36321572 DOI: 10.1002/jbt.23251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 09/01/2022] [Accepted: 10/12/2022] [Indexed: 02/10/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) originates from the hepatopathy of fatty liver. Pirfenidone is a novel broad-spectrum anti-fibrosis agent used for treating various kinds of tissue fibrosis. The present study will evaluate the effects of Pirfenidone on liver injury in high-fat diet (HFD)-fed mice to evaluate the value of Pirfenidone in treating NAFLD. The pathology of NAFLD was simulated by feeding mice with an HFD in the present study, followed by treating the HFD mice with 150 and 300 mg/kg/day Pirfenidone once a day. The pathological state of HFD mice was identified by the elevated liver weight, promoted serum triglyceride (TG), total cholesterol (TC), and low-density lipoprotein cholesterol (LDL-C) levels, declined serum high-density lipoprotein cholesterol (HDL-C) levels, increased alanine aminotransferase and aspartate aminotransferase activity, and histopathological changes to the liver tissues, all of which were dramatically ameliorated by 150 and 300 mg/kg Pirfenidone administration. Furthermore, the excessive production of interleukin-1β (IL-1β), tumor necrosis factor-α (TNF-α), and IL-6, as well as upregulated phosphorylated nuclear factor kappa-B (p- NF-κB p65), were observed in HFD-fed mice, but significantly reversed by Pirfenidone. Finally, activated oxidative stress, identified by promoted malondialdehyde (MDA) levels and declined catalase (CAT) activity, was observed in HFD-fed mice, accompanied by the downregulation of nuclear factor erythroid 2-related factor 2 (Nrf2) and sterol-regulatory element-binding proteins-1c (SREBP-1c). After the treatment with Pirfenidone, oxidative stress was greatly mitigated. Our results imply that Pirfenidone ameliorated the progression of NAFLD by mediating inflammation and oxidative stress.
Collapse
Affiliation(s)
- Rong Xie
- Department of the Traditional Chinese Medicine, The First Affiliated Hospital of Hainan Medical College, Hainan Province, Haikou City, China
| | - Shaodong Chen
- Department of Traditional Chinese Medicine, School of Medicine, Xiamen University, Fujian Province, Xiamen City, China
| | - Fang Li
- Department of the Traditional Chinese Medicine, The First Affiliated Hospital of Hainan Medical College, Hainan Province, Haikou City, China
| | - Liu Yang
- Department of Traditional Chinese Medicine, Hainan Medical University, Hainan Province, Haikou City, China
| | - Bangliang Yu
- Department of Traditional Chinese Medicine, Hainan Medical University, Hainan Province, Haikou City, China
| |
Collapse
|
10
|
Exploring the Validity of Available Markers and Indices in the Diagnosis of Nonalcoholic Fatty Liver Disease (NAFLD) in People with Type 2 Diabetes in Saudi Arabia. Diseases 2023; 11:diseases11010010. [PMID: 36648875 PMCID: PMC9887592 DOI: 10.3390/diseases11010010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Revised: 01/12/2023] [Accepted: 01/12/2023] [Indexed: 01/18/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is common among Saudi patients with type 2 diabetes (T2DM). However, recommended clinical procedures to detect it are unavailable in many locations. Therefore, better and more available diagnostic biomarkers for NAFLD are needed. Various serum parameters were suggested, and algorithms that employ routine measurements in clinical practice have been developed for the prediction of fat stores in the liver in different populations. However, no such studies have been conducted on Saudis. We aimed to compare selected biochemical markers and calculated indices in T2DM patients diagnosed with NAFLD and patients without NAFLD to find the best markers associated with NAFLD. A cross-sectional study was employed to recruit 67 people with T2DM from endocrine outpatient clinics at King Abdul-Aziz University Hospital. NAFLD was detected by ultrasonography in 28 patients. Demographic information, anthropometric, and blood pressure (BP) measurements were taken. Fasting blood samples were obtained to measure glucose, glycated haemoglobin, lipid profile, liver function tests, and highly sensitive C-reactive protein. Fatty liver index, hepatic steatosis index, NAFLD-liver fat score, and triglyceride and glucose index were calculated. Following stepwise forward likelihood ratio regression with independent variables included in one model using binary logistic regression with age and waist circumference (WC) entered as covariates, elevated diastolic BP and low high-density lipoprotein- cholesterol remained significantly associated with NAFLD (p = 0.002 and 0.03, respectively). However, none of the investigated indices could be used to diagnose the disease adequately due to low specificity, even after calculating new cut-off values. Investigating novel markers and adjusting existing equations used to calculate indices to improve sensitivity and specificity in our population is needed.
Collapse
|
11
|
Du Y, Su D, Ratnapradipa KL, Dong J, Rochling FA, Farazi PA. Factors Associated with Awareness and Knowledge of Nonalcoholic Fatty Liver Disease, a Liver Cancer Etiological Factor, Among Chinese Young Adults. JOURNAL OF CANCER EDUCATION : THE OFFICIAL JOURNAL OF THE AMERICAN ASSOCIATION FOR CANCER EDUCATION 2022:10.1007/s13187-022-02246-9. [PMID: 36527545 DOI: 10.1007/s13187-022-02246-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 12/04/2022] [Indexed: 06/17/2023]
Abstract
Despite the increasing prevalence of nonalcoholic fatty liver disease (NAFLD), a liver cancer etiological factor, among Chinese young adults (CYA), there is a lack of study assessing awareness and knowledge of NAFLD among them. This study aimed to assess awareness and knowledge of NAFLD, and identify the factors associated with awareness and knowledge of NAFLD among CYA. We conducted a survey to assess awareness and knowledge of NAFLD among students who enrolled in one university undergraduate program in 2018, 2019, and 2020 through a newly developed web-based, self-administered questionnaire. The outcomes included awareness of NAFLD and knowledge score of NAFLD. We conducted unadjusted and adjusted analyses of associated factors with these outcomes. A total of 1373 students participated in the survey. Of all participants, 26.2% reported that they had heard of NAFLD, 7.7% reported that they had heard of lean NAFLD, and 11.1% received ≥ 20 points in the knowledge section of NAFLD. The mean and median scores were 9.35 (Std = 7.67) and 9 (range 0-27). Factors associated with having awareness and knowledge (≥ 20 points) of NAFLD included medical background, high medical knowledge level, and family history of NAFLD. There was low NAFLD awareness and knowledge among CYA aged 18 to 25 years. Our findings point to the need to develop an educational intervention for CYA to improve their NAFLD awareness and knowledge. Further studies should include information on physical activity and dietary habits to test whether NAFLD awareness and knowledge impact those habits.
Collapse
Affiliation(s)
- Yi Du
- University of Nebraska Medical Center, 42nd and Emile, Omaha, NE, 68198-4395, USA
| | - Dejun Su
- University of Nebraska Medical Center, 42nd and Emile, Omaha, NE, 68198-4395, USA
| | | | - Jianghu Dong
- University of Nebraska Medical Center, 42nd and Emile, Omaha, NE, 68198-4395, USA
| | - Fedja A Rochling
- University of Nebraska Medical Center, 42nd and Emile, Omaha, NE, 68198-4395, USA
| | - Paraskevi A Farazi
- University of Nebraska Medical Center, 42nd and Emile, Omaha, NE, 68198-4395, USA.
| |
Collapse
|
12
|
Yang J, Yao W, Yang H, Shen Y, Zhang Y. Design and synthesis of ERα agonists: Effectively reduce lipid accumulation. Front Chem 2022; 10:1104249. [PMID: 36569962 PMCID: PMC9772986 DOI: 10.3389/fchem.2022.1104249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 11/28/2022] [Indexed: 12/13/2022] Open
Abstract
In recent years, the incidence of non-alcoholic fatty liver disease (NAFLD) has been increasing worldwide. Hepatic lipid deposition is a major feature of NAFLD, and insulin resistance is one of the most important causes of lipid deposition. Insulin resistance results in the disruption of lipid metabolism homeostasis characterized by increased lipogenesis and decreased lipolysis. Estrogen receptor α (ERα) has been widely reported to be closely related to lipid metabolism. Activating ERa may be a promising strategy to improve lipid metabolism. Here, we used computer-aided drug design technology to discover a highly active compound, YRL-03, which can effectively reduce lipid accumulation. Cellular experimental results showed that YRL-03 could effectively reduce lipid accumulation by targeting ERα, thereby achieving alleviation of insulin resistance. We believe this study provides meaningful guidance for future molecular development of drugs to prevent and treat NAFLD.
Collapse
Affiliation(s)
- Jinfei Yang
- School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao, China,*Correspondence: Jinfei Yang, ; Yuanyuan Zhang,
| | - Weiwei Yao
- School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao, China
| | - Huihui Yang
- School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao, China
| | - Yajing Shen
- School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao, China
| | - Yuanyuan Zhang
- West China School of Pharmacy, Sichuan University, Chengdu, China,*Correspondence: Jinfei Yang, ; Yuanyuan Zhang,
| |
Collapse
|
13
|
Konieczna J, Fiol M, Colom A, Martínez-González MÁ, Salas-Salvadó J, Corella D, Soria-Florido MT, Martínez JA, Alonso-Gómez ÁM, Wärnberg J, Vioque J, López-Miranda J, Estruch R, Bernal-López MR, Lapetra J, Serra-Majem L, Bueno-Cavanillas A, Tur JA, Martín Sánchez V, Pintó X, Gaforio JJ, Matía-Martín P, Vidal J, Vázquez C, Daimiel L, Ros E, Bes-Rastrollo M, Pascual M, Sorlí JV, Goday A, Zulet MÁ, Moreno-Rodriguez A, Carmona González FJ, Valls-Enguix R, Janer JM, Garcia-Rios A, Casas R, Gomez-Perez AM, Santos-Lozano JM, Basterra-Gortari FJ, Martínez MÁ, Ortega-Azorin C, Bayó J, Abete I, Salaverria-Lete I, Ruiz-Canela M, Babio N, Carres L, Romaguera D. Does Consumption of Ultra-Processed Foods Matter for Liver Health? Prospective Analysis among Older Adults with Metabolic Syndrome. Nutrients 2022; 14:4142. [PMID: 36235794 PMCID: PMC9570694 DOI: 10.3390/nu14194142] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 09/22/2022] [Accepted: 09/27/2022] [Indexed: 11/06/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) includes a spectrum of liver alterations that can result in severe disease and even death. Consumption of ultra-processed foods (UPF) has been associated with obesity and related comorbidities. However, the link between UPF and NAFLD has not been sufficiently assessed. We aimed to investigate the prospective association between UPF consumption and liver health biomarkers. Methods: We followed for 1 year 5867 older participants with overweight/obesity and metabolic syndrome (MetS) from the PREDIMED-Plus trial. A validated 143-item semi-quantitative food frequency questionnaire was used to evaluate consumption of UPF at baseline, 6, and 12 months. The degree of processing for foods and beverages (g/day) was established according to the NOVA classification system. The non-invasive fatty liver index (FLI) and hepatic steatosis index (HSI) were used to evaluate liver health at three points in time. The associations between changes in UPF consumption (percentage of total daily dietary intake (g)) and liver biomarkers were assessed using mixed-effects linear models with repeated measurements. Results: In this cohort, UPF consumption at baseline was 8.19% (SD 6.95%) of total daily dietary intake in grams. In multivariable models, each 10% daily increment in UPF consumption in 1 year was associated with significantly greater FLI (β 1.60 points, 95% CI 1.24;1.96 points) and HSI (0.43, 0.29; 0.57) scores (all p-values < 0.001). These associations persisted statistically significant after adjusting for potential dietary confounders and NAFLD risk factors. Conclusions: A higher UPF consumption was associated with higher levels of NAFLD-related biomarkers in older adults with overweight/obesity and MetS.
Collapse
Grants
- PI13/00673, PI13/00492, PI13/00272, PI13/01123, PI13/00462, PI13/00233, PI13/02184, PI13/00728, PI13/01090, PI13/01056, PI14/01722, PI14/00636, PI14/00618, PI14/00696, PI14/01206, PI14/01919, PI14/00853, PI14/01374, PI14/00972, PI14/00728, PI14/01471, PI1 Instituto de Salud Carlos III
- PI0458/2013, PS0358/2016, PI0137/2018 the Consejería de Salud de la Junta de Andalucía
- PROMETEO/2017/017 Generalitat Valenciana
- N/A SEMERGEN
- ICREA Academia programme Institució Catalana de Recerca i Estudis Avançats
- #340918 European Research Council
- N/A Especial Action Project
- 2013ACUP00194 Recercaixa
- IJC2019-042420-I Spanish Ministry of Economy, Industry and Competitiveness and European Social Funds
Collapse
Affiliation(s)
- Jadwiga Konieczna
- Research Group on Nutritional Epidemiology & Cardiovascular Physiopathology (NUTRECOR), Health Research Institute of the Balearic Islands (IdISBa), University Hospital Son Espases (HUSE), 07120 Palma de Mallorca, Spain
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Institute of Health Carlos III, 28029 Madrid, Spain
| | - Miguel Fiol
- Research Group on Nutritional Epidemiology & Cardiovascular Physiopathology (NUTRECOR), Health Research Institute of the Balearic Islands (IdISBa), University Hospital Son Espases (HUSE), 07120 Palma de Mallorca, Spain
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Institute of Health Carlos III, 28029 Madrid, Spain
| | - Antoni Colom
- Research Group on Nutritional Epidemiology & Cardiovascular Physiopathology (NUTRECOR), Health Research Institute of the Balearic Islands (IdISBa), University Hospital Son Espases (HUSE), 07120 Palma de Mallorca, Spain
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Institute of Health Carlos III, 28029 Madrid, Spain
| | - Miguel Ángel Martínez-González
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Institute of Health Carlos III, 28029 Madrid, Spain
- Department of Preventive Medicine and Public Health, IDISNA, University of Navarra, 31008 Pamplona, Spain
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Jordi Salas-Salvadó
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Institute of Health Carlos III, 28029 Madrid, Spain
- Universitat Rovira i Virgili, Departament de Bioquímica i Biotecnologia, Unitat de Nutrició, 43201 Reus, Spain
- University Hospital of Sant Joan de Reus, Nutrition Unit, 43204 Reus, Spain
- Institut d’Investigació Sanitària Pere Virgili (IISPV), 43204 Reus, Spain
| | - Dolores Corella
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Institute of Health Carlos III, 28029 Madrid, Spain
- Department of Preventive Medicine, University of Valencia,46010 Valencia, Spain
| | - María Trinidad Soria-Florido
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Institute of Health Carlos III, 28029 Madrid, Spain
- Unit of Cardiovascular Risk and Nutrition, Institut Hospital del Mar de Investigaciones Médicas Municipal d’Investigació Médica (IMIM), 08003 Barcelona, Spain
| | - J. Alfredo Martínez
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Institute of Health Carlos III, 28029 Madrid, Spain
- Department of Nutrition, Food Sciences, and Physiology, University of Navarra, IDISNA, 31008 Pamplona, Spain
- Precision Nutrition and Cardiometabolic Health Program, IMDEA Food, CEI UAM + CSIC, 28049 Madrid, Spain
| | - Ángel M. Alonso-Gómez
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Institute of Health Carlos III, 28029 Madrid, Spain
- Bioaraba Health Research Institute, Cardiovascular, Respiratory and Metabolic Area, Osakidetza Basque Health Service, Araba University Hospital, University of the Basque Country UPV/EHU, 01009 Vitoria-Gasteiz, Spain
| | - Julia Wärnberg
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Institute of Health Carlos III, 28029 Madrid, Spain
- Department of Nursing, University of Málaga, Institute of Biomedical Research in Malaga (IBIMA), 29016 Málaga, Spain
| | - Jesús Vioque
- CIBER de Epidemiología y Salud Pública (CIBERESP), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Alicante Institute for Health and Biomedical Research (ISABIAL-UMH), 03010 Alicante, Spain
| | - José López-Miranda
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Institute of Health Carlos III, 28029 Madrid, Spain
- Department of Internal Medicine, Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Reina Sofia University Hospital, University of Cordoba, 14004 Cordoba, Spain
| | - Ramon Estruch
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Institute of Health Carlos III, 28029 Madrid, Spain
- Department of Internal Medicine, Institut d’Investigacions Biomèdiques August Pi Sunyer (IDIBAPS), Hospital Clinic, University of Barcelona, 08007 Barcelona, Spain
| | - M. Rosa Bernal-López
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Institute of Health Carlos III, 28029 Madrid, Spain
- Department of Internal Medicine, Regional University Hospital of Malaga, Instituto de Investigación Biomédica de Málaga (IBIMA), University of Málaga, 29009 Málaga, Spain
| | - José Lapetra
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Institute of Health Carlos III, 28029 Madrid, Spain
- Department of Family Medicine, Research Unit, Distrito Sanitario Atención Primaria Sevilla, 41013 Sevilla, Spain
| | - Lluís Serra-Majem
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Institute of Health Carlos III, 28029 Madrid, Spain
- Research Institute of Biomedical and Health Sciences (IUIBS), University of Las Palmas de Gran Canaria & Centro Hospitalario Universitario Insular Materno Infantil (CHUIMI), Canarian Health Service, 35001 Las Palmas de Gran Canaria, Spain
| | - Aurora Bueno-Cavanillas
- CIBER de Epidemiología y Salud Pública (CIBERESP), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Department of Preventive Medicine and Public Health, University of Granada, 18011 Granada, Spain
| | - Josep A. Tur
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Institute of Health Carlos III, 28029 Madrid, Spain
- Research Group on Community Nutrition & Oxidative Stress, University of Balearic Islands, 07122 Palma de Mallorca, Spain
| | - Vicente Martín Sánchez
- CIBER de Epidemiología y Salud Pública (CIBERESP), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Institute of Biomedicine (IBIOMED), University of León, 24071 León, Spain
| | - Xavier Pintó
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Institute of Health Carlos III, 28029 Madrid, Spain
- Lipids and Vascular Risk Unit, Internal Medicine, Hospital Universitario de Bellvitge Idibell-UB, Hospitalet de Llobregat, 08907 Barcelona, Spain
| | - José J. Gaforio
- CIBER de Epidemiología y Salud Pública (CIBERESP), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Departamento de Ciencias de la Salud, Centro de Estudios Avanzados en Olivar y Aceites de Oliva, Universidad de Jaén, 23071 Jaén, Spain
| | - Pilar Matía-Martín
- Department of Endocrinology and Nutrition, Instituto de Investigación Sanitaria Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain
| | - Josep Vidal
- CIBER Diabetes y Enfermedades Metabólicas (CIBERDEM), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain
- Department of Endocrinology, Institut d’ Investigacions Biomédiques August Pi Sunyer (IDIBAPS), Hospital Clinic, University of Barcelona, 08007 Barcelona, Spain
| | - Clotilde Vázquez
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Institute of Health Carlos III, 28029 Madrid, Spain
- Department of Endocrinology and Nutrition, Hospital Fundación Jimenez Díaz, Instituto de Investigaciones Biomédicas IISFJD, University Autonoma, 28040 Madrid, Spain
| | - Lidia Daimiel
- Nutritional Control of the Epigenome Group, Precision Nutrition and Obesity Program, IMDEA Food, CEI UAM + CSIC, 28049 Madrid, Spain
| | - Emilio Ros
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Institute of Health Carlos III, 28029 Madrid, Spain
- Lipid Clinic, Department of Endocrinology and Nutrition, IDIBAPS, Hospital Clínic, 08036 Barcelona, Spain
| | - Maira Bes-Rastrollo
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Institute of Health Carlos III, 28029 Madrid, Spain
- Department of Preventive Medicine and Public Health, IDISNA, University of Navarra, 31008 Pamplona, Spain
| | - María Pascual
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Institute of Health Carlos III, 28029 Madrid, Spain
- Universitat Rovira i Virgili, Departament de Bioquímica i Biotecnologia, Unitat de Nutrició, 43201 Reus, Spain
- University Hospital of Sant Joan de Reus, Nutrition Unit, 43204 Reus, Spain
- Institut d’Investigació Sanitària Pere Virgili (IISPV), 43204 Reus, Spain
| | - Jose V. Sorlí
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Institute of Health Carlos III, 28029 Madrid, Spain
- Department of Preventive Medicine, University of Valencia,46010 Valencia, Spain
| | - Albert Goday
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Institute of Health Carlos III, 28029 Madrid, Spain
- Unit of Cardiovascular Risk and Nutrition, Institut Hospital del Mar de Investigaciones Médicas Municipal d’Investigació Médica (IMIM), 08003 Barcelona, Spain
- Departament de Medicina, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain
| | - María Ángeles Zulet
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Institute of Health Carlos III, 28029 Madrid, Spain
- Department of Nutrition, Food Sciences, and Physiology, University of Navarra, IDISNA, 31008 Pamplona, Spain
| | - Anai Moreno-Rodriguez
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Institute of Health Carlos III, 28029 Madrid, Spain
- Bioaraba Health Research Institute, Cardiovascular, Respiratory and Metabolic Area, Osakidetza Basque Health Service, Araba University Hospital, University of the Basque Country UPV/EHU, 01009 Vitoria-Gasteiz, Spain
| | - Francisco Jesús Carmona González
- Unidad de Gestión Clínica Torrequebrada, Distrito de Atención Primaria Costa del Sol, Servicio Andaluz de Salud, 29640 Benalmádena, Spain
| | | | - Juana M. Janer
- Research Group on Nutritional Epidemiology & Cardiovascular Physiopathology (NUTRECOR), Health Research Institute of the Balearic Islands (IdISBa), University Hospital Son Espases (HUSE), 07120 Palma de Mallorca, Spain
- Centro de Salud Camp Redó (UBS Son Sardina) Gerència Atenció Primària de Mallorca, 07010 Palma de Mallorca, Spain
| | - Antonio Garcia-Rios
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Institute of Health Carlos III, 28029 Madrid, Spain
- Department of Internal Medicine, Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Reina Sofia University Hospital, University of Cordoba, 14004 Cordoba, Spain
| | - Rosa Casas
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Institute of Health Carlos III, 28029 Madrid, Spain
- Department of Internal Medicine, Institut d’Investigacions Biomèdiques August Pi Sunyer (IDIBAPS), Hospital Clinic, University of Barcelona, 08007 Barcelona, Spain
| | - Ana M. Gomez-Perez
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Institute of Health Carlos III, 28029 Madrid, Spain
- Virgen de la Victoria Hospital, Department of Endocrinology, Instituto de Investigación Biomédica de Málaga (IBIMA), University of Málaga, 29071 Málaga, Spain
| | - José Manuel Santos-Lozano
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Institute of Health Carlos III, 28029 Madrid, Spain
- Department of Family Medicine, Research Unit, Distrito Sanitario Atención Primaria Sevilla, 41013 Sevilla, Spain
| | - F. Javier Basterra-Gortari
- Department of Preventive Medicine and Public Health, IDISNA, University of Navarra, 31008 Pamplona, Spain
- Servicio de Endocrinología. Complejo Hospitalario de Navarra, Servicio Navarro de Salud, 31003 Pamplona, Spain
| | - María Ángeles Martínez
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Institute of Health Carlos III, 28029 Madrid, Spain
- Universitat Rovira i Virgili, Departament de Bioquímica i Biotecnologia, Unitat de Nutrició, 43201 Reus, Spain
- University Hospital of Sant Joan de Reus, Nutrition Unit, 43204 Reus, Spain
- Institut d’Investigació Sanitària Pere Virgili (IISPV), 43204 Reus, Spain
| | - Carolina Ortega-Azorin
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Institute of Health Carlos III, 28029 Madrid, Spain
- Department of Preventive Medicine, University of Valencia,46010 Valencia, Spain
| | - Joan Bayó
- CAP El Clot, Institut Català de la Salut, 08018 Barcelona, Spain
| | - Itziar Abete
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Institute of Health Carlos III, 28029 Madrid, Spain
- Department of Nutrition, Food Sciences, and Physiology, University of Navarra, IDISNA, 31008 Pamplona, Spain
| | - Itziar Salaverria-Lete
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Institute of Health Carlos III, 28029 Madrid, Spain
- Bioaraba Health Research Institute, Cardiovascular, Respiratory and Metabolic Area, Osakidetza Basque Health Service, Araba University Hospital, University of the Basque Country UPV/EHU, 01009 Vitoria-Gasteiz, Spain
| | - Miguel Ruiz-Canela
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Institute of Health Carlos III, 28029 Madrid, Spain
- Department of Preventive Medicine and Public Health, IDISNA, University of Navarra, 31008 Pamplona, Spain
| | - Nancy Babio
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Institute of Health Carlos III, 28029 Madrid, Spain
- Universitat Rovira i Virgili, Departament de Bioquímica i Biotecnologia, Unitat de Nutrició, 43201 Reus, Spain
- University Hospital of Sant Joan de Reus, Nutrition Unit, 43204 Reus, Spain
- Institut d’Investigació Sanitària Pere Virgili (IISPV), 43204 Reus, Spain
| | - Lourdes Carres
- Atención Primaria Sant Martí, Institut Català de la Salut, 08020 Barcelona, Spain
| | - Dora Romaguera
- Research Group on Nutritional Epidemiology & Cardiovascular Physiopathology (NUTRECOR), Health Research Institute of the Balearic Islands (IdISBa), University Hospital Son Espases (HUSE), 07120 Palma de Mallorca, Spain
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Institute of Health Carlos III, 28029 Madrid, Spain
| |
Collapse
|
14
|
Dementia and non-alcoholic fatty liver disease – An unprecedented relationship. Ann Med Surg (Lond) 2022; 81:104359. [PMID: 36147178 PMCID: PMC9486664 DOI: 10.1016/j.amsu.2022.104359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Accepted: 08/11/2022] [Indexed: 11/23/2022] Open
|
15
|
Roy N, Alencastro F, Roseman BA, Wilson SR, Delgado ER, May MC, Bhushan B, Bello FM, Jurczak MJ, Shiva S, Locker J, Gingras S, Duncan AW. Dysregulation of Lipid and Glucose Homeostasis in Hepatocyte-Specific SLC25A34 Knockout Mice. THE AMERICAN JOURNAL OF PATHOLOGY 2022; 192:1259-1281. [PMID: 35718058 PMCID: PMC9472157 DOI: 10.1016/j.ajpath.2022.06.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 05/18/2022] [Accepted: 06/08/2022] [Indexed: 10/18/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is an epidemic affecting 30% of the US population. It is characterized by insulin resistance, and by defective lipid metabolism and mitochondrial dysfunction in the liver. SLC25A34 is a major repressive target of miR-122, a miR that has a central role in NAFLD and liver cancer. However, little is known about the function of SLC25A34. To investigate SLC25A34 in vitro, mitochondrial respiration and bioenergetics were examined using hepatocytes depleted of Slc25a34 or overexpressing Slc25a34. To test the function of SLC25A34 in vivo, a hepatocyte-specific knockout mouse was generated, and loss of SLC25A34 was assessed in mice maintained on a chow diet and a fast-food diet (FFD), a model for NAFLD. Hepatocytes depleted of Slc25a34 displayed increased mitochondrial biogenesis, lipid synthesis, and ADP/ATP ratio; Slc25a34 overexpression had the opposite effect. In the knockout model on chow diet, SLC25A34 loss modestly affected liver function (altered glucose metabolism was the most pronounced defect). RNA-sequencing revealed changes in metabolic processes, especially fatty acid metabolism. After 2 months on FFD, knockouts had a more severe phenotype, with increased lipid content and impaired glucose tolerance, which was attenuated after longer FFD feeding (6 months). This work thus presents a novel model for studying SLC25A34 in vivo in which SLC25A34 plays a role in mitochondrial respiration and bioenergetics during NAFLD.
Collapse
Affiliation(s)
- Nairita Roy
- Department of Pathology, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; McGowan Institute for Regenerative Medicine, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; Pittsburgh Liver Research Center, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Frances Alencastro
- Department of Pathology, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; McGowan Institute for Regenerative Medicine, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; Pittsburgh Liver Research Center, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Bayley A Roseman
- Department of Pathology, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; McGowan Institute for Regenerative Medicine, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; Pittsburgh Liver Research Center, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Sierra R Wilson
- Department of Pathology, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; McGowan Institute for Regenerative Medicine, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; Pittsburgh Liver Research Center, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Evan R Delgado
- Department of Pathology, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; McGowan Institute for Regenerative Medicine, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; Pittsburgh Liver Research Center, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Meredith C May
- Department of Pathology, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; McGowan Institute for Regenerative Medicine, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; Pittsburgh Liver Research Center, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Bharat Bhushan
- Department of Pathology, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; Pittsburgh Liver Research Center, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Fiona M Bello
- Pittsburgh Liver Research Center, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; Department of Medicine, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Michael J Jurczak
- Pittsburgh Liver Research Center, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; Department of Medicine, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Sruti Shiva
- Departments of Pharmacology and Chemical Biology, Vascular Medicine Institute, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Joseph Locker
- Department of Pathology, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; Pittsburgh Liver Research Center, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Sebastien Gingras
- Department of Immunology, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Andrew W Duncan
- Department of Pathology, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; McGowan Institute for Regenerative Medicine, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; Pittsburgh Liver Research Center, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; Department of Bioengineering, School of Engineering, University of Pittsburgh, Pittsburgh, Pennsylvania.
| |
Collapse
|
16
|
Tang Y, Wei ZM, Li N, Sun LL, Jin ZY, Wu Z, Sun H. Quantitative analysis of the risk of type 2 diabetes and fatty liver in non-obese individuals by computed tomography. Abdom Radiol (NY) 2022; 47:2099-2105. [PMID: 35389075 DOI: 10.1007/s00261-022-03506-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 03/19/2022] [Accepted: 03/21/2022] [Indexed: 11/25/2022]
Abstract
PURPOSE To explore the risk of fatty liver and type 2 diabetes with quantitative parameters of abdominal computed tomography (CT) in a non-obese population. METHODS A retrospective analysis of abdominal CT and hospitalization records of inpatients admitted from May 2019 to May 2021 were divided into a non-obese control group (n = 143 cases) and a non-obese diabetes group (n = 105 cases). The measured abdominal CT parameters included body width, liver and spleen CT values, and the ratio of the liver CT value to the spleen CT value (L/S ratio). Logistic regression was used to analyze the risk factors for diabetes in non-obese individuals. RESULTS Three variables including body width (P < 0.001), liver CT value (P = 0.013), and L/S ratio (P = 0.002) were significantly correlated with the presence of diabetes in non-obese individuals. CONCLUSION Body width, liver CT value, and L/S ratio can be used to indicate the risk of type 2 diabetes in non-obese individuals.
Collapse
Affiliation(s)
- Yi Tang
- Department of Endocrinology, Fushun Central Hospital, Fushun, 113006, Liaoning Province, China
| | - Ze-Min Wei
- Department of Endocrinology, Fushun Central Hospital, Fushun, 113006, Liaoning Province, China
| | - Ning Li
- Department of Radiology, Fushun Central Hospital, Fushun, 113006, Liaoning Province, China
| | - Lin-Lin Sun
- Department of Endocrinology, Fushun Central Hospital, Fushun, 113006, Liaoning Province, China
| | - Zheng-Yu Jin
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Zhe Wu
- Department of Radiology, Fushun Central Hospital, Fushun, 113006, Liaoning Province, China.
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, 100730, China.
| | - Hao Sun
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, 100730, China.
| |
Collapse
|
17
|
Eugenol alleviated nonalcoholic fatty liver disease in rat via a gut-brain-liver axis involving glucagon-like Peptide-1. Arch Biochem Biophys 2022; 725:109269. [PMID: 35508252 DOI: 10.1016/j.abb.2022.109269] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 04/27/2022] [Accepted: 04/28/2022] [Indexed: 12/16/2022]
|
18
|
Baig MH, Gupta P, Khan MI, Alajmi MF, Hussain A, Hassan MI, Dong JJ. Probing the interaction of Selonsertib with human serum albumin: In silico and in vitro approaches. Curr Top Med Chem 2022; 22:879-890. [PMID: 35352662 DOI: 10.2174/1568026622666220330012032] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 12/30/2021] [Accepted: 01/18/2022] [Indexed: 11/22/2022]
Abstract
Introduction - Selonsertib, the most recently developed selective inhibitor of apoptosis signal-regulating kinase 1, was found to be highly productive in patients with stage 2 or 3 non-alcoholic steatohepatitis. We elucidated the binding characteristics, mechanism of interaction, and dynamic behaviors of selonsertib with human serum albumin (HSA), a major circulatory transport protein. Method- Different biophysical approaches (fluorescence quenching and isothermal titration calorimetry (ITC) were combined with various in silico techniques to examine the binding of selonsertib to HSA. Molecular docking results, analysis of molecular dynamics trajectories, and essential dynamics investigations indicated the stable binding of selonsertib to HSA. Further in vitro studies were performed to validate the observed interaction. Result- ITC results confirmed the robust binding and high affinity of selonsertib and HSA. Likewise, the fluorescence quenching results highlighted the binding affinity of selonsertib and HSA. Collectively, our findings offer deeper insight into the binding mechanism of selonsertib and HSA, emphasizing the selonsertib-mediated structural changes within HSA, along with a comprehensive rationale for the biological transport and accumulation of selonsertib in the blood plasma. Conclusion- Therefore, considering the bioavailability and effectiveness of selonsertib, assessing the interactions of this inhibitor with carrier proteins is crucial to elucidate its biological processes at the molecular level. This evidence carries the considerable scientific potential for future drug design.
Collapse
Affiliation(s)
- Mohammad Hassan Baig
- Department of Family Medicine, Yonsei University College of Medicine, Gangnam Severance Hospital, 211 Eonju-ro, Gangnam-gu, Seoul, Republic of Korea
| | - Preeti Gupta
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi- 110025, INDIA
| | - Mohd Imran Khan
- Department of Internal Medicine, Yonsei University College of Medicine, Gangnam Severance Hospital, 211 Eonju-ro, Gangnam-gu, Seoul, Republic of Korea
| | - Mohamed F Alajmi
- Department of Pharmacognosy College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Afzal Hussain
- Department of Pharmacognosy College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Md Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi- 110025, INDIA
| | - Jae-June Dong
- Department of Family Medicine, Yonsei University College of Medicine, Gangnam Severance Hospital, 211 Eonju-ro, Gangnam-gu, Seoul, Republic of Korea
| |
Collapse
|
19
|
Kathak RR, Sumon AH, Molla NH, Hasan M, Miah R, Tuba HR, Habib A, Ali N. The association between elevated lipid profile and liver enzymes: a study on Bangladeshi adults. Sci Rep 2022; 12:1711. [PMID: 35110625 PMCID: PMC8810783 DOI: 10.1038/s41598-022-05766-y] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Accepted: 01/14/2022] [Indexed: 12/14/2022] Open
Abstract
Dyslipidemia, a major contributor to cardiovascular diseases, is rapidly increasing in Asian countries including Bangladesh. In addition to the cardiovascular system, abnormal lipid levels are also known to cause complications in renal and hepatic systems. The data regarding dyslipidemia and its relationship with liver enzymes are scarce for the Bangladeshi population. Therefore, this study was conducted to estimate the prevalence of dyslipidemia and determine the relationship between lipid profile and liver enzymes in Bangladeshi adults. A total of 405 participants (318 males and 87 females) were enrolled in the study. Serum levels of TG, TC, LDL, HDL and liver enzymes including ALT, AST, GGT and ALP were analyzed using standard methods. Dyslipidemia and liver function tests abnormalities were defined according to the international standard guidelines. The association between elevated lipid profile markers and liver enzyme abnormalities was assessed by logistic regression analysis. Overall, the prevalence of elevated TG, TC, LDL and low HDL were 30.9%, 23.7%, 26.2% and 78.8%, respectively. On the other hand, the prevalence of elevated liver enzymes ALT, AST, GGT and ALP were 18.8%, 21.6%, 12.9% and 21.9%, respectively. Dyslipidemia and liver enzyme abnormalities were higher in diabetic and hypertensive participants than in the healthy participants. About 61% of participants with dyslipidemia had at least one or more elevated liver enzymes. In regression analysis, an independent association was observed between serum GGT and all lipid components. In conclusion, a high prevalence of dyslipidemia and liver enzyme abnormalities were observed among the study participants. Of the four liver enzymes, the serum levels of GGT showed an independent association with all lipid components. Moreover, this study indicates that subjects with dyslipidemia often have a higher chance of having liver diseases than subjects with no dyslipidemia. However, large-scale prospective studies are needed to understand the underlying mechanisms of lipid-induced hepatic dysfunction in the Bangladeshi population.
Collapse
Affiliation(s)
- Rahanuma Raihanu Kathak
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet, 3114, Bangladesh
| | - Abu Hasan Sumon
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet, 3114, Bangladesh
| | - Noyan Hossain Molla
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet, 3114, Bangladesh
| | - Mahmudul Hasan
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet, 3114, Bangladesh
| | - Rakib Miah
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet, 3114, Bangladesh
| | - Humaira Rashid Tuba
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet, 3114, Bangladesh
| | - Ahsan Habib
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet, 3114, Bangladesh
| | - Nurshad Ali
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet, 3114, Bangladesh.
| |
Collapse
|
20
|
Taylor JM, Li A, McLachlan CS. Immune cell profile and immune-related gene expression of obese peripheral blood and liver tissue. FEBS Lett 2022; 596:199-210. [PMID: 34850389 DOI: 10.1002/1873-3468.14248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 11/24/2021] [Accepted: 11/26/2021] [Indexed: 11/10/2022]
Abstract
Obesity is associated with changes in immune cell subpopulations. However, tissue and blood obesity-responsive immune phenotypic pathways have not been contrasted. Here, the local niche immune cell population and gene expression in fatty liver is compared to peripheral blood of obese individuals. The Cibersort algorithm enumerated increased fractions of memory CD4+ T lymphocytes and reductions in natural killer and memory B cells in obese liver tissue and obese blood, with similar reductions found in nonalcoholic fatty liver disease tissue. Gene expression analysis identified inflammatory immune signatures of regulatory CD4+ T cells with inferred Th1, Th17, Th2, or Treg phenotypes that differed between liver and blood. Our study suggests that the local tissue-specific immune phenotype in the liver differs from the obese peripheral circulation, with the latter reflective of multisystemic persistent inflammation that is characteristic of obesity.
Collapse
Affiliation(s)
- Jude M Taylor
- Centre for Healthy Futures, Torrens University Australia, Pyrmont, Australia
| | - Amy Li
- Centre for Healthy Futures, Torrens University Australia, Pyrmont, Australia
- Department of Pharmacy & Biomedical Sciences, La Trobe University, Bendigo, Australia
| | - Craig S McLachlan
- Centre for Healthy Futures, Torrens University Australia, Pyrmont, Australia
| |
Collapse
|
21
|
Lai JL, Lian YE, Wu JY, Wang YD, Bai YN. Verapamil induces autophagy to improve liver regeneration in non-alcoholic fatty liver mice. Adipocyte 2021; 10:532-545. [PMID: 34699301 PMCID: PMC8555529 DOI: 10.1080/21623945.2021.1983241] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 09/15/2021] [Accepted: 09/16/2021] [Indexed: 12/13/2022] Open
Abstract
Verapamil can restore intracellular calcium homeostasis, increase the fusion of autophagosomes and lysosomes, reduce lipid droplet accumulation and inhibit inflammation and insulin resistance in high-fat-fed mice. The present study aimed to investigate verapamil's effect and its underlying liver regeneration mechanism in mice with non-alcoholic fatty liver. After 50% hepatectomy was performed, the changes of autophagy and liver regeneration were evaluated by detecting cell proliferation and autophagy at each time point. Then, 25mg/kg verapamil was injected intraperitoneally for 10 d before an operation in the mild to moderate fatty liver and severe fatty liver groups. The control group and mild to moderate fatty liver group reached the peak of proliferation at 24-48h after operation, and the mice with severe fatty liver and steatohepatitis reached the peak at 48-72h. Autophagy in the normal group and mild to moderate fatty liver group reached the peak 48 hours after operation. Verapamil injection can enhance autophagy, reduce the weight of fatty liver mice, improve liver function and liver regeneration. Verapamil can induce autophagy, improve hepatocyte function and promote hepatocyte regeneration through the mTOR independent signaling pathway, thus improving the process of liver regeneration after partial hepatectomy.
Collapse
Affiliation(s)
- Jian-Lin Lai
- Shengli Clinical Medical College of Fujian Medical University, Department of Hepatobiliary and Pancreatic Surgery, Fujian Provincial Hospital, Fuzhou, Fujian, 350001, China
| | - Yuan-E Lian
- Department of Pathology, The Affiliated Union Hospital of Fujian Medical University, Fuzhou, 350001, China
| | - Jun-Yi Wu
- Shengli Clinical Medical College of Fujian Medical University, Department of Hepatobiliary and Pancreatic Surgery, Fujian Provincial Hospital, Fuzhou, Fujian, 350001, China
| | - Yao-Dong Wang
- Shengli Clinical Medical College of Fujian Medical University, Department of Hepatobiliary and Pancreatic Surgery, Fujian Provincial Hospital, Fuzhou, Fujian, 350001, China
| | - Yan-Nan Bai
- Shengli Clinical Medical College of Fujian Medical University, Department of Hepatobiliary and Pancreatic Surgery, Fujian Provincial Hospital, Fuzhou, Fujian, 350001, China
| |
Collapse
|
22
|
Li Y, Yang Y, Li Y, Zhang P, Ge G, Jin J, Du T, Ma M, Na L, Ding L, Sheng H. Use of GP73 in the diagnosis of non-alcoholic steatohepatitis and the staging of hepatic fibrosis. J Int Med Res 2021; 49:3000605211055378. [PMID: 34772312 PMCID: PMC8593324 DOI: 10.1177/03000605211055378] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 10/06/2021] [Indexed: 12/22/2022] Open
Abstract
OBJECTIVE To evaluate the utility of Golgi protein 73 (GP73) in the diagnosis of non-alcoholic steatohepatitis (NASH) and hepatic fibrosis (HF) staging. METHODS Ninety-one patients with non-alcoholic fatty liver disease (NAFLD) were allocated to NAFL (n = 46) and NASH (n = 45) groups according to their NAFLD activity score (NAS), and there were 30 healthy controls. Serum GP73 was measured by ELISA, GP73 protein expression was evaluated using immunohistochemistry, and FibroScan was used to determine liver hardness. RESULTS The serum GP73 concentrations of the NAFL and NASH groups were significantly higher than those of controls. GP73 expression in the liver of the patients gradually progressed from absent or low to moderate or high. Serum GP73 positively correlated with liver expression, and the serum and liver GP73 of the patients positively correlated with FibroScan value and HF stage. There was a strong positive correlation of the combination of alanine aminotransferase, gamma glutamyl transferase and GP73 with NASH. The combination of serum GP73 and FibroScan value was found to predict NASH (NAS > 4) and advanced HF (stage ≥2) in patients with NAFLD using receiver operating characteristic analysis. CONCLUSION Serum GP73 may be useful in the diagnosis of NASH and the staging of HF.
Collapse
Affiliation(s)
- Yadi Li
- Department of Clinical Medicine, Ningxia Medical University, Ningxia Medical University, Yinchuan, China
| | - Yan Yang
- Department of Infectious Diseases, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Yufang Li
- Department of Infectious Diseases, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Ping Zhang
- Department of Infectious Diseases, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Gaiying Ge
- Department of Clinical Medicine, Ningxia Medical University, Ningxia Medical University, Yinchuan, China
| | - Jing Jin
- Department of Clinical Medicine, Ningxia Medical University, Ningxia Medical University, Yinchuan, China
| | - Ting Du
- Department of Clinical Medicine, Ningxia Medical University, Ningxia Medical University, Yinchuan, China
| | - Maiyan Ma
- Department of Clinical Medicine, Ningxia Medical University, Ningxia Medical University, Yinchuan, China
| | - Li Na
- Biobank of General Hospital of Ningxia Medical University, Yinchuan, China
| | - Lu Ding
- Biobank of General Hospital of Ningxia Medical University, Yinchuan, China
| | - Huiping Sheng
- Department of Infectious Diseases, General Hospital of Ningxia Medical University, Yinchuan, China
| |
Collapse
|
23
|
Padda J, Khalid K, Khedr A, Tasnim F, Al-Ewaidat OA, Cooper AC, Jean-Charles G. Non-Alcoholic Fatty Liver Disease and Its Association With Diabetes Mellitus. Cureus 2021; 13:e17321. [PMID: 34557367 PMCID: PMC8449987 DOI: 10.7759/cureus.17321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/20/2021] [Indexed: 11/29/2022] Open
Abstract
There is a bidirectional relationship between non-alcoholic fatty liver disease (NAFLD) and type 2 diabetes mellitus (T2DM). The liver has a vital role in the pathophysiology of both diseases as it leads to the development of insulin resistance (IR), which in turn results in NAFLD and T2DM. It has been shown that T2DM increases the risk of NAFLD progression. Furthermore, the presence of NAFLD raises the probability of T2DM complications, which explains the increased rates of NAFLD screening in patients with T2DM. In addition, there are common management options for the two diseases. Lifestyle changes can play a role in the initial management of both diseases. Medications that are used to treat T2DM are also used in the management of NAFLD, such as metformin, thiazolidinediones (TZD), glucagon-like peptide-1 (GLP-1) analogues, and dipeptidyl peptidase-4 (DPP4) inhibitors. Bariatric surgery is often used as a last resort and has shown promising results. Lifestyle interventions with diet and exercise are important postoperatively to maintain the weight loss. There are many novel treatments that are being investigated for the treatment of NAFLD, targeting multiple pathophysiologic pathways. This review aims to shed some light on the intricate relationship between NAFLD and T2DM and how IR links both diseases. We also try to raise awareness among clinicians about this relationship and how the presence of one disease should raise a high index of suspicion for the existence of the other.
Collapse
Affiliation(s)
| | | | - Anwar Khedr
- Internal Medicine, JC Medical Center, Orlando, USA
| | | | | | | | - Gutteridge Jean-Charles
- Internal Medicine, JC Medical Center, Orlando, USA.,Internal Medicine, Advent Health & Orlando Health Hospital, Orlando, USA
| |
Collapse
|
24
|
Navarroza AMC, Wong SN. Comparison of clinical and metabolic profiles of lean versus non-lean nonalcoholic fatty liver disease. Indian J Gastroenterol 2021; 40:380-388. [PMID: 34213749 DOI: 10.1007/s12664-021-01184-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 04/19/2021] [Indexed: 02/08/2023]
Abstract
BACKGROUND Data on nonalcoholic fatty liver disease (NAFLD) in the Philippines are scarce. We aimed to compare the clinical and biochemical profiles of lean (BMI<23) vs. non-lean (BMI≥23) NAFLD patients. METHODS Consecutive patients diagnosed with NAFLD on ultrasound in two outpatient hepatology clinics from February 2007-January 2017 were included. Patients with significant alcohol intake, alternative causes of steatosis, and incomplete data were excluded. RESULTS A total of 663 patients (57.9% male) were included. Most patients were non-lean (88.1%) and had an elevated alanine aminotransferase (ALT) (63%). Cirrhosis or hepatocellular carcinoma (HCC) were already present in 8.4% on initial consultation. Concomitant hepatitis B was equally common in patients with and without cirrhosis (20.7% vs. 17.5%; p=0.660) or HCC (17.9% vs. 12.8%; p=0.415). Independent factors associated with HCC/cirrhosis on initial consultation were older age (OR=1.038), low albumin (OR=0.428), high BARD score (BMI, AST/ALT ratio, T2 diabetes mellitus; OR=2.548) and the presence of symptoms (OR=1.808). Compared to lean NAFLD patients, non-lean patients were more likely to be younger (51.5±14.4 vs. 55±14.3; p=0.003), have DM (47.9% vs. 29.1%; p=0.002), hypertension (57.5% vs. 38%; p=0.001), dyslipidemia (73.1% vs. 54.4%; p=0.001) and metabolic syndrome (60.3% vs. 30.4%; p<0.0001), abnormal metabolic parameters (LDL-C, HDL-C, triglycerides, uric acid and FBS), and with elevated ALT (65.2% vs. 46.8%; p=0.002) and AST (41.1±29.6 vs. 35.3±28.3; p=0.008). CONCLUSIONS The proportion of lean NAFLD was 11.9%. Although metabolic derangements and its clinical consequences were present in about a third of lean patients, these were still more common in non-lean NAFLD. Cirrhosis or HCC were already present in a significant proportion (8.4%) of NAFLD patients on initial presentation.
Collapse
Affiliation(s)
| | - Stephen N Wong
- Section of Gastroenterology, University of Santo Tomas Hospital, Manila, Philippines. .,Section of Gastroenterology, Chinese General Hospital and Medical Center, Manila, Philippines.
| |
Collapse
|
25
|
Shi Y, Pizzini J, Wang H, Das F, Abdul Azees PA, Ghosh Choudhury G, Barnes JL, Zang M, Weintraub ST, Yeh CK, Katz MS, Kamat A. β2-Adrenergic receptor agonist induced hepatic steatosis in mice: modeling nonalcoholic fatty liver disease in hyperadrenergic states. Am J Physiol Endocrinol Metab 2021; 321:E90-E104. [PMID: 34029162 PMCID: PMC8321826 DOI: 10.1152/ajpendo.00651.2020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 04/23/2021] [Accepted: 05/08/2021] [Indexed: 12/11/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a spectrum of disorders ranging from hepatic steatosis [excessive accumulation of triglycerides (TG)] to nonalcoholic steatohepatitis, which can progress to cirrhosis and hepatocellular carcinoma. The molecular pathogenesis of steatosis and progression to more severe NAFLD remains unclear. Obesity and aging, two principal risk factors for NAFLD, are associated with a hyperadrenergic state. β-Adrenergic responsiveness in liver increases in animal models of obesity and aging, and in both is linked to increased hepatic expression of β2-adrenergic receptors (β2-ARs). We previously showed that in aging rodents intracellular signaling from elevated hepatic levels of β2-ARs may contribute to liver steatosis. In this study we demonstrate that injection of formoterol, a highly selective β2-AR agonist, to mice acutely results in hepatic TG accumulation. Further, we have sought to define the intrahepatic mechanisms underlying β2-AR mediated steatosis by investigating changes in hepatic expression and cellular localization of enzymes, transcription factors, and coactivators involved in processes of lipid accrual and disposition-and also functional aspects thereof-in livers of formoterol-treated animals. Our results suggest that β2-AR activation by formoterol leads to increased hepatic TG synthesis and de novo lipogenesis, increased but incomplete β-oxidation of fatty acids with accumulation of potentially toxic long-chain acylcarnitine intermediates, and reduced TG secretion-all previously invoked as contributors to fatty liver disease. Experiments are ongoing to determine whether sustained activation of hepatic β2-AR signaling by formoterol might be utilized to model fatty liver changes occurring in hyperadrenergic states of obesity and aging, and thereby identify novel molecular targets for the prevention or treatment of NAFLD.NEW & NOTEWORTHY Results of our study suggest that β2-adrenergic receptor (β2-AR) activation by agonist formoterol leads to increased hepatic TG synthesis and de novo lipogenesis, incomplete β-oxidation of fatty acids with accumulation of long-chain acylcarnitine intermediates, and reduced TG secretion. These findings may, for the first time, implicate a role for β2-AR responsive dysregulation of hepatic lipid metabolism in the pathogenetic processes underlying NAFLD in hyperadrenergic states such as obesity and aging.
Collapse
Affiliation(s)
- Yun Shi
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - Jason Pizzini
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - Hanzhou Wang
- Department of Comprehensive Dentistry, University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - Falguni Das
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - Parveez Ahamed Abdul Azees
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas
- Department of Comprehensive Dentistry, University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - Goutam Ghosh Choudhury
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - Jeffrey L Barnes
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - Mengwei Zang
- Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, Texas
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, Texas
- Geriatric Research, Education and Clinical Center, Audie L. Murphy Division, South Texas Veterans Health Care System, San Antonio, Texas
| | - Susan T Weintraub
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - Chih-Ko Yeh
- Department of Comprehensive Dentistry, University of Texas Health Science Center at San Antonio, San Antonio, Texas
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, Texas
- Geriatric Research, Education and Clinical Center, Audie L. Murphy Division, South Texas Veterans Health Care System, San Antonio, Texas
| | - Michael S Katz
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas
- Geriatric Research, Education and Clinical Center, Audie L. Murphy Division, South Texas Veterans Health Care System, San Antonio, Texas
| | - Amrita Kamat
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, Texas
- Geriatric Research, Education and Clinical Center, Audie L. Murphy Division, South Texas Veterans Health Care System, San Antonio, Texas
| |
Collapse
|
26
|
Namkhah Z, Naeini F, Ostadrahimi A, Tutunchi H, Hosseinzadeh-Attar MJ. The association of the adipokine zinc-alpha2-glycoprotein with non-alcoholic fatty liver disease and related risk factors: A comprehensive systematic review. Int J Clin Pract 2021; 75:e13985. [PMID: 33404166 DOI: 10.1111/ijcp.13985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Accepted: 01/03/2021] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND AND AIM The adipokine zinc-alpha2-glycoprotein (ZAG), a multidisciplinary protein, is involved in lipid metabolism, glucose homeostasis and energy balance. Accumulating evidence demonstrates that the expression of ZAG is mainly downregulated in obesity and obesity-related conditions. In the present study, we assessed the association of ZAG with non-alcoholic fatty liver disease (NAFLD) and the related risk factors including obesity, metabolic factors and inflammatory parameters, with emphasis on potential mechanisms underlying these associations. METHODS PRISMA guidelines were followed in this review. Systematic searches were performed using the PubMed/Medline, ScienceDirect, Scopus, EMBASE, ProQuest and Google Scholar databases, up to August 2020 for all relevant published papers. RESULTS Out of 362 records screened, 34 articles were included in the final analysis. According to the studies reviewed here, ZAG appears to exert a protective effect against NAFLD by enhancing mRNA expression levels of peroxisome proliferator-activated receptor α (PPARα) and PPARγ, promoting mRNA expression levels of the lipolysis-related genes, reducing mRNA expression levels of the lipogenesis-related genes, increasing hepatic fatty acid oxidation, ameliorating hepatic steatosis, promoting the activity of brown adipose tissue and the expression of thermogenesis-related genes, modulating energy balance and glucose homeostasis, and elevating plasma levels of healthy adipokines such as adiponectin. ZAG can also be involved in the regulation of inflammatory responses by attenuation of the expression of pro-inflammatory and pro-fibrotic mediators. CONCLUSION According to the studies reviewed here, ZAG is suggested to be a promising therapeutic target for NAFLD. However, the favourable effects of ZAG need to be confirmed in prospective cohort studies.
Collapse
Affiliation(s)
- Zahra Namkhah
- Department of Clinical Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Science, Tehran, Iran
| | - Fatemeh Naeini
- Department of Clinical Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Science, Tehran, Iran
| | - Alireza Ostadrahimi
- Nutrition Research Center, Department of Clinical Nutrition, School of Nutrition & Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Helda Tutunchi
- Nutrition Research Center, Department of Clinical Nutrition, School of Nutrition & Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | | |
Collapse
|
27
|
Current Perspectives on the Beneficial Effects of Soybean Isoflavones and Their Metabolites for Humans. Antioxidants (Basel) 2021; 10:antiox10071064. [PMID: 34209224 PMCID: PMC8301030 DOI: 10.3390/antiox10071064] [Citation(s) in RCA: 128] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 06/27/2021] [Accepted: 06/28/2021] [Indexed: 12/13/2022] Open
Abstract
Soybeans are rich in proteins and lipids and have become a staple part of the human diet. Besides their nutritional excellence, they have also been shown to contain various functional components, including isoflavones, and have consequently received increasing attention as a functional food item. Isoflavones are structurally similar to 17-β-estradiol and bind to estrogen receptors (ERα and ERβ). The estrogenic activity of isoflavones ranges from a hundredth to a thousandth of that of estrogen itself. Isoflavones play a role in regulating the effects of estrogen in the human body, depending on the situation. Thus, when estrogen is insufficient, isoflavones perform the functions of estrogen, and when estrogen is excessive, isoflavones block the estrogen receptors to which estrogen binds, thus acting as an estrogen antagonist. In particular, estrogen antagonistic activity is important in the breast, endometrium, and prostate, and such antagonistic activity suppresses cancer occurrence. Genistein, an isoflavone, has cancer-suppressing effects on estrogen receptor-positive (ER+) cancers, including breast cancer. It suppresses the function of enzymes such as tyrosine protein kinase, mitogen-activated kinase, and DNA polymerase II, thus inhibiting cell proliferation and inducing apoptosis. Genistein is the most biologically active and potent isoflavone candidate for cancer prevention. Furthermore, among the various physiological functions of isoflavones, they are best known for their antioxidant activities. S-Equol, a metabolite of genistein and daidzein, has strong antioxidative effects; however, the ability to metabolize daidzein into S-equol varies based on racial and individual differences. The antioxidant activity of isoflavones may be effective in preventing dementia by inhibiting the phosphorylation of Alzheimer's-related tau proteins. Genistein also reduces allergic responses by limiting the expression of mast cell IgE receptors, which are involved in allergic responses. In addition, they have been known to prevent and treat various diseases, including cardiovascular diseases, metabolic syndromes, osteoporosis, diabetes, brain-related diseases, high blood pressure, hyperlipidemia, obesity, and inflammation. Further, it also has positive effects on menstrual irregularity in non-menopausal women and relieving menopausal symptoms in middle-aged women. Recently, soybean consumption has shown steep increasing trend in Western countries where the intake was previously only 1/20-1/50 of that in Asian countries. In this review, I have dealt with the latest research trends that have shown substantial interest in the biological efficacy of isoflavones in humans and plants, and their related mechanisms.
Collapse
|
28
|
Duan J, Song Y, Zhang X, Wang C. Effect of ω-3 Polyunsaturated Fatty Acids-Derived Bioactive Lipids on Metabolic Disorders. Front Physiol 2021; 12:646491. [PMID: 34113260 PMCID: PMC8185290 DOI: 10.3389/fphys.2021.646491] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Accepted: 04/26/2021] [Indexed: 12/23/2022] Open
Abstract
Arachidonic acid (ARA) is an important ω-6 polyunsaturated fatty acid (PUFA), and docosahexaenoic acid (DHA), eicosapentaenoic acid (EPA) and n-3 docosapentaenoic acid (n-3 DPA) are three well-known ω-3 PUFAs. These fatty acids can be metabolized into a number of bioactive lipids. Eicosanoids derived from ARA have drawn great attention because of their important and complex biofunctions. Although EPA, DHA and n-3 DPA have also shown powerful biofunctions, we have fewer studies of metabolites derived from them than those from ARA. Recently, growing research has focused on the bioaction of ω-3 PUFA-derived metabolites, which indicates their great potential for treating metabolic disorders. Most of the functional studies of these bioactive lipids focused on their anti-inflammatory effects. However, several studies elucidated their direct effects on pancreatic β cells, hepatocytes, adipocytes, skeletal muscle cells, and endothelial cells. These researches revealed the importance of studying the functions of metabolites derived from ω-3 polyunsaturated fatty acids other than themselves. The current review summarizes research into the effects of ω-3 PUFA-derived oxylipins on metabolic disorders, including diabetes, non-alcoholic fatty liver disease, adipose tissue dysfunction, and atherosclerosis.
Collapse
Affiliation(s)
- Jinjie Duan
- Department of Physiology and Pathophysiology, The Province and Ministry Co-Sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Medical University, Tianjin, China.,Tianjin Key Laboratory of Medical Epigenetics, Tianjin Medical University, Tianjin, China
| | - Yayue Song
- Department of Physiology and Pathophysiology, The Province and Ministry Co-Sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Medical University, Tianjin, China.,Tianjin Key Laboratory of Medical Epigenetics, Tianjin Medical University, Tianjin, China
| | - Xu Zhang
- Department of Physiology and Pathophysiology, The Province and Ministry Co-Sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Medical University, Tianjin, China
| | - Chunjiong Wang
- Department of Physiology and Pathophysiology, The Province and Ministry Co-Sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Medical University, Tianjin, China.,Tianjin Key Laboratory of Medical Epigenetics, Tianjin Medical University, Tianjin, China
| |
Collapse
|
29
|
Hossein Bagheri M, Azamian-Jazi A, Banitalebi E, Kazeminasab F, Hossein Nasr-Esfahani M. Both high-intensity interval training and low-intensity endurance training decrease intrahepatic lipid deposits via alterations of the expression of HIF-1α, HIG2 in a murine model of non alcoholic fatty liver disease (NAFLD). Sci Sports 2021. [DOI: 10.1016/j.scispo.2020.01.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
30
|
Diesinger T, Lautwein A, Buko V, Belonovskaya E, Lukivskaya O, Naruta E, Kirko S, Andreev V, Dvorsky R, Buckert D, Bergler S, Renz C, Müller‐Enoch D, Wirth T, Haehner T. ω-Imidazolyl-alkyl derivatives as new preclinical drug candidates for treating non-alcoholic steatohepatitis. Physiol Rep 2021; 9:e14795. [PMID: 33769703 PMCID: PMC7995547 DOI: 10.14814/phy2.14795] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 02/15/2021] [Accepted: 02/16/2021] [Indexed: 01/15/2023] Open
Abstract
Cytochrome P450 2E1 (CYP2E1)-associated reactive oxygen species production plays an important role in the development and progression of inflammatory liver diseases such as alcoholic steatohepatitis. We developed two new inhibitors for this isoenzyme, namely 12-imidazolyl-1-dodecanol (I-ol) and 1-imidazolyldodecane (I-an), and aimed to test their effects on non-alcoholic steatohepatitis (NASH). The fat-rich and CYP2E1 inducing Lieber-DeCarli diet was administered over 16 weeks of the experimental period to induce the disease in a rat model, and the experimental substances were administered simultaneously over the last four weeks. The high-fat diet (HFD) pathologically altered the balance of reactive oxygen species and raised the activities of the liver enzymes alanine aminotransferase (ALT), aspartate aminotransferase (AST), alkaline phosphatase (AP) and γ-glutamyl-transferase (γ-GT); lowered the level of adiponectine and raised the one of tumor necrosis factor (TNF)-α; increased the hepatic triglyceride and phospholipid content and diminished the serum HDL cholesterol concentration. Together with the histological findings, we concluded that the diet led to the development of NASH. I-ol and, to a lesser extent, I-an shifted the pathological values toward the normal range, despite the continued administration of the noxious agent (HFD). The hepatoprotective drug ursodeoxycholic acid (UDCA), which is used off-label in clinical practice, showed a lower effectiveness overall. I-ol, in particular, showed extremely good tolerability during the acute toxicity study in rats. Therefore, cytochrome P450 2E1 may be considered a suitable drug target, with I-ol and I-an being promising drug candidates for the treatment of NASH.
Collapse
Affiliation(s)
- Torsten Diesinger
- Chair of Biochemistry and Molecular MedicineFaculty of Health/School of MedicineWitten/Herdecke UniversityWittenGermany
- Department of Internal MedicineNeu‐Ulm HospitalNeu‐UlmGermany
- Institute of Physiological ChemistryUniversity of UlmUlmGermany
| | - Alfred Lautwein
- Institute of Physiological ChemistryUniversity of UlmUlmGermany
| | - Vyacheslav Buko
- Division of Biochemical PharmacologyInstitute of Biochemistry of Biologically Active CompoundsNational Academy of SciencesBulvar Leninskogo KomsomolaGrodnoBelarus
- Department of BiotechnologyUniversity of Medical SciencesBiałystokPoland
| | - Elena Belonovskaya
- Division of Biochemical PharmacologyInstitute of Biochemistry of Biologically Active CompoundsNational Academy of SciencesBulvar Leninskogo KomsomolaGrodnoBelarus
| | - Oksana Lukivskaya
- Division of Biochemical PharmacologyInstitute of Biochemistry of Biologically Active CompoundsNational Academy of SciencesBulvar Leninskogo KomsomolaGrodnoBelarus
| | - Elena Naruta
- Division of Biochemical PharmacologyInstitute of Biochemistry of Biologically Active CompoundsNational Academy of SciencesBulvar Leninskogo KomsomolaGrodnoBelarus
| | - Siarhei Kirko
- Division of Biochemical PharmacologyInstitute of Biochemistry of Biologically Active CompoundsNational Academy of SciencesBulvar Leninskogo KomsomolaGrodnoBelarus
| | - Viktor Andreev
- Department of Medical Biology and GeneticsGrodno State Medical UniversityGrodnoBelarus
| | - Radovan Dvorsky
- Institute of Biochemistry and Molecular Biology IIMedical Faculty of the Heinrich Heine University DüsseldorfDüsseldorfGermany
- Max Planck Institute of Molecular PhysiologyDortmundGermany
| | - Dominik Buckert
- Institute of Physiological ChemistryUniversity of UlmUlmGermany
- Department of Internal Medicine IIUniversity Hospital UlmUlmGermany
| | | | - Christian Renz
- Institute of Physiological ChemistryUniversity of UlmUlmGermany
| | | | - Thomas Wirth
- Institute of Physiological ChemistryUniversity of UlmUlmGermany
| | - Thomas Haehner
- Institute of Physiological ChemistryUniversity of UlmUlmGermany
| |
Collapse
|
31
|
Bagherieh M, Kheirollahi A, Zamani-Garmsiri F, Emamgholipour S, Meshkani R. Folic acid ameliorates palmitate-induced inflammation through decreasing homocysteine and inhibiting NF-κB pathway in HepG2 cells. Arch Physiol Biochem 2021:1-8. [PMID: 33596128 DOI: 10.1080/13813455.2021.1878539] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
OBJECTIVE Prevention of inflammation is one of the possible remedy procedure for steatohepatitis during NAFLD. In this study, we researched the folic acid (FA) potency to attenuate the inflammation of palmitate-treated HepG2 cells and the related signalling pathways. METHODS The molecular mechanisms related to FA anti-inflammatory effect in palmitate and Hcy-treated HepG2 cell line were assessed. RESULTS The results indicated that while palmitate enhances the expression and secretion of TNF-α, IL-6, and IL-1β, and also intracellular ROS level, FA at concentrations of 25, 50, and 75 µg/mL significantly reversed these effects in HepG2 cells. In addition, FA could ameliorate inflammation and decrease ROS production induced by Hcy. Furthermore, FA pre-treatment suppress palmitate -induced (NF-κB) p65 level in palmitate or Hcy stimulated cells. CONCLUSIONS Overall, these results suggest that FA reduces inflammation in HepG2 cells through decreasing ROS and Hcy concentration level resulting in inhibiting the NF-κB pathway.
Collapse
Affiliation(s)
- Molood Bagherieh
- Department of Clinical Biochemistry, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Asma Kheirollahi
- Department of Comparative Biosciences, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Fahimeh Zamani-Garmsiri
- Department of Clinical Biochemistry, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Solaleh Emamgholipour
- Department of Clinical Biochemistry, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Reza Meshkani
- Department of Clinical Biochemistry, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
32
|
Evaluating the Efficacy of Carum Copticum Seeds on the Treatment of Patients with Nonalcoholic Fatty Liver Disease: A Multi-Center, Randomized, Triple–Blind, Placebo-Controlled Clinical Trial Study. HEPATITIS MONTHLY 2021. [DOI: 10.5812/hepatmon.110488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/13/2023]
Abstract
Background: The increased prevalence of Nonalcoholic Fatty Liver Disease (NAFLD) has caused several concerns. Besides, there are concerns about the side effects of the drugs used to treat this condition. Since the current treatments are not effective in treating NAFLD, developing a novel therapeutic option based on some medicinal plants is necessary. Few studies have investigated the natural drugs and their effects (e.g., extracts of Carum Copticum Seeds (CCS)) on these patients. Objectives: The current study intended to evaluate the safety and efficacy of CCS extracts on liver enzymes, serum lipids, as well as the grade of fatty liver, and anthropometric measurements in patients with nonalcoholic fatty liver disease. Methods: In this multi-center, randomized, triple-blind, placebo-controlled clinical trial study, 90 patients with grades of 1 to 3 of nonalcoholic fatty liver disease referred to two medical centers located in Tehran (Iran) in 2019 are examined. Diet and exercise were recommended for patients three times a day in eight weeks, in addition to taking 500 mg capsules (aqueous extracts of Ajwain seeds and placebo for treatment and placebo groups). Results: Medical records of 68 patients were reviewed, 48 (71%) male and 20 (29%) female. Both groups were similar concerning demographic and baseline characteristics. The total score of the Leeds questionnaire [-14.03 vs. -7.49, P = 0.048], Triglyceride (TG) [-4.45 vs. 10.03, P = 0.017], and Alanine Aminotransferase (ALT) [-14.71 vs. -4.79, P = 0.012] were significantly changed in the treatment group compared to the control group. Nevertheless, a different situation was observed for Aspartate Aminotransferase (AST) [-7.08 vs. -4.84, P = 0.314], Fasting Blood Sugar (FBS) [-2.88 vs. -2.81, P = 0.207], and Body Mass Index (BMI) [-0.59 vs. -0.39, P = 0.095]. Although a significant change was found in both groups, the amount of decline was similar for both groups. In both groups, the cholesterol, High-Density Lipoprotein (HDL), and Low-Density Lipoprotein (LDL) did not change significantly. The sonographic findings indicated significantly higher improvements in the intervention group than the placebo group (RR = 2.43, 95% CI (1.15-5.65), and P value = 0.034). Conclusions: The result of this study supports the efficacy of Carum copticum seeds in the treatment of nonalcoholic fatty liver disease patients. It was found that CCS with a significant reduction in ALT, TG, and relative reduction of BMI can help physicians to manage other metabolic disorders associated with NAFLD, such as obesity and hyperlipidemia.
Collapse
|
33
|
Kiyak A, Elibol S, Barutcu O, Saruc M, Tozun N. Subtracted Adulthood Mass Index - a new index to predict NAFLD risk in non-obese individuals. HEPATOLOGY FORUM 2021; 2:26-30. [PMID: 35782892 PMCID: PMC9138927 DOI: 10.14744/hf.2020.2020.0031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 12/28/2020] [Indexed: 11/21/2022]
Abstract
Background and Aim Nonalcoholic fatty liver disease (NAFLD) is a common chronic liver disease. The aims of the current study are to determine the relationship between NAFLD in non-obese individuals and weight gain during adulthood and develop a new index for the identification of NAFLD risk. Materials and Methods For this cross-sectional study, 362 patients who underwent abdominal ultrasonography (USG) in our clinic were included. Seventy-eight individuals were obese (>30 kg/m2). A history of weight gain during adulthood and systemic metabolic diseases was collected at the time of the study. A new index termed "Subtracted Adulthood Mass Index" (SAMI) was created to estimate the risk of NAFLD development for non-obese people. SAMI is the ratio of the difference between the individual's current weight and his/her weight at 20 years old to his/her height squared (kg/m2). Results When the SAMI cut-off was set at 3 kg/m2, the sensitivity for predicting NAFLD risk was 85.2%, the specificity was 66.9%, the PPV was 79.1%, and the NPV was 75.4%. Conclusion In this innovational study, a new index named SAMI was developed to identify non-obese people who are at risk of developing NAFLD. The SAMI is easy to calculate and appropriate for clinical use.
Collapse
Affiliation(s)
- Ata Kiyak
- Acibadem University School of Medicine, Istanbul, Turkey
| | - Serra Elibol
- Acibadem University School of Medicine, Istanbul, Turkey
| | - Ozlem Barutcu
- Department of Radiology, Acibadem University School of Medicine; Istanbul, Turkey
| | - Murat Saruc
- Department of Gastroenterology Acibadem University School of Medicine, Istanbul, Turkey
| | - Nurdan Tozun
- Department of Gastroenterology Acibadem University School of Medicine, Istanbul, Turkey
| |
Collapse
|
34
|
Gómez-Zorita S, Milton-Laskíbar I, Aguirre L, Fernández-Quintela A, Xiao J, Portillo MP. Effects of Pterostilbene on Diabetes, Liver Steatosis and Serum Lipids. Curr Med Chem 2021; 28:238-252. [PMID: 31663469 DOI: 10.2174/0929867326666191029112626] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Revised: 10/16/2019] [Accepted: 10/27/2019] [Indexed: 02/08/2023]
Abstract
Pterostilbene, a phenolic compound derived from resveratrol, possesses greater bioavailability than its parent compound due to the presence of two methoxyl groups. In this review, the beneficial effects of pterostilbene on diabetes, liver steatosis and dyslipidemia are summarized. Pterostilbene is a useful bioactive compound in preventing type 1 diabetes, insulin resistance and type 2 diabetes in animal models. Concerning type 1 diabetes, the main mechanisms described to justify the positive effects of this phenolic compound are increased liver glycogen content and hepatic glucokinase and phosphofructokinase activities, the recovery of pancreatic islet architecture, cytoprotection and a decrease in serum and pancreatic pro-inflammatory cytokines. As for type 2 diabetes, increased liver glucokinase and glucose-6-phosphatase and decreased fructose-1,6-biphosphatase activities are reported. When insulin resistance is induced by diets, a greater activation of insulin signaling cascade has been reported, increased cardiotrophin-1 levels and liver glucokinase and glucose- 6-phosphatase activities, and a decreased fructose-1,6-biphosphatase activity. Data concerning pterostilbene and liver steatosis are scarce so far, but the reduction in oxidative stress induced by pterostilbene may be involved since oxidative stress is related to the progression of steatosis to steatohepatitis. Finally, pterostilbene effectively reduces total cholesterol, LDL-cholesterol and serum triglyceride levels, while increases HDL-cholesterol in animal models of dyslipidemia.
Collapse
Affiliation(s)
- Saioa Gómez-Zorita
- Nutrition and Obesity Group, Department of Pharmacy and Food Sciences, University of the Basque Country (UPV/EHU) and Lucio Lascaray Research Institute, Vitoria, Spain
| | - Iñaki Milton-Laskíbar
- Nutrition and Obesity Group, Department of Pharmacy and Food Sciences, University of the Basque Country (UPV/EHU) and Lucio Lascaray Research Institute, Vitoria, Spain
| | - Leixuri Aguirre
- Nutrition and Obesity Group, Department of Pharmacy and Food Sciences, University of the Basque Country (UPV/EHU) and Lucio Lascaray Research Institute, Vitoria, Spain
| | - Alfredo Fernández-Quintela
- Nutrition and Obesity Group, Department of Pharmacy and Food Sciences, University of the Basque Country (UPV/EHU) and Lucio Lascaray Research Institute, Vitoria, Spain
| | - Jianbo Xiao
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Control in Chinese Medicine, University of Macau, Macau SAR, China
| | - María P Portillo
- Nutrition and Obesity Group, Department of Pharmacy and Food Sciences, University of the Basque Country (UPV/EHU) and Lucio Lascaray Research Institute, Vitoria, Spain
| |
Collapse
|
35
|
Chao HC, Lin HY. Comparison of Body Mass Index and Fat Indices in Predicting the Severity of Nonalcoholic Fatty Liver Disease Among Children Who Are Overweight and Obese. Front Pediatr 2021; 9:724426. [PMID: 34513769 PMCID: PMC8432615 DOI: 10.3389/fped.2021.724426] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2021] [Accepted: 07/26/2021] [Indexed: 12/12/2022] Open
Abstract
Background: Information of the relationships between body mass parameters and the severity of fatty liver is deficient in pediatric nonalcoholic fatty liver disease (NAFLD). Methods: The relationships between body mass parameters (waist circumference [WC], body mass index [BMI], and abdominal subcutaneous fat thickness [ASFT]) and the severity of fatty liver were prospectively evaluated in pediatric patients who are overweight or obese, suffering from NAFLD. Ultrasonography was performed to assess fatty liver and its severity on a three-grade scale (low-grade fatty liver [LGFL], grade 1 or 2; high-grade fatty liver [HGFL], grade 3). Results: A total of 110 subjects (55 LGFL and 55 HGFL) aged 6.2-17.9 years were included. The WC, BMI, and ASFT values were significantly higher in the HGFL group compared to those in the LGFL group (p = 0.00004, 0.01, and 0.04, respectively). WC had the greatest power to predict HGFL under receiver-operating characteristic curve analyses and was positively correlated with the severity of fatty liver in subjects aged 6-12-year old and 13-17-year old (p = 0.007, and 0.0039, respectively). ASFT showed a positive correlation with the severity of fatty liver in subjects aged 13-17-year old (p = 0.04). Conclusions: WC, BMI, and ASFT are predictive of severe NAFLD among children who are overweight and obese; particularly, WC has the most predictive accuracy. Among the parameters, WC and ASFT are predictive in specific age groups.
Collapse
Affiliation(s)
- Hsun-Chin Chao
- Division of Pediatric Gastroenterology, Department of Pediatrics, Chang Gung Children's Medical Center, Chang Gung Memorial Hospital, Taoyuan City, Taiwan.,Chang Gung University College of Medicine, Taoyuan City, Taiwan
| | - Hsin-Yeh Lin
- Division of Pediatric Gastroenterology, Department of Pediatrics, Chang Gung Children's Medical Center, Chang Gung Memorial Hospital, Taoyuan City, Taiwan
| |
Collapse
|
36
|
Zain SM, Tan H, Mohamed Z, Chan W, Mahadeva S, Basu RC, Mohamed R. Use of simple scoring systems for a public health approach in the management of non‐alcoholic fatty liver disease patients. JGH Open 2020; 4:1155-1161. [PMID: 33319051 PMCID: PMC7731808 DOI: 10.1002/jgh3.12414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 08/23/2020] [Accepted: 08/25/2020] [Indexed: 11/08/2022]
Abstract
Background and Aim Methods Results Conclusion
Collapse
Affiliation(s)
- Shamsul Mohd Zain
- Department of Pharmacology, Faculty of Medicine University of Malaya Kuala Lumpur Malaysia
| | - Hwa‐Li Tan
- Department of Pharmacology, Faculty of Medicine University of Malaya Kuala Lumpur Malaysia
| | - Zahurin Mohamed
- Department of Pharmacology, Faculty of Medicine University of Malaya Kuala Lumpur Malaysia
| | - Wah‐Kheong Chan
- Department of Medicine, Faculty of Medicine University of Malaya Kuala Lumpur Malaysia
| | - Sanjiv Mahadeva
- Department of Medicine, Faculty of Medicine University of Malaya Kuala Lumpur Malaysia
| | - Roma Choudhury Basu
- Clinical Investigation Centre University Malaya Medical Centre Kuala Lumpur Malaysia
| | - Rosmawati Mohamed
- Department of Medicine, Faculty of Medicine University of Malaya Kuala Lumpur Malaysia
| |
Collapse
|
37
|
Verlinden W, Van Mieghem E, Depauw L, Vanwolleghem T, Vonghia L, Weyler J, Driessen A, Callens D, Roosens L, Dirinck E, Verrijken A, Gaal LV, Francque S. Non-Alcoholic Steatohepatitis Decreases Microsomal Liver Function in the Absence of Fibrosis. Biomedicines 2020; 8:E546. [PMID: 33261113 PMCID: PMC7760673 DOI: 10.3390/biomedicines8120546] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 11/23/2020] [Accepted: 11/26/2020] [Indexed: 12/21/2022] Open
Abstract
The incidence of non-alcoholic fatty liver disease (NAFLD) is rising across the globe, with the presence of steatohepatitis leading to a more aggressive clinical course. Currently, the diagnosis of non-alcoholic steatohepatitis (NASH) is based on histology, though with the high prevalence of NAFLD, a non-invasive method is needed. The 13C-aminopyrine breath test (ABT) evaluates the microsomal liver function and could be a potential candidate. We aimed to evaluate a potential change in liver function in NASH patients and to evaluate the diagnostic power of ABT to detect NASH. We performed a retrospective analysis on patients suspected of NAFLD who underwent a liver biopsy and ABT. 440 patients were included. ABT did not decrease in patients with isolated liver steatosis but decreased significantly in the presence of NASH without fibrosis and decreased even further with the presence of significant fibrosis. The predictive power of ABT as a single test for NASH was low but improved in combination with ALT and ultrasonographic steatosis. We conclude that microsomal liver function of patients with NASH is significantly decreased, even in the absence of fibrosis. The ABT is thus a valuable tool in assessing the presence of NASH; and could be used as a supplementary diagnostic tool in clinical practice.
Collapse
Affiliation(s)
- Wim Verlinden
- Laboratory of Experimental Medicine and Pediatrics, Division of Gastroenterology and Hepatology, University of Antwerp, 2610 Antwerp, Belgium; (E.V.M.); (L.D.); (T.V.); (L.V.); (J.W.)
- Department of Gastroenterology and Hepatology, Antwerp University Hospital, 2650 Antwerp, Belgium
| | - Eugénie Van Mieghem
- Laboratory of Experimental Medicine and Pediatrics, Division of Gastroenterology and Hepatology, University of Antwerp, 2610 Antwerp, Belgium; (E.V.M.); (L.D.); (T.V.); (L.V.); (J.W.)
| | - Laura Depauw
- Laboratory of Experimental Medicine and Pediatrics, Division of Gastroenterology and Hepatology, University of Antwerp, 2610 Antwerp, Belgium; (E.V.M.); (L.D.); (T.V.); (L.V.); (J.W.)
| | - Thomas Vanwolleghem
- Laboratory of Experimental Medicine and Pediatrics, Division of Gastroenterology and Hepatology, University of Antwerp, 2610 Antwerp, Belgium; (E.V.M.); (L.D.); (T.V.); (L.V.); (J.W.)
- Department of Gastroenterology and Hepatology, Antwerp University Hospital, 2650 Antwerp, Belgium
| | - Luisa Vonghia
- Laboratory of Experimental Medicine and Pediatrics, Division of Gastroenterology and Hepatology, University of Antwerp, 2610 Antwerp, Belgium; (E.V.M.); (L.D.); (T.V.); (L.V.); (J.W.)
- Department of Gastroenterology and Hepatology, Antwerp University Hospital, 2650 Antwerp, Belgium
| | - Jonas Weyler
- Laboratory of Experimental Medicine and Pediatrics, Division of Gastroenterology and Hepatology, University of Antwerp, 2610 Antwerp, Belgium; (E.V.M.); (L.D.); (T.V.); (L.V.); (J.W.)
- Department of Gastroenterology and Hepatology, Antwerp University Hospital, 2650 Antwerp, Belgium
| | - Ann Driessen
- Department of Pathology, Antwerp University Hospital, 2650 Antwerp, Belgium;
| | - Dirk Callens
- Department of Clinical Biology, Antwerp University Hospital, 2650 Antwerp, Belgium; (D.C.); (L.R.)
| | - Laurence Roosens
- Department of Clinical Biology, Antwerp University Hospital, 2650 Antwerp, Belgium; (D.C.); (L.R.)
| | - Eveline Dirinck
- Department of Endocrinology, Diabetology and Metabolism, Antwerp University Hospital, 2650 Antwerp, Belgium; (E.D.); (A.V.); (L.V.G.)
| | - An Verrijken
- Department of Endocrinology, Diabetology and Metabolism, Antwerp University Hospital, 2650 Antwerp, Belgium; (E.D.); (A.V.); (L.V.G.)
| | - Luc Van Gaal
- Department of Endocrinology, Diabetology and Metabolism, Antwerp University Hospital, 2650 Antwerp, Belgium; (E.D.); (A.V.); (L.V.G.)
| | - Sven Francque
- Laboratory of Experimental Medicine and Pediatrics, Division of Gastroenterology and Hepatology, University of Antwerp, 2610 Antwerp, Belgium; (E.V.M.); (L.D.); (T.V.); (L.V.); (J.W.)
- Department of Gastroenterology and Hepatology, Antwerp University Hospital, 2650 Antwerp, Belgium
| |
Collapse
|
38
|
Chiu LS, Pedley A, Massaro J, Benjamin EJ, Mitchell GF, McManus DD, Aragam J, Vasan RS, Cheng S, Long MT. The association of non-alcoholic fatty liver disease and cardiac structure and function-Framingham Heart Study. Liver Int 2020; 40:2445-2454. [PMID: 32654390 PMCID: PMC7669676 DOI: 10.1111/liv.14600] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 06/29/2020] [Accepted: 07/01/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND & AIMS Non-alcoholic fatty liver disease confers increased risk for cardiovascular disease, including heart failure (HF), for reasons that remain unclear. Possible pathways could involve an association of liver fat with cardiac structural or functional abnormalities even after accounting for body size. METHODS We analysed N = 2356 Framingham Heart Study participants (age 52 ± 12 years, 52% women) who underwent echocardiography and standardized computed tomography measures of liver fat. RESULTS In cross-sectional multivariable regression models adjusted for age, gender, cohort and cardiovascular risk factors, liver fat was positively associated with left ventricular (LV) mass (β = 1.45; 95% confidence interval (CI): 0.01, 2.88), LV wall thickness (β = 0.01; 95% CI: 0.00, 0.02), mass volume ratio (β = 0.02; 95% CI 0.01, 0.03), mitral peak velocity (E) (β = 0.83; 95% CI 0.31, 1.36) and LV filling pressure (E/e' ratio) (β = 0.16; 95% CI 0.09, 0.23); and inversely associated with global systolic longitudinal strain (β = 0.20, 95% CI 0.07, 0.33), diastolic annular velocity (e') (β = -0.12; 95% CI - 0.22, -0.03), and E/A ratio (β = -0.01; 95% CI - 0.02, -0.00). After additional adjustment for body mass index (BMI), statistical significance was attenuated for all associations except for that of greater liver fat with increased LV filling pressure, a possible precursor to HF (β = 0.11; 95% CI 0.03, 0.18). CONCLUSION Increased liver fat was associated with multiple subclinical cardiac dysfunction measures, with most of associations mediated by obesity. Interestingly, the association of liver fat and LV filling pressure was only partially mediated by BMI, suggesting a possible direct effect of liver fat on LV filling pressure. Further confirmatory studies are needed.
Collapse
Affiliation(s)
- Laura S. Chiu
- Section of Gastroenterology, Boston Medical Center, Boston University School of Medicine, Boston, MA
| | | | - Joseph Massaro
- National Heart, Lung, and Blood Institute’s and Boston University’s Framingham Heart Study, Framingham, MA.,Department of Mathematics and Statistics, Boston University, Boston, MA, United States
| | - Emelia J. Benjamin
- National Heart, Lung, and Blood Institute’s and Boston University’s Framingham Heart Study, Framingham, MA.,Section of Cardiovascular Medicine, Boston Medical Center, Boston University School of Medicine, Boston, MA,Department of Epidemiology, Boston University School of Public Health, Boston, MA
| | | | - David D. McManus
- Cardiology Division, Department of Medicine and the Department of Quantitative Health Sciences, University of Massachusetts Medical School, Worchester, MA
| | - Jayashri Aragam
- Cardiovascular Division, VA Boston Healthcare System, West Roxbury, MA,Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Boston, MA,Harvard Medical School, Boston, MA
| | - Ramachandran S. Vasan
- National Heart, Lung, and Blood Institute’s and Boston University’s Framingham Heart Study, Framingham, MA.,Section of Cardiovascular Medicine, Boston Medical Center, Boston University School of Medicine, Boston, MA,Department of Epidemiology, Boston University School of Public Health, Boston, MA,Section of Preventive Medicine and Epidemiology, Department of Medicine, Boston Medical Center, Boston, MA
| | - Susan Cheng
- Barbra Streisand Women’s Heart Center, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Michelle T. Long
- Section of Gastroenterology, Boston Medical Center, Boston University School of Medicine, Boston, MA
| |
Collapse
|
39
|
Canbay A, Kachru N, Haas JS, Sowa JP, Meise D, Ozbay AB. Patterns and predictors of mortality and disease progression among patients with non-alcoholic fatty liver disease. Aliment Pharmacol Ther 2020; 52:1185-1194. [PMID: 33016540 DOI: 10.1111/apt.16016] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 05/05/2020] [Accepted: 07/15/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND Factors associated with mortality and disease progression in non-alcoholic fatty liver disease (NAFLD) and non-alcoholic steatohepatitis (NASH) are poorly understood. AIMS To assess the impact of liver disease severity, demographics and comorbidities on all-cause mortality and liver disease progression in a large, real-world cohort of NAFLD patients. METHODS Claims data from the German Institut für angewandte Gesundheitsforschung database between 2011 and 2016 were analyzed retrospectively. Adult patients diagnosed with NAFLD and/or NASH were categorised as NAFLD, NAFLD non-progressors, compensated cirrhosis, decompensated cirrhosis, liver transplant or hepatocellular carcinoma (HCC). The longitudinal probability of mortality and incidence of progression were calculated for disease severity cohorts and multivariable analyses performed for adjusted mortality. RESULTS Among 4 580 434 patients in the database, prevalence of NAFLD was 4.7% (n = 215 655). Of those, 36.8% were non-progressors, 0.2% compensated cirrhosis, 9.6% decompensated cirrhosis, 0.0005% liver transplant and 0.2% HCC. Comorbidity rates were significantly higher in compensated cirrhosis, decompensated cirrhosis and HCC compared with non-progressors. The longitudinal probability of mortality for non-progressors, compensated cirrhosis, decompensated cirrhosis and HCC was 3.6%, 18.7%, 28.8% and 68%, respectively. Independent predictors of mortality included cardiovascular disease, type 2 diabetes mellitus, hypertension, obesity and renal impairment. The cumulative incidence of progression in NAFLD and compensated cirrhosis patients was 10.7% and 16.7%, respectively, over 5 years of follow-up. CONCLUSION NAFLD patients were severely under-diagnosed and had a high probability of mortality that increased with disease progression. Early identification and effective management to halt or reverse fibrosis are essential to prevent progression.
Collapse
Affiliation(s)
- Ali Canbay
- Department of Internal Medicine, Ruhr-University, Bochum, Germany
| | - Nandita Kachru
- Health Economics Outcomes Research, Gilead Sciences Inc., Foster City, CA, USA
| | | | - Jan-Peter Sowa
- Department of Internal Medicine, Ruhr-University, Bochum, Germany
| | | | - Ahmet Burak Ozbay
- Health Economics Outcomes Research, Gilead Sciences Inc., Foster City, CA, USA
| |
Collapse
|
40
|
Shi Y, Lam SM, Liu H, Luo G, Zhang J, Yao S, Li J, Zheng L, Xu N, Zhang X, Shui G. Comprehensive lipidomics in apoM -/- mice reveals an overall state of metabolic distress and attenuated hepatic lipid secretion into the circulation. J Genet Genomics 2020; 47:523-534. [PMID: 33309167 DOI: 10.1016/j.jgg.2020.08.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 08/03/2020] [Accepted: 08/05/2020] [Indexed: 12/14/2022]
Abstract
Apolipoprotein M (apoM) participates in both high-density lipoprotein and cholesterol metabolism. Little is known about how apoM affects lipid composition of the liver and serum. In this study, we systemically investigated the effects of apoM on liver and plasma lipidomes and how apoM participates in lipid cycling, via apoM knockout in mice and the human SMMC-7721 cell line. We used integrated mass spectrometry-based lipidomics approaches to semiquantify more than 600 lipid species from various lipid classes, which include free fatty acids, glycerolipids, phospholipids, sphingolipids, glycosphingolipids, cholesterol, and cholesteryl esters (CEs), in apoM-/- mouse. Hepatic accumulation of neutral lipids, including CEs, triacylglycerols, and diacylglycerols, was observed in apoM-/- mice; while serum lipidomic analyses showed that, in contrast to the liver, the overall levels of CEs and saturated/monounsaturated fatty acids were markedly diminished. Furthermore, the level of ApoB-100 was dramatically increased in the liver, whereas significant reductions in both ApoB-100 and low-density lipoprotein (LDL) cholesterol were observed in the serum of apoM-/- mice, which indicated attenuated hepatic LDL secretion into the circulation. Lipid profiles and proinflammatory cytokine levels indicated that apoM-/- leads to hepatic steatosis and an overall state of metabolic distress. Taken together, these results revealed that apoM knockout leads to hepatic steatosis, impaired lipid secretion, and an overall state of metabolic distress.
Collapse
Affiliation(s)
- Yuanping Shi
- Department of Comprehensive Laboratory, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, China
| | - Sin Man Lam
- Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Hong Liu
- Department of Cardiothoracic Surgery, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, China
| | - Guanghua Luo
- Department of Comprehensive Laboratory, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, China
| | - Jun Zhang
- Department of Comprehensive Laboratory, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, China
| | - Shuang Yao
- Department of Comprehensive Laboratory, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, China
| | - Jie Li
- Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Lu Zheng
- Department of Comprehensive Laboratory, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, China
| | - Ning Xu
- Section of Clinical Chemistry and Pharmacology, Institute of Laboratory Medicine, Lunds University, Klinikgatan 19, S-22185, Lund, Sweden
| | - Xiaoying Zhang
- Department of Cardiothoracic Surgery, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, China.
| | - Guanghou Shui
- Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China.
| |
Collapse
|
41
|
Fernández-Ramos D, Lopitz-Otsoa F, Delacruz-Villar L, Bilbao J, Pagano M, Mosca L, Bizkarguenaga M, Serrano-Macia M, Azkargorta M, Iruarrizaga-Lejarreta M, Sot J, Tsvirkun D, van Liempd SM, Goni FM, Alonso C, Martínez-Chantar ML, Elortza F, Hayardeny L, Lu SC, Mato JM. Arachidyl amido cholanoic acid improves liver glucose and lipid homeostasis in nonalcoholic steatohepatitis via AMPK and mTOR regulation. World J Gastroenterol 2020; 26:5101-5117. [PMID: 32982112 PMCID: PMC7495035 DOI: 10.3748/wjg.v26.i34.5101] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 06/19/2020] [Accepted: 08/13/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Arachidyl amido cholanoic acid (Aramchol) is a potent downregulator of hepatic stearoyl-CoA desaturase 1 (SCD1) protein expression that reduces liver triglycerides and fibrosis in animal models of steatohepatitis. In a phase IIb clinical trial in patients with nonalcoholic steatohepatitis (NASH), 52 wk of treatment with Aramchol reduced blood levels of glycated hemoglobin A1c, an indicator of glycemic control. AIM To assess lipid and glucose metabolism in mouse hepatocytes and in a NASH mouse model [induced with a 0.1% methionine and choline deficient diet (0.1MCD)] after treatment with Aramchol. METHODS Isolated primary mouse hepatocytes were incubated with 20 μmol/L Aramchol or vehicle for 48 h. Subsequently, analyses were performed including Western blot, proteomics by mass spectrometry, and fluxomic analysis with 13C-uniformly labeled glucose. For the in vivo part of the study, male C57BL/6J mice were randomly fed a control or 0.1MCD for 4 wk and received 1 or 5 mg/kg/d Aramchol or vehicle by intragastric gavage for the last 2 wk. Liver metabolomics were assessed using ultra-high-performance liquid chromatography-time of flight-MS for the determination of glucose metabolism-related metabolites. RESULTS Combination of proteomics and Western blot analyses showed increased AMPK activity while the activity of nutrient sensor mTORC1 was decreased by Aramchol in hepatocytes. This translated into changes in the content of their downstream targets including proteins involved in fatty acid (FA) synthesis and oxidation [P-ACCα/β(S79), SCD1, CPT1A/B, HADHA, and HADHB], oxidative phosphorylation (NDUFA9, NDUFB11, NDUFS1, NDUFV1, ETFDH, and UQCRC2), tricarboxylic acid (TCA) cycle (MDH2, SUCLA2, and SUCLG2), and ribosome (P-p70S6K[T389] and P-S6[S235/S236]). Flux experiments with 13C-uniformely labeled glucose showed that TCA cycle cataplerosis was reduced by Aramchol in hepatocytes, as indicated by the increase in the number of rounds that malate remained in the TCA cycle. Finally, liver metabolomic analysis showed that glucose homeostasis was improved by Aramchol in 0.1MCD fed mice in a dose-dependent manner, showing normalization of glucose, G6P, F6P, UDP-glucose, and Rbl5P/Xyl5P. CONCLUSION Aramchol exerts its effect on glucose and lipid metabolism in NASH through activation of AMPK and inhibition of mTORC1, which in turn activate FA β-oxidation and oxidative phosphorylation.
Collapse
Affiliation(s)
- David Fernández-Ramos
- Precision Medicine and Metabolism Laboratory, Centro de Investigación Cooperativa en Biociencias (CIC bioGUNE), Derio 48160, Bizkaia, Spain
- CIBERehd - Centro de Investigación Biomédica en Red de enfermedades hepáticas y digestivas, Madrid 28029, Spain
| | - Fernando Lopitz-Otsoa
- Precision Medicine and Metabolism Laboratory, Centro de Investigación Cooperativa en Biociencias (CIC bioGUNE), Derio 48160, Bizkaia, Spain
| | - Laura Delacruz-Villar
- Precision Medicine and Metabolism Laboratory, Centro de Investigación Cooperativa en Biociencias (CIC bioGUNE), Derio 48160, Bizkaia, Spain
| | - Jon Bilbao
- Precision Medicine and Metabolism Laboratory, Centro de Investigación Cooperativa en Biociencias (CIC bioGUNE), Derio 48160, Bizkaia, Spain
| | - Martina Pagano
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Naples 80138, Italy
| | - Laura Mosca
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Naples 80138, Italy
| | - Maider Bizkarguenaga
- Precision Medicine and Metabolism Laboratory, Centro de Investigación Cooperativa en Biociencias (CIC bioGUNE), Derio 48160, Bizkaia, Spain
| | - Marina Serrano-Macia
- Liver Disease Laboratory, Centro de Investigación Cooperativa en Biociencias (CIC bioGUNE), Derio 48160, Spain
| | - Mikel Azkargorta
- Proteomics Platform, Centro de Investigación Cooperativa en Biociencias (CIC bioGUNE), Derio 48160, Spain
| | | | - Jesús Sot
- Instituto Biofisika (UPV/EHU, CSIC), Leioa 48940, Spain; Departamento de Bioquímica y Biología Molecular, Universidad del País Vasco, Leioa 48940, Spain
| | - Darya Tsvirkun
- Pre-clinical and Chemistry, Manufacturing and Controls, Galmed Pharmaceuticals, Tel Aviv 6578317, Israel
| | | | - Felix M Goni
- Instituto Biofisika (UPV/EHU, CSIC), Leioa 48940, Spain; Departamento de Bioquímica y Biología Molecular, Universidad del País Vasco, Leioa 48940, Spain
| | | | - María Luz Martínez-Chantar
- CIBERehd - Centro de Investigación Biomédica en Red de enfermedades hepáticas y digestivas, Madrid 28029, Spain
- Liver Disease Laboratory, Centro de Investigación Cooperativa en Biociencias (CIC bioGUNE), Derio 48160, Spain
| | - Felix Elortza
- Proteomics Platform, Centro de Investigación Cooperativa en Biociencias (CIC bioGUNE), Derio 48160, Spain
| | - Liat Hayardeny
- Pre-clinical and Chemistry, Manufacturing and Controls, Galmed Pharmaceuticals, Tel Aviv 6578317, Israel
| | - Shelly C Lu
- Division of Digestive and Liver Diseases, Cedars-Sinai Medical Center, Los Angeles, CA 90048, United States
| | - José M Mato
- Precision Medicine and Metabolism Laboratory, Centro de Investigación Cooperativa en Biociencias (CIC bioGUNE), Derio 48160, Bizkaia, Spain
- CIBERehd - Centro de Investigación Biomédica en Red de enfermedades hepáticas y digestivas, Madrid 28029, Spain
| |
Collapse
|
42
|
Abstract
There are >1.5 billion people with chronic liver disease worldwide, causing liver diseases to be a significant global health issue. Diffuse parenchymal liver diseases, including hepatic steatosis, fibrosis, metabolic diseases, and hepatitis cause chronic liver injury and may progress to fibrosis and eventually hepatocellular carcinoma. As early diagnosis and treatment of these diseases impact the progression and outcome, the need for assessment of the liver parenchyma has increased. While the current gold standard for evaluation of the hepatic parenchymal tissue, biopsy has disadvantages and limitations. Consequently, noninvasive methods have been developed based on serum biomarkers and imaging techniques. Conventional imaging modalities such as ultrasound, computed tomography scan, and magnetic resonance imaging provide noninvasive options for assessment of liver tissue. However, several recent advances in liver imaging techniques have been introduced. This review article focuses on the current status of imaging methods for diffuse parenchymal liver diseases assessment including their diagnostic accuracy, advantages and disadvantages, and comparison between different techniques.
Collapse
|
43
|
Nonalcoholic Fatty Liver Disease (NAFLD) and Hepatic Cytochrome P450 (CYP) Enzymes. Pharmaceuticals (Basel) 2020; 13:ph13090222. [PMID: 32872474 PMCID: PMC7560175 DOI: 10.3390/ph13090222] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 08/21/2020] [Indexed: 12/12/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is characterized by excessive fat in the liver. An international consensus panel has recently proposed to rename the disease to metabolic dysfunction associated with fatty liver disease (MAFLD). The disease can range from simple steatosis (fat accumulation) to nonalcoholic steatohepatitis (NASH) which represents a severe form of NAFLD and is accompanied by inflammation, fibrosis, and hepatocyte damage in addition to significant steatosis. This review collates current knowledge of changes in human hepatic cytochrome P450 enzymes in NAFLD. While the expression of these enzymes is well studied in healthy volunteers, our understanding of the alterations of these proteins in NAFLD is limited. Much of the existing knowledge on the subject is derived from preclinical studies, and clinical translation of these findings is poor. Wherever available, the effect of NAFLD on these proteins in humans is debatable and currently lacks a consensus among different reports. Protein expression is an important in vitro physiological parameter controlling the pharmacokinetics of drugs and the last decade has seen a rise in the accurate estimation of these proteins for use with physiologically based pharmacokinetic (PBPK) modeling to predict drug pharmacokinetics in special populations. The application of label-free, mass spectrometry-based quantitative proteomics as a promising tool to study NAFLD-associated changes has also been discussed.
Collapse
|
44
|
Kalaki-Jouybari F, Shanaki M, Delfan M, Gorgani-Firouzjae S, Khakdan S. High-intensity interval training (HIIT) alleviated NAFLD feature via miR-122 induction in liver of high-fat high-fructose diet induced diabetic rats. Arch Physiol Biochem 2020; 126:242-249. [PMID: 30318957 DOI: 10.1080/13813455.2018.1510968] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Background: Exercise intervention is strongly recommended to manage metabolic diseases. In this study, we investigate, whether HIIT and CET can induce hepatic miR-122 expression, NAFLD rats with diabetes.Methods: 40 Wistar rats divided into 2 groups, non-diabetic (NDC) and diabetic .Type 2 diabetes was induced by high-fat high-fructose diet (HFHFD). Then diabetic rats were subdivided into three groups: diabetic control (HFHFD + DC), CET (HFHFD + CET), and HIIT (HFHFD + HIIT). After eight weeks of exercise on a rodent treadmill, we measured miR-122 and its target genes expression in the liver of rats.Results: HIIT decreased the expression of FAS, ACC, SREBP-1c compared with HFHFD + DC (p = .004, p = .032, p = .043, respectively), and could partially increase miR-122 expression as compared with HFHFD + DC (26.8%, p = .68).Conclusions: Exercise training could be a non-pharmacological intervention for improvement of NAFLD of diabetic rats by induction of miR-122. HIIT had a greater effect on NAFLD amelioration than CET.
Collapse
Affiliation(s)
- Fatemeh Kalaki-Jouybari
- Department of Medical Laboratory Sciences, School of Allied Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mehrnoosh Shanaki
- Department of Medical Laboratory Sciences, School of Allied Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Maryam Delfan
- Department of Exercise Physiology, Faculty of Physical Education and Sport Sciences, Alzahra University, Tehran, Iran
| | - Sattar Gorgani-Firouzjae
- Department of Medical Laboratory Sciences, School of Allied Health Medicine, AJA University of Medical Sciences, Tehran, Iran
| | - Soheyla Khakdan
- Department of Medical Laboratory Sciences, School of Allied Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
45
|
Loomba R, Wong R, Fraysse J, Shreay S, Li S, Harrison S, Gordon SC. Nonalcoholic fatty liver disease progression rates to cirrhosis and progression of cirrhosis to decompensation and mortality: a real world analysis of Medicare data. Aliment Pharmacol Ther 2020; 51:1149-1159. [PMID: 32372515 DOI: 10.1111/apt.15679] [Citation(s) in RCA: 130] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 01/17/2020] [Accepted: 02/12/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND Risk factors and timing associated with disease progression and mortality in nonalcoholic fatty liver disease (NAFLD) are poorly understood. AIMS To evaluate the impact of disease severity, demographics and comorbidities on risk of mortality and time to progression in a large, real-world cohort of diagnosed NAFLD patients. METHODS Claims data from a 20% Medicare representative sample between 2007 and 2015 were analysed retrospectively. Adults were categorised into disease severity groups: NAFLD/nonalcoholic steatohepatitis (NASH) alone, compensated cirrhosis, decompensated cirrhosis, liver transplant or hepatocellular carcinoma. Cumulative incidence of mortality and disease progression were calculated for each group and multivariate analyses performed adjusting for demographics, comorbidities and disease severity. RESULTS A total of 10 826 456, patients were assessed and the prevalence of NAFLD was 5.7% (N = 621 253). Among patients with NAFLD, 71.1% had NAFLD/NASH alone and 28.9% had NAFLD cirrhosis. Overall, 85.5% of patients had hypertension, 84.1% dyslipidemia, 68.7% had cardiovascular disease and 55.5% diabetes. The cumulative risk of progression of NAFLD to cirrhosis, and compensated cirrhosis to decompensated cirrhosis was 39% and 45%, respectively, over 8 years of follow-up. The independent predictors of progression included cardiovascular disease, renal impairment, dyslipidemia and diabetes. The cumulative risk of mortality for NAFLD, NAFLD cirrhosis, decompensated cirrhosis and hepatocellular carcinoma was 12.6%, 31.1%, 51.4% and 76.2%, respectively. CONCLUSIONS The present report (a) demonstrates that NAFLD is grossly underdiagnosed in real-world clinical settings and (b) provides new evidence on the progression rates of NAFLD and risk factors of mortality across the spectrum of severity of NAFLD and cirrhosis.
Collapse
Affiliation(s)
- Rohit Loomba
- Department of Medicine, NAFLD Research Center, La Jolla, CA, USA.,Division of Gastroenterology, Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Robert Wong
- Division of Gastroenterology and Hepatology, Alameda Health System, Highland Hospital, Oakland, CA, USA
| | - Jeremy Fraysse
- Health Economics Outcomes Research, Gilead Sciences, Foster City, CA, USA
| | - Sanatan Shreay
- Health Economics Outcomes Research, Gilead Sciences, Foster City, CA, USA
| | - Suying Li
- Chronic Disease Research Group, Minneapolis, MN, USA
| | | | - Stuart C Gordon
- Department of Gastroenterology and Hepatology, Henry Ford Hospital, Wayne State University School of Medicine, Detroit, MI, USA
| |
Collapse
|
46
|
Tsai YS, Lin SW, Chen YL, Chen CC. Effect of probiotics Lactobacillus paracasei GKS6, L. plantarum GKM3, and L. rhamnosus GKLC1 on alleviating alcohol-induced alcoholic liver disease in a mouse model. Nutr Res Pract 2020; 14:299-308. [PMID: 32765811 PMCID: PMC7390740 DOI: 10.4162/nrp.2020.14.4.299] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 01/09/2020] [Accepted: 04/14/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND/OBJECTIVES Heavy alcohol consumption causes the development of alcoholic liver disease (ALD), a neglected but important public health problem. Many studies have pointed out that probiotics could improve gut health, which is also considered to be a cause of ALD. Therefore, this study screened the probiotics, Lactobacillus casei GKC1 (GKC1), L. fermentum GKF3 (GKF3), Bifidobacterium lactis GKK2 (GKK2), L. rhamnosus GKLC1 (GKLC1), L. paracasei GKS6 (GKS6), and L. plantarum GKM3 (GKM3), for their potential benefits in alleviating ALD for applications to disease prevention. SUBJECTS/METHODS C57BL/6N mice were divided into 8 groups (n = 6 in each): normal control, positive control (alcohol-diet fed), and treatments of feeding probiotics GKC1, GKF3, GKK2, GKLC1, GKS6, and GKM3 under an oral dose 0.82 g/kg B.W. per day by oral gavage. The experiment was conducted for 8 weeks, and the concentrations of alanine aminotransferase (ALT), aspartate aminotransferase, triglyceride (TG), and total cholesterol (TC) in mice were measured. The glutathione (GSH), catalase (CAT), and histology were analyzed after sacrifice. RESULTS The results showed a decrease in the serum ALT, liver TG, and liver TC levels in the GKS6, GKM3, and GKLC1 groups compared to the positive control. In addition, the decreasing GSH and CAT levels were inhibited in the GKS6 and GKM3 groups. The histopathological results showed that all probiotics could reduce the accumulation of liver fat. Furthermore, there was a significant difference in GKLC1 with lower stomach damage compared to the alcohol-fed mice without any addition of probiotics. CONCLUSIONS GKLC1, GKS6, and GKM3 can be used as supplements for alleviating the development of ALD.
Collapse
Affiliation(s)
- You-Shan Tsai
- Biotech Research Institute, Grape King Bio Ltd, Taoyuan 32542, Taiwan
| | - Shih-Wei Lin
- Biotech Research Institute, Grape King Bio Ltd, Taoyuan 32542, Taiwan
| | - Yen-Lien Chen
- Biotech Research Institute, Grape King Bio Ltd, Taoyuan 32542, Taiwan
| | - Chin-Chu Chen
- Institute of Food Science and Technology, National Taiwan University, Taipei 10617, Taiwan.,Department of Food Science, Nutrition and Nutraceutical Biotechnology, Shih Chien University, Taipei 10462, Taiwan.,Department of Bioscience Technology, Chung Yuan Christian University, Taoyuan 32023, Taiwan
| |
Collapse
|
47
|
Lüchtenborg C, Niederhaus B, Brügger B, Popovic B, Fricker G. Lipid Profiles of Five Essential Phospholipid Preparations for the Treatment of Nonalcoholic Fatty Liver Disease: A Comparative Study. Lipids 2020; 55:271-278. [PMID: 32255515 DOI: 10.1002/lipd.12236] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 02/26/2020] [Accepted: 03/09/2020] [Indexed: 11/10/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is associated with an imbalance in fatty acid composition and can progress from simple steatosis to steatohepatitis, liver cirrhosis, and hepatocellular carcinoma. Essential phospholipids (EPL), which contain high levels of 1,2-dilinoleoylphosphatidylcholine, can be used to treat NAFLD. Polyenylphosphatidylcholine (PPC) preparations are external, commercially available EPL products. The lipid composition of five commercially available PPC preparations, including Essentiale Forte, Fortifikat, Hepatoprotect Regenerator, Fortifikat Forte, and Esentin Forte were compared, the outcome of which may impact physician choice in the treatment of NAFLD. Following lipid extraction, a comparative analysis of key lipid content was performed using a QTRAP6500+ triple quadruple ion trap hybrid mass spectrometer (Sciex) in nanoelectrospray ionization mode. The glycerophospholipid composition of each PPC was determined, including levels of phosphatidylcholine (PtdCho), and phosphatidylethanolamine (PtdEtn) species, as well as PtdCho:PtdEtn ratio. Of the five preparations analyzed, Essentiale Forte contained the highest PtdCho levels (61.9 mol%) and lowest PtdEtn levels (4.9 mol%). PtdCho 36:4 levels, a polyunsaturated species of PtdCho, were highest in Esentin Forte (39.3 mol%) and Essentiale Forte (38.3 mol%) compared with other PPCs (28.7-35.8 mol%). Levels of lysophosphatidylcholine, phosphatidylinositol, phosphatidic acid, and phosphatidylglycerol were low in all five preparations. Lipid composition was consistent between the preparations. The high PtdCho:PtdEtn ratio composition of Essentiale Forte compared with the other PPC analyzed, as well as the presence of polyunsaturated fatty acids, suggest it could be the most clinically beneficial commercially available hepatoprotective product in the treatment of NAFLD.
Collapse
Affiliation(s)
| | | | - Britta Brügger
- Heidelberg University Biochemistry Center, Heidelberg, 69120, Germany
| | - Branko Popovic
- Sanofi-Aventis Deutschland GmbH, Frankfurt, 65929, Germany
| | - Gert Fricker
- Institute of Pharmacy and Molecular Biotechnology, Ruprecht-Karls University, Heidelberg, 69120, Germany
| |
Collapse
|
48
|
Wang W, Xu AL, Li ZC, Li Y, Xu SF, Sang HC, Zhi F. Combination of Probiotics and Salvia miltiorrhiza Polysaccharide Alleviates Hepatic Steatosis via Gut Microbiota Modulation and Insulin Resistance Improvement in High Fat-Induced NAFLD Mice. Diabetes Metab J 2020; 44:336-348. [PMID: 31950772 PMCID: PMC7188963 DOI: 10.4093/dmj.2019.0042] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Accepted: 05/09/2019] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Nonalcoholic fatty liver disease (NAFLD) increases the risk of hepatocellular carcinoma, which is currently the leading cause of obesity-related cancer deaths in middle-aged men. METHODS Probiotics with lipid-lowering function were screened from the fecal microbiota of healthy adults. Polysaccharide from different sources was screened for improving insulin resistance. The combination of probiotics and Salvia miltiorrhiza polysaccharide (LBM) was investigated for alleviating hepatic steatosis. RESULTS First, Bifidobacterium bifidum V (BbV) and Lactobacillus plantarum X (LpX) were obtained from the fecal microbiota of healthy adults. Second, to improve insulin resistance, a Salvia miltiorrhiza Bunge polysaccharide showing good performance in reducing insulin resistance was obtained. The liver total cholesterol (TC) and total triglyceride (TG) levels and the serum levels of free fatty acid, alanine transaminase, aspartate transaminase, low density lipoprotein cholesterol, TG, and TC can be significantly reduced through supplementation with LpX-BbV (LB) in NAFLD mice. Interestingly, the function of the probiotic LB can be enhanced by S. miltiorrhiza Bunge polysaccharide. Furthermore, the gut microbiota was modulated by LpX-BbV+S. miltiorrhiza Bunge polysaccharide (LBM). The lipopolysaccharide concentration of the LBM group was decreased by 73.6% compared to the NAFLD group. Ultimately, the mRNA concentrations of the proinflammatory cytokines (tumor necrosis factor α, interleukin 1β [IL-1β], and IL-6) decreased with LB and LBM treatment. CONCLUSION The results of this this study indicate that the LBM combination can be used as a therapeutic for ameliorating NAFLD via modulating the gut microbiota and improving insulin resistance.
Collapse
Affiliation(s)
- Wei Wang
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Institute of Gastroenterology of Guangdong Province, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Department of Gastroenterology, Sir Run Run Hospital, Nanjing Medical University, Nanjing, China
| | - Ai Lei Xu
- Department of Gastroenterology, Sir Run Run Hospital, Nanjing Medical University, Nanjing, China
| | - Zheng Chao Li
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Institute of Gastroenterology of Guangdong Province, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yi Li
- Department of Gastroenterology, Sir Run Run Hospital, Nanjing Medical University, Nanjing, China
| | - Shun Fu Xu
- Department of Gastroenterology, Sir Run Run Hospital, Nanjing Medical University, Nanjing, China
| | - Hua Chao Sang
- Department of Gastroenterology, Sir Run Run Hospital, Nanjing Medical University, Nanjing, China
| | - Fachao Zhi
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Institute of Gastroenterology of Guangdong Province, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
49
|
Chang Y, Noh JW, Cheon JY, Kim Y, Kwon YD, Ryu S. Self-rated health and risk of incident non-alcoholic fatty liver disease: A cohort study. Sci Rep 2020; 10:3826. [PMID: 32123241 PMCID: PMC7052149 DOI: 10.1038/s41598-020-60823-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Accepted: 02/18/2020] [Indexed: 11/28/2022] Open
Abstract
Although self-rated health (SRH), a subjective measure of overall health status, associates with metabolic abnormalities, studies on the relationship between SRH and non-alcoholic fatty liver disease (NAFLD), a hepatic manifestation of metabolic syndrome, are limited. In this study, we evaluated whether or not SRH predicts the risk of incident NAFLD. This cohort study was performed in a sample of 148,313 Korean adults free of ultrasound-diagnosed NAFLD at baseline with annual or biennial follow-up for a median of 3.7 years. SRH and NAFLD were measured at baseline and follow-up visits. NAFLD was determined based on the ultrasound-diagnosed fatty liver without excessive alcohol consumption or any other cause. Hazard ratios with 95% confidence intervals were estimated via a parametric proportional hazards model. During 522,696.1 person-years of follow-up, 23,855 individuals with new-onset NAFLD were identified (incidence rate, 45.6 per 1,000 person-years). After adjustments for possible confounders including total calorie intake, sleep duration, and depressive symptoms, the multivariate-adjusted hazard ratios (95% confidence intervals) for incident NAFLD comparing good, fair, and poor or very poor SRH to very good SRH were 1.06 (0.97-1.14), 1.18 (1.09-1.27), and 1.24 (1.13-1.37), respectively. This association of SRH with incident NAFLD remained significant after accounting for changes in SRH and confounders during follow-up and was similar across clinically relevant subgroups. In a large-scale cohort study of apparently healthy Korean adults, poor SRH was independently and positively associated with incident NAFLD risk, indicating a predictive role of SRH as a health measure in NAFLD.
Collapse
Affiliation(s)
- Yoosoo Chang
- Department of Occupational and Environmental Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- Center for Cohort Studies, Total Healthcare Center, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine Seoul, Seoul, Republic of Korea
- Department of Clinical Research Design & Evaluation, SAIHST, Sungkyunkwan University, Seoul, Republic of Korea
| | - Jin-Won Noh
- Department of Health Administration, College of Health Science, Dankook University, Cheonan, Republic of Korea
- Institute of Health Promotion and Policy, Dankook University, Cheonan, Republic of Korea
- Global Health Unit, Department of Health Sciences, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Joo Young Cheon
- Department of Nursing Science, Sungshin University, Seoul, Republic of Korea
| | - Yejin Kim
- Center for Cohort Studies, Total Healthcare Center, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine Seoul, Seoul, Republic of Korea
| | - Young Dae Kwon
- Department of Humanities and Social Medicine, College of Medicine and Catholic Institute for Healthcare Management, The Catholic University of Korea, Seoul, Republic of Korea
| | - Seungho Ryu
- Department of Occupational and Environmental Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea.
- Center for Cohort Studies, Total Healthcare Center, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine Seoul, Seoul, Republic of Korea.
- Department of Clinical Research Design & Evaluation, SAIHST, Sungkyunkwan University, Seoul, Republic of Korea.
| |
Collapse
|
50
|
Heeba GH, El-Deen RM, Abdel-Latif RG, Khalifa MMA. Combined treatments with metformin and phosphodiesterase inhibitors alleviate nonalcoholic fatty liver disease in high-fat diet fed rats: a comparative study. Can J Physiol Pharmacol 2020; 98:498-505. [PMID: 32083947 DOI: 10.1139/cjpp-2019-0487] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is an excessive accumulation of fats in the liver resulting in hepatic inflammation and fibrous tissue formation along with insulin resistance. This study was designed to investigate the possible protective effects of metformin alone and in combination with different phosphodiesterase inhibitors (PDEIs). Rats were fed a high-fat diet (HFD) for 16 weeks to induce NAFLD. Starting from week 12, rats received metformin alone or in combination with pentoxifylline, cilostazol, or sildenafil. HFD administration resulted in hepatic steatosis and inflammation in rats. In addition, liver index, body composition index, activities of liver enzymes, and serum lipids deviated from normal. Further, significant elevations were recorded compared to control in terms of serum glucose, insulin, and HOMA-IR (homeostasis model assessment index for insulin resistance), oxidative stress parameters, hepatic TNF-α and NF-κB gene expression, and iNOS protein expression. Rats treated with metformin showed a significant improvement in the aforementioned parameters. However, the addition of pentoxifylline to metformin treatment synergized its action and produced a fortified effect against HFD-induced NAFLD better than other PDEIs. Data from this study indicated that combined treatment of metformin and pentoxifylline had the most remarkable ameliorated effects against HFD-induced NAFLD; further clinical investigations are needed to approve PDEIs for NAFLD treatment.
Collapse
Affiliation(s)
- Gehan H Heeba
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Minia University, El-Minia 61111, Egypt
| | - Reham M El-Deen
- Ministry of Health and Population, Undersecretary preventive sector, General administration viral hepatitis, Cairo, Egypt
| | - Rania G Abdel-Latif
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Minia University, El-Minia 61111, Egypt
| | - Mohamed M A Khalifa
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Minia University, El-Minia 61111, Egypt
| |
Collapse
|