1
|
Michalopoulou E, Thymis J, Lampsas S, Pavlidis G, Katogiannis K, Vlachomitros D, Katsanaki E, Kostelli G, Pililis S, Pliouta L, Kountouri A, Papanikolaou IS, Lambadiari V, Ikonomidis I. The Triad of Risk: Linking MASLD, Cardiovascular Disease and Type 2 Diabetes; From Pathophysiology to Treatment. J Clin Med 2025; 14:428. [PMID: 39860434 PMCID: PMC11765821 DOI: 10.3390/jcm14020428] [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: 11/29/2024] [Revised: 12/30/2024] [Accepted: 01/08/2025] [Indexed: 01/27/2025] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is an emerging global health concern, and it is not only the keystone precursor of eventual liver-related morbidity, but it also places patients at considerably higher cardiovascular risk, which is still a leading cause of death in these patients. The most important common underlying pathophysiological mechanisms in these diseases are primarily related to insulin resistance, chronic inflammation and oxidative stress. The presence of MASLD with cardiovascular disease (CVD) and type 2 diabetes mellitus (T2DM) elevates the risk for poor outcomes, thus this review highlights a method to the therapeutic approaches. Given the intertwined nature of MASLD, T2DM, and CVD, there is an urgent need for therapeutic strategies that address all three conditions. Although lifestyle changes are important as treatment, medication plays a crucial role in managing hyperglycemia, enhancing liver function and lowering cardiovascular risk. The onset and progression of MASLD should be addressed through a multifaceted therapeutic approach, targeting inflammatory, immune, metabolic, oxidative stress, hormonal and gutaxis pathways, alongside the treatment strategies for T2DM. In this review, we discuss the effects of antidiabetic drugs with an impact on both liver outcomes and cardiovascular risk in patients affected by MASLD, T2DM and CDV.
Collapse
Affiliation(s)
- Eleni Michalopoulou
- 2nd Cardiology Department, Attikon University Hospital, National and Kapodistrian University of Athens, Rimini 1, Chaidari, 12462 Athens, Greece; (E.M.); (J.T.); (G.P.); (K.K.); (D.V.); (E.K.); (G.K.)
| | - John Thymis
- 2nd Cardiology Department, Attikon University Hospital, National and Kapodistrian University of Athens, Rimini 1, Chaidari, 12462 Athens, Greece; (E.M.); (J.T.); (G.P.); (K.K.); (D.V.); (E.K.); (G.K.)
| | - Stamatios Lampsas
- Diabetes Center, 2nd Department of Internal Medicine, Attikon University Hospital, Medical School, National and Kapodistrian University of Athens, Rimini 1, Chaidari, 12462 Athens, Greece; (S.L.); (S.P.); (L.P.); (A.K.); (V.L.)
| | - George Pavlidis
- 2nd Cardiology Department, Attikon University Hospital, National and Kapodistrian University of Athens, Rimini 1, Chaidari, 12462 Athens, Greece; (E.M.); (J.T.); (G.P.); (K.K.); (D.V.); (E.K.); (G.K.)
| | - Konstantinos Katogiannis
- 2nd Cardiology Department, Attikon University Hospital, National and Kapodistrian University of Athens, Rimini 1, Chaidari, 12462 Athens, Greece; (E.M.); (J.T.); (G.P.); (K.K.); (D.V.); (E.K.); (G.K.)
| | - Dimitrios Vlachomitros
- 2nd Cardiology Department, Attikon University Hospital, National and Kapodistrian University of Athens, Rimini 1, Chaidari, 12462 Athens, Greece; (E.M.); (J.T.); (G.P.); (K.K.); (D.V.); (E.K.); (G.K.)
| | - Eleni Katsanaki
- 2nd Cardiology Department, Attikon University Hospital, National and Kapodistrian University of Athens, Rimini 1, Chaidari, 12462 Athens, Greece; (E.M.); (J.T.); (G.P.); (K.K.); (D.V.); (E.K.); (G.K.)
| | - Gavriella Kostelli
- 2nd Cardiology Department, Attikon University Hospital, National and Kapodistrian University of Athens, Rimini 1, Chaidari, 12462 Athens, Greece; (E.M.); (J.T.); (G.P.); (K.K.); (D.V.); (E.K.); (G.K.)
| | - Sotirios Pililis
- Diabetes Center, 2nd Department of Internal Medicine, Attikon University Hospital, Medical School, National and Kapodistrian University of Athens, Rimini 1, Chaidari, 12462 Athens, Greece; (S.L.); (S.P.); (L.P.); (A.K.); (V.L.)
| | - Loukia Pliouta
- Diabetes Center, 2nd Department of Internal Medicine, Attikon University Hospital, Medical School, National and Kapodistrian University of Athens, Rimini 1, Chaidari, 12462 Athens, Greece; (S.L.); (S.P.); (L.P.); (A.K.); (V.L.)
| | - Aikaterini Kountouri
- Diabetes Center, 2nd Department of Internal Medicine, Attikon University Hospital, Medical School, National and Kapodistrian University of Athens, Rimini 1, Chaidari, 12462 Athens, Greece; (S.L.); (S.P.); (L.P.); (A.K.); (V.L.)
| | - Ioannis S. Papanikolaou
- Hepatogastroenterology Unit, Second Department of Internal Medicine-Propaedeutic, Attikon University Hospital, Rimini 1, Chaidari, 12462 Athens, Greece;
| | - Vaia Lambadiari
- Diabetes Center, 2nd Department of Internal Medicine, Attikon University Hospital, Medical School, National and Kapodistrian University of Athens, Rimini 1, Chaidari, 12462 Athens, Greece; (S.L.); (S.P.); (L.P.); (A.K.); (V.L.)
| | - Ignatios Ikonomidis
- 2nd Cardiology Department, Attikon University Hospital, National and Kapodistrian University of Athens, Rimini 1, Chaidari, 12462 Athens, Greece; (E.M.); (J.T.); (G.P.); (K.K.); (D.V.); (E.K.); (G.K.)
| |
Collapse
|
2
|
Jelenkovic A, Ibáñez-Zamacona ME, Rebato E. Human adaptations to diet: Biological and cultural coevolution. ADVANCES IN GENETICS 2024; 111:117-147. [PMID: 38908898 DOI: 10.1016/bs.adgen.2024.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/24/2024]
Abstract
Modern humans evolved in Africa some 200,000 years ago, and since then, human populations have expanded and diversified to occupy a broad range of habitats and use different subsistence modes. This has resulted in different adaptations, such as differential responses to diseases and different abilities to digest or tolerate certain foods. The shift from a subsistence strategy based on hunting and gathering during the Palaeolithic to a lifestyle based on the consumption of domesticated animals and plants in the Neolithic can be considered one of the most important dietary transitions of Homo sapiens. In this text, we review four examples of gene-culture coevolution: (i) the persistence of the enzyme lactase after weaning, which allows the digestion of milk in adulthood, related to the emergence of dairy farming during the Neolithic; (ii) the population differences in alcohol susceptibility, in particular the ethanol intolerance of Asian populations due to the increased accumulation of the toxic acetaldehyde, related to the spread of rice domestication; (iii) the maintenance of gluten intolerance (celiac disease) with the subsequent reduced fitness of its sufferers, related to the emergence of agriculture and (iv) the considerable variation in the biosynthetic pathway of long-chain polyunsaturated fatty acids in native populations with extreme diets.
Collapse
Affiliation(s)
- Aline Jelenkovic
- Department of Genetics, Physical Anthropology and Animal Physiology, Faculty of Science and Technology, University of the Basque Country (UPV/EHU), Bilbao, Spain.
| | - María Eugenia Ibáñez-Zamacona
- Department of Genetics, Physical Anthropology and Animal Physiology, Faculty of Science and Technology, University of the Basque Country (UPV/EHU), Bilbao, Spain
| | - Esther Rebato
- Department of Genetics, Physical Anthropology and Animal Physiology, Faculty of Science and Technology, University of the Basque Country (UPV/EHU), Bilbao, Spain
| |
Collapse
|
3
|
Ochoa-Rios S, Grauzam SE, Gregory R, Angel PM, Drake RR, Helke KL, Mehta AS. Spatial Omics Reveals that Cancer-Associated Glycan Changes Occur Early in Liver Disease Development in a Western Diet Mouse Model of MASLD. J Proteome Res 2024; 23:786-796. [PMID: 38206822 DOI: 10.1021/acs.jproteome.3c00672] [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] [Indexed: 01/13/2024]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is a progressive disease and comprises different stages of liver damage; it is significantly associated with obese and overweight patients. Untreated MASLD can progress to life-threatening end-stage conditions, such as cirrhosis and liver cancer. N-Linked glycosylation is one of the most common post-translational modifications in the cell surface and secreted proteins. N-Linked glycan alterations have been established to be signatures of liver diseases. However, the N-linked glycan changes during the progression of MASLD to liver cancer are still unknown. Here, we induced different stages of MASLD in mice and liver-cancer-related phenotypes and elucidated the N-glycome profile during the progression of MASLD by quantitative and qualitative profiling in situ using matrix-assisted laser desorption ionization (MALDI) imaging mass spectrometry (IMS). Importantly, we identified specific N-glycan structures including fucosylated and highly branched N-linked glycans at very early stages of liver injury (steatosis), which in humans are associated with cancer development, establishing the importance of these modifications with disease progression. Finally, we report that N-linked glycan alterations can be observed in our models by MALDI-IMS before liver injury is identified by histological analysis. Overall, we propose these findings as promising biomarkers for the early diagnosis of liver injury in MASLD.
Collapse
Affiliation(s)
- Shaaron Ochoa-Rios
- Department of Cell and Molecular Pharmacology, Medical University of South Carolina, Charleston, South Carolina 29425, United States
| | - Stéphane Elie Grauzam
- Department of Cell and Molecular Pharmacology, Medical University of South Carolina, Charleston, South Carolina 29425, United States
| | - Rebecca Gregory
- Department of Comparative Medicine, Medical University of South Carolina, Charleston, South Carolina 29425, United States
| | - Peggi M Angel
- Department of Cell and Molecular Pharmacology, Medical University of South Carolina, Charleston, South Carolina 29425, United States
| | - Richard R Drake
- Department of Cell and Molecular Pharmacology, Medical University of South Carolina, Charleston, South Carolina 29425, United States
| | - Kristi L Helke
- Department of Comparative Medicine, Medical University of South Carolina, Charleston, South Carolina 29425, United States
| | - Anand S Mehta
- Department of Cell and Molecular Pharmacology, Medical University of South Carolina, Charleston, South Carolina 29425, United States
| |
Collapse
|
4
|
Barazesh M, Jalili S, Akhzari M, Faraji F, Khorramdin E. Recent Progresses on Pathophysiology, Diagnosis, Therapeutic Modalities,
and Management of Non-alcoholic Fatty Liver Disorder. CURRENT DRUG THERAPY 2024; 19:20-48. [DOI: 10.2174/1574885518666230417111247] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 01/30/2023] [Accepted: 02/06/2023] [Indexed: 01/03/2025]
Abstract
Abstract:
Non-alcoholic fatty liver disease (NAFLD) is currently the utmost common chronic liver
disorder that happens through all age groups and is identified to occur in 14%-30% of the general
population, demonstrating a critical and grossing clinical issue because of the growing incidence of
obesity and overweight. From the histological aspect, it looks like alcoholic liver damage, but it happens in patients who avoid remarkable alcohol usage. NAFLD comprises a broad spectrum, ranging
from benign hepatocellular steatosis to inflammatory nonalcoholic steatohepatitis (NASH), different
levels of fibrosis, and cirrhosis. Patients with NASH are more susceptible to more rapid progression to
cirrhosis and hepatocellular carcinoma. There is no single factor that drives proceeding from simple
steatosis to NASH. However, a combination of multi parameters such as genetic background, gut microflora, intake of high fat/ fructose dietary contents or methionine/choline-deficient diet, and consequently accumulated hepatocellular lipids mainly including triglycerides and also other bio-analytes,
such as free fatty acids, cholesterol, and phospholipids display a crucial role in disease promotion.
NAFLD is related to overweight and insulin resistance (IR) and is regarded as the hepatic presentation
of the metabolic syndrome, an amalgamation of medical statuses such as hyperlipidemia, hypertension, type 2 diabetes, and visceral obesity. Despite the increasing prevalence of this disease, which
imposes a remarkable clinical burden, most affected patients remain undiagnosed in a timely manner,
largely related to the asymptomatic entity of NAFLD patients and the unavailability of accurate and
efficient noninvasive diagnostic tests. However, liver biopsy is considered a gold standard for NAFLD
diagnosis, but due to being expensive and invasiveness is inappropriate for periodic disease screening.
Some noninvasive monitoring approaches have been established recently for NAFLD assessment. In
addition to the problem of correct disease course prediction, no effective therapeutic modalities are
approved for disease treatment. Imaging techniques can commonly validate the screening and discrimination of NAFLD; nevertheless, staging the disease needs a liver biopsy. The present therapeutic approaches depend on weight loss, sports activities, and dietary modifications, although different insulin-sensitizing drugs, antioxidants, and therapeutic agents seem hopeful. This review aims to focus on
the current knowledge concerning epidemiology, pathogenesis, and different biochemical experiments
and imaging modalities applied to diagnose the different grades of NAFLD and its management, as
well as new data about pharmacological therapies for this disorder.
Collapse
Affiliation(s)
- Mahdi Barazesh
- School of Paramedical, Gerash University of Medical Sciences, Gerash, Iran
| | - Sajad Jalili
- Department of Orthopedics, School of
Medicine, Ahvaz Jundishapour University of Medical Sciences, Ahvaz, Iran
| | - Morteza Akhzari
- School of Nursing, Larestan University of
Medical Sciences, Larestan, Iran
| | - Fouzieyeh Faraji
- School of Paramedical, Gerash University of Medical Sciences, Gerash, Iran
| | - Ebrahim Khorramdin
- Department of Orthopedics, School of
Medicine, Ahvaz Jundishapour University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
5
|
Tiniakos DG, Anstee QM, Brunt EM, Burt AD. Fatty Liver Disease. MACSWEEN'S PATHOLOGY OF THE LIVER 2024:330-401. [DOI: 10.1016/b978-0-7020-8228-3.00005-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
6
|
Mounika N, Yadav A, Kamboj P, Banerjee SK, Deka UJ, Kaur S, Adela R. Circulatory bone morphogenetic protein (BMP) 8B is a non-invasive predictive biomarker for the diagnosis of non-alcoholic steatohepatitis (NASH). PLoS One 2023; 18:e0295839. [PMID: 38127951 PMCID: PMC10734958 DOI: 10.1371/journal.pone.0295839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 11/28/2023] [Indexed: 12/23/2023] Open
Abstract
BACKGROUND Nonalcoholic fatty liver disease (NAFLD) is a complex disease which is characterized by the deposition of fats in the hepatocytes. Further, it progresses to nonalcoholic steatohepatitis (NASH), fibrosis, and hepatocellular carcinoma. The increasing prevalence of NAFLD urges to find the non-invasive predictive biomarkers. In this study, we sought to determine increased BMP8B levels as predictors for the progression of NAFLD. METHODS In the present cross-sectional study, circulatory BMP8B levels were measured in healthy controls (n = 56), NAFL patients (n = 72) and NASH patients (n = 77) by using an ELISA kit. Human hepatic BMP8B mRNA expression was measured in the liver tissue of control and NASH patients. In addition, BMP8B expression was confirmed by immunohistochemistry analysis. Furthermore, hepatic BMP8B mRNA expression was measured in wild type (WT) mice, WT mice fed with choline deficient high fat diet (WT+CDHF), iNOS (inducible nitric oxide synthase) knockout (iNOS-/-) mice, iNOS-/- fed with CDHF diet (iNOS-/-+CDHF). RESULTS Increased circulatory BMP8B levels and BMP8B mRNA expression in hepatic tissue were significantly higher in NASH patients as compared with the control subjects. BMP8B expression was increased parallel to the fibrosis score in the hepatic tissues of NASH patients. It was observed that increased BMP8B levels have shown a significant positive correlation between aspartate aminotransferase (r = 0.31, p = 0.005), alanine aminotransferase (r = 0.23, p = 0.045), APRI (r = 0.30, p = 0.009), and Fib-4 score (r = 0.25, p = 0.036) in NASH patients. BMP8B has maintained a significant association with NASH and shown high sensitivity (92.91%) and specificity (92.73%) in NASH patients. Furthermore, increased BMP8B mRNA expression levels were observed in iNOS-/-+CDHF mice. CONCLUSION Our study findings confirmed that BMP8B increases with the severity of the disease and BMP8B shows potential as a non-invasive predictive biomarker to identify NAFLD progression. However, future studies should investigate circulatory BMP8B levels in a large number of patients and also its impact on liver during NAFLD progression.
Collapse
Affiliation(s)
- Nadella Mounika
- Department of Pharmacy Practice, NIPER-Guwahati, Sila Katamur, Halugurisuk, Changsari, Dist.Kamrup, Guwahati, Assam, India
| | - Angeera Yadav
- Department of Pharmacy Practice, NIPER-Guwahati, Sila Katamur, Halugurisuk, Changsari, Dist.Kamrup, Guwahati, Assam, India
| | - Parul Kamboj
- Senior Researcher, Translational Health Science and Technology Institute (THSTI), Faridabad, India
| | - Sanjay K. Banerjee
- Senior Researcher, Translational Health Science and Technology Institute (THSTI), Faridabad, India
- Department of Biotechnology, NIPER-Guwahati, Sila Katamur, Halugurisuk, Changsari, Dist.Kamrup, Guwahati, Assam, India
| | - Utpal Jyoti Deka
- Department of Gastroenterology, Downtown Hospital, GS Rd, Bormotoria, Guwahati, Assam, India
| | - Savneet Kaur
- Department of Molecular and Cellular Medicine, Institute of Liver & Biliary Science (ILBS), Vasant Kunj, New Delhi, India
| | - Ramu Adela
- Department of Pharmacy Practice, NIPER-Guwahati, Sila Katamur, Halugurisuk, Changsari, Dist.Kamrup, Guwahati, Assam, India
| |
Collapse
|
7
|
Nadolsky K, Cryer DR, Articolo A, Fisher T, Schneider J, Rinella M. Nonalcoholic steatohepatitis diagnosis and treatment from the perspective of patients and primary care physicians: a cross-sectional survey. Ann Med 2023; 55:2211349. [PMID: 37171239 PMCID: PMC10184582 DOI: 10.1080/07853890.2023.2211349] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/13/2023] Open
Abstract
BACKGROUND The global prevalence of nonalcoholic steatohepatitis (NASH) is rising. Despite this, NASH is underdiagnosed and does not yet have approved pharmacological treatments. We sought to understand the path to diagnosis, patient interactions with healthcare professionals, treatment regimens, and disease management for patients with NASH. METHODS Cross-sectional online surveys of patients with a self-reported diagnosis of NASH and healthcare professionals treating patients with NASH were conducted from 10th November 2020, to 1st January 2021. This manuscript focuses on responses from 152 patients with NASH and 101 primary care physicians (PCPs). RESULTS Patients (n = 152, mean age = 40, SD = 11) and healthcare professionals (n = 226) were located throughout the US. In the most common patient journey, 72% of patients had initial discussions about symptoms with a PCP but only 30% report receiving their NASH diagnosis from a PCP. Almost half of PCPs (47%) were not aware of any clinical practice guidelines for diagnosis and management of NASH. For ongoing management of NASH, PCPs most frequently prescribed lifestyle changes such as exercise (89%), lifestyle changes focused on diet (79%), and/or metformin (57%). Other healthcare professionals rarely referred patients to PCPs for treatment, but when they did, the primary reasons were patients struggling with lifestyle modifications (58%), needing to lose weight (46%), and needing treatment of comorbidities (42%). CONCLUSIONS PCPs may benefit from greater awareness of NASH and guidelines for its diagnosis and treatment. Given the absence of pharmacological treatments approved for NASH, PCPs can offer support in obesity management, comorbidity management, and risk stratification for liver disease progression.
Collapse
Affiliation(s)
- Karl Nadolsky
- MI State University College of Human Medicine, Holland Hospital Endocrinology, Obesity & Diabetes, Holland, MI, USA
| | | | | | | | | | - Mary Rinella
- Department of Medicine, University of Chicago Medicine, Chicago, IL, USA
| |
Collapse
|
8
|
Janini E, Fteiha B, Ramlawi I, Mahamid M. Clinical Trajectory and Predictors of Intensive Care Unit Mortality Among Nonalcoholic Fatty Liver Disease Patients: A Retrospective Case-Control Study. J Clin Exp Hepatol 2023; 13:218-224. [PMID: 36950493 PMCID: PMC10025687 DOI: 10.1016/j.jceh.2022.11.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 11/18/2022] [Indexed: 12/02/2022] Open
Abstract
Background Despite being the most common liver disease worldwide, the clinical trajectory and inpatient crude mortality rate of nonalcoholic fatty liver disease (NAFLD) patients admitted to the intensive care unit (ICU) have not been thoroughly studied. Methods We conducted a single-center retrospective case-control study of patients admitted to a general ICU setting between the years 2015 and 2020. Medical records from patients who met the diagnostic criteria for NAFLD, as well as age- and gender-matched control group, were reviewed. The primary endpoint was crude ICU mortality, defined as death within 30 days of ICU admission. The secondary outcomes included presentation with septic shock and severe sepsis, Sequential Organ Failure Assessment score and Acute Physiology and Chronic Health Evaluation II scores, vasopressor requirements, mechanical ventilation need, and admission-to-ICU transfer time. Results Two hundred fifty subjects were enrolled and were equally divided into the NAFLD and control groups. NAFLD group subjects had higher overall 30-day ICU mortality (63.9% vs 36.1%, P < 0.05), more frequent presentation with septic shock and severe sepsis (55.2% vs 33.6%, P < 0.05), higher Acute Physiology and Chronic Health Evaluation II and Sequential Organ Failure Assessment scores at presentation (21.3 ± 12.5 vs 16.6 ± 10.5 and 11.36 ± 5.2 vs 8.3 ± 6.2, P < 0.05), higher need for mechanical ventilation (18.4 vs 7.2%, P = 0.05), and vasopressor (15.2% vs 7.2%, P = 0.05) dependency on admission with a shorter admission-to-ICU transfer mean interval (3 vs 6 days, P < 0.05). There were no differences in the need for blood transfusions, steroids, or dialysis between the two groups. Higher fibrosis-4 (FIB-4) and NAFLD fibrosis scores were found to be associated with mortality in ICU-admitted NAFLD patients. Conclusion NAFLD patients are more likely than non-NAFLD admitted ICU patients to present with severe sepsis and septic shock, have a shorter admission-to-ICU transfer time, and have a higher crude ICU mortality rate.
Collapse
Affiliation(s)
- Eiad Janini
- Department of Internal Medicine, Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | - Bashar Fteiha
- Faculty of Medicine, Hebrew University of Jerusalem, Israel; The Digestive Diseases Institute, Shaare Zedek Medical Center, Jerusalem 9103102, Israel
| | - Imran Ramlawi
- ICU Ward and the Faculty of Medicine, Hebrew University of Jerusalem, Shaare Zedek Medical Center, Jerusalem, Israel
| | - Mahmud Mahamid
- Faculty of Medicine, Hebrew University of Jerusalem, Israel; The Digestive Diseases Institute, Shaare Zedek Medical Center, Jerusalem 9103102, Israel
| |
Collapse
|
9
|
Creatine supplementation protects against diet-induced non-alcoholic fatty liver but exacerbates alcoholic fatty liver. Life Sci 2022; 310:121064. [PMID: 36220368 DOI: 10.1016/j.lfs.2022.121064] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 09/26/2022] [Accepted: 10/06/2022] [Indexed: 11/06/2022]
Abstract
AIMS This work investigated the effects of creatine supplementation on different pathways related to the pathogenesis of non-alcoholic fatty liver disease and alcoholic liver disease. MAIN METHODS To induce alcoholic liver disease, male Swiss mice were divided into three groups: control, ethanol and ethanol supplemented with creatine. To induce non-alcoholic fatty liver disease, mice were divided into three groups: control, high-fat diet and high-fat diet supplemented with creatine. Each group consisted of eight animals. In both cases, creatine monohydrate was added to the diets (1 %; weight/vol). KEY FINDINGS Creatine supplementation prevented high-fat diet-induced non-alcoholic fatty liver disease progression, demonstrated by attenuated liver fat accumulation and liver damage. On the other hand, when combined with ethanol, creatine supplementation up-regulated key genes related to ethanol metabolism, oxidative stress, inflammation and lipid synthesis, and exacerbated ethanol-induced liver steatosis and damage, demonstrated by increased liver fat accumulation and histopathological score, as well as elevated oxidative damage markers and inflammatory mediators. SIGNIFICANCE Our results clearly demonstrated creatine supplementation exerts different outcomes in relation to non-alcoholic fatty liver disease and alcoholic liver disease, namely it protects against high-fat diet-induced non-alcoholic fatty liver disease but exacerbates ethanol-induced alcoholic liver disease. The exacerbating effects of the creatine and ethanol combination appear to be related to oxidative stress and inflammation-mediated up-regulation of ethanol metabolism.
Collapse
|
10
|
Hidalgo I, Ortiz-Flores M, Villarreal F, Fonseca-Coronado S, Ceballos G, Meaney E, Nájera N. Is it possible to treat nonalcoholic liver disease using a flavanol-based nutraceutical approach? Basic and clinical data. J Basic Clin Physiol Pharmacol 2022; 33:703-714. [PMID: 35119232 DOI: 10.1515/jbcpp-2021-0285] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 01/15/2022] [Indexed: 01/05/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is characterized by a spectrum of diseases, ranging from simple steatosis to hepatocellular carcinoma. The main factors for NAFLD are closely related to obesity, insulin resistance, intestinal microbiota alterations, hyperinsulinism, low-grade systemic inflammation, nitroxidative stress, lipid peroxidation, and mitochondrial dysfunction. Currently, the treatment of NAFLD is based on diet and exercise because, to date, there is no specific pharmacological agent, already approved, that raises the need for new therapeutic strategies. Nutraceuticals, such as polyphenols, have potential beneficial effects for health. In this article, the beneficial effects of epigallocatechin-3-gallate (EGCG) and (-)-epicatechin (EC) are discussed. EGCG is the main catechin in green tea, which has shown in various studies its potential effect preventing and treating NAFLD since it has shown antihyperlipidemic, anti-inflammatory, antifibrotic, antioxidant, and improvement of liver lipid metabolism. However, it has been found that excessive consumption may cause hepatotoxicity. EC is widely distributed in nature (fruits and vegetables). This flavanol has shown many beneficial effects, including antihypertensive, anti-inflammatory, anti-hyperglycemic, antithrombotic, and antifibrotic properties. It increases mitochondrial biogenesis, and it also has effects on the regulation of synthesis and metabolism of lipids. This flavanol is a nontoxic substance; it has been classified by the United States Food and Drug Administration as harmless. The EC-induced effects can be useful for the prevention and/or treatment of NAFLD.
Collapse
Affiliation(s)
- Isabel Hidalgo
- Unidad de Investigación Laboratorio de Investigación en Inmunología y Salud Publica, Facultad de Estudios Superiores Cuautitlán, Universidad Nacional Autónoma de México, Estado de México, Mexico
| | - Miguel Ortiz-Flores
- Laboratorio de investigación integral cardiometabólica, Escuela Superior de Medicina, Instituto Politécnico Nacional, CDMX, Mexico
| | | | - Salvador Fonseca-Coronado
- Unidad de Investigación Laboratorio de Investigación en Inmunología y Salud Publica, Facultad de Estudios Superiores Cuautitlán, Universidad Nacional Autónoma de México, Estado de México, Mexico
| | - Guillermo Ceballos
- Laboratorio de investigación integral cardiometabólica, Escuela Superior de Medicina, Instituto Politécnico Nacional, CDMX, Mexico
| | - Eduardo Meaney
- Laboratorio de investigación integral cardiometabólica, Escuela Superior de Medicina, Instituto Politécnico Nacional, CDMX, Mexico
| | - Nayelli Nájera
- Laboratorio de investigación integral cardiometabólica, Escuela Superior de Medicina, Instituto Politécnico Nacional, CDMX, Mexico
| |
Collapse
|
11
|
Briand F, Sencio V, Robil C, Heumel S, Deruyter L, Machelart A, Barthelemy J, Bogard G, Hoffmann E, Infanti F, Domenig O, Chabrat A, Richard V, Prévot V, Nogueiras R, Wolowczuk I, Pinet F, Sulpice T, Trottein F. Diet-Induced Obesity and NASH Impair Disease Recovery in SARS-CoV-2-Infected Golden Hamsters. Viruses 2022; 14:v14092067. [PMID: 36146875 PMCID: PMC9503118 DOI: 10.3390/v14092067] [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: 08/05/2022] [Revised: 09/12/2022] [Accepted: 09/14/2022] [Indexed: 01/08/2023] Open
Abstract
Obese patients with non-alcoholic steatohepatitis (NASH) are prone to severe forms of COVID-19. There is an urgent need for new treatments that lower the severity of COVID-19 in this vulnerable population. To better replicate the human context, we set up a diet-induced model of obesity associated with dyslipidemia and NASH in the golden hamster (known to be a relevant preclinical model of COVID-19). A 20-week, free-choice diet induces obesity, dyslipidemia, and NASH (liver inflammation and fibrosis) in golden hamsters. Obese NASH hamsters have higher blood and pulmonary levels of inflammatory cytokines. In the early stages of a SARS-CoV-2 infection, the lung viral load and inflammation levels were similar in lean hamsters and obese NASH hamsters. However, obese NASH hamsters showed worse recovery (i.e., less resolution of lung inflammation 10 days post-infection (dpi) and lower body weight recovery on dpi 25). Obese NASH hamsters also exhibited higher levels of pulmonary fibrosis on dpi 25. Unlike lean animals, obese NASH hamsters infected with SARS-CoV-2 presented long-lasting dyslipidemia and systemic inflammation. Relative to lean controls, obese NASH hamsters had lower serum levels of angiotensin-converting enzyme 2 activity and higher serum levels of angiotensin II—a component known to favor inflammation and fibrosis. Even though the SARS-CoV-2 infection resulted in early weight loss and incomplete body weight recovery, obese NASH hamsters showed sustained liver steatosis, inflammation, hepatocyte ballooning, and marked liver fibrosis on dpi 25. We conclude that diet-induced obesity and NASH impair disease recovery in SARS-CoV-2-infected hamsters. This model might be of value for characterizing the pathophysiologic mechanisms of COVID-19 and evaluating the efficacy of treatments for the severe forms of COVID-19 observed in obese patients with NASH.
Collapse
Affiliation(s)
| | - Valentin Sencio
- Univ. Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019-UMR 9017-CIIL-Center for Infection and Immunity of Lille, F-59000 Lille, France
| | - Cyril Robil
- Univ. Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019-UMR 9017-CIIL-Center for Infection and Immunity of Lille, F-59000 Lille, France
| | - Séverine Heumel
- Univ. Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019-UMR 9017-CIIL-Center for Infection and Immunity of Lille, F-59000 Lille, France
| | - Lucie Deruyter
- Univ. Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019-UMR 9017-CIIL-Center for Infection and Immunity of Lille, F-59000 Lille, France
| | - Arnaud Machelart
- Univ. Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019-UMR 9017-CIIL-Center for Infection and Immunity of Lille, F-59000 Lille, France
| | - Johanna Barthelemy
- Univ. Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019-UMR 9017-CIIL-Center for Infection and Immunity of Lille, F-59000 Lille, France
| | - Gemma Bogard
- Univ. Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019-UMR 9017-CIIL-Center for Infection and Immunity of Lille, F-59000 Lille, France
| | - Eik Hoffmann
- Univ. Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019-UMR 9017-CIIL-Center for Infection and Immunity of Lille, F-59000 Lille, France
| | | | | | | | | | - Vincent Prévot
- Univ. Lille, INSERM, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, UMR-S 1172, European Genomic Institute for Diabetes (EGID), F-59000 Lille, France
| | - Ruben Nogueiras
- Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), S-15781 Santiago de Compostela, Spain
| | - Isabelle Wolowczuk
- Univ. Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019-UMR 9017-CIIL-Center for Infection and Immunity of Lille, F-59000 Lille, France
| | - Florence Pinet
- Univ. Lille, INSERM, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE-Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, F-59000 Lille, France
| | | | - François Trottein
- Univ. Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019-UMR 9017-CIIL-Center for Infection and Immunity of Lille, F-59000 Lille, France
- Correspondence:
| |
Collapse
|
12
|
Li H, Sun J, Li B, Jiang A, Tao J, Ning C, Li R, Liu H. AMPK-PPARγ-Cidec Axis Drives the Fasting-Induced Lipid Droplet Aggregation in the Liver of Obese Mice. Front Nutr 2022; 9:917801. [PMID: 35859752 PMCID: PMC9289538 DOI: 10.3389/fnut.2022.917801] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 05/17/2022] [Indexed: 11/13/2022] Open
Abstract
Intermittent fasting is one of the most common clinical treatments for the obesity, a main risk factor of the metabolic syndrome which can lead to a variety of diseases. Fasting-induced fat mobilization alters the metabolic state of lipid in the liver, predisposing to increase the hepatic lipid droplet aggregation and triglyceride levels. However, the underlying mechanisms regarding the lipid droplet aggregation in the liver after fasting remains elusive. Here, we report that a lipid droplet surface binding protein Cidec (cell death inducing DFFA like effector C) is activated by AMPK to regulate the hepatic lipid droplet fusion following fasting in obese mice. Specifically, we found that lipid droplets were significantly aggregated in the liver of high-fat-diet and ob/ob mice after 16 and 24 h of fasting, accompanied by the dramatically up-regulated expression of Cidec. Consistently, overexpression of Cidec in the AML12 cells resulted in the intracellular lipid droplet aggregation. Furthermore, we showed that fasting caused the up-regulated expression of AMPK, which in turn activated the transcription of Cidec through the transcription factor PPARγ. Altogether, our observations reveal that fasting-induced hepatic lipid droplet aggregation is mediated by the AMPK-activated expression of Cidec via PPARγ, extending our understanding about the molecular mechanism of the impact of fasting on the obesity and providing potential targets for the treatment of human obesity.
Collapse
Affiliation(s)
- Hongqiang Li
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
- Hebei Key Laboratory of Specialty Animal Germplasm Resources Exploration and Innovation, College of Animal Science and Technology, Hebei Normal University of Science and Technology, Qinhuangdao, China
| | - Jian Sun
- Hebei Key Laboratory of Specialty Animal Germplasm Resources Exploration and Innovation, College of Animal Science and Technology, Hebei Normal University of Science and Technology, Qinhuangdao, China
| | - Bojiang Li
- College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, China
| | - Aiwen Jiang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Jingli Tao
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Caibo Ning
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Rongyang Li
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Honglin Liu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
- *Correspondence: Honglin Liu
| |
Collapse
|
13
|
Ochoa-Rios S, O'Connor IP, Kent LN, Clouse JM, Hadjiyannis Y, Koivisto C, Pecot T, Angel PM, Drake RR, Leone G, Mehta AS, Rockey DC. Imaging Mass Spectrometry Reveals Alterations in N-Linked Glycosylation That Are Associated With Histopathological Changes in Nonalcoholic Steatohepatitis in Mouse and Human. Mol Cell Proteomics 2022; 21:100225. [PMID: 35331917 PMCID: PMC9092512 DOI: 10.1016/j.mcpro.2022.100225] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 03/14/2022] [Accepted: 03/17/2022] [Indexed: 01/20/2023] Open
Abstract
Nonalcoholic steatohepatitis (NASH) is the progressive form of nonalcoholic fatty liver disease (NAFLD) and is characterized by inflammation, hepatocyte injury, and fibrosis. Further, NASH is a risk factor for cirrhosis and hepatocellular carcinoma. Previous research demonstrated that serum N-glycan profiles can be altered in NASH patients. Here, we hypothesized that these N-glycan modifications may be associated with specific liver damage in NAFLD and NASH. To investigate the N-glycome profile in tissue, imaging mass spectrometry was used for a qualitative and quantitative in situ N-linked glycan analysis of mouse and human NAFLD/NASH tissue. A murine model was used to induce NAFLD and NASH through ad libitum feeding with either a high-fat diet or a Western diet, respectively. Mice fed a high-fat diet or Western diet developed inflammation, steatosis, and fibrosis, consistent with NAFLD/NASH phenotypes. Induction of NAFLD/NASH for 18 months using high caloric diets resulted in increased expression of mannose, complex/fucosylated, and hybrid N-glycan structures compared to control mouse livers. To validate the animal results, liver biopsy specimens from 51 human NAFLD/NASH patients representing the full range of NASH Clinical Research Network fibrosis stages were analyzed. Importantly, the same glycan alterations observed in mouse models were observed in human NASH biopsies and correlated with the degree of fibrosis. In addition, spatial glycan alterations were localized specifically to histopathological changes in tissue like fibrotic and fatty areas. We demonstrate that the use of standard staining's combined with imaging mass spectrometry provide a full profile of the origin of N-glycan modifications within the tissue. These results indicate that the spatial distribution of abundances of released N-glycans correlate with regions of tissue steatosis associated with NAFLD/NASH.
Collapse
Affiliation(s)
- Shaaron Ochoa-Rios
- Department of Cell and Molecular Pharmacology, Medical University of South Carolina, Charleston, South Carolina, USA.
| | - Ian P O'Connor
- Digestive Disease Research Center, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Lindsey N Kent
- Department of Obstetrics and Gynecology, Washington University in St Louis Center for Reproductive Health Sciences, St Louis, Missouri, USA
| | - Julian M Clouse
- Department of Cancer Biology and Genetics, The Ohio State University Comprehensive Cancer Center, Columbus, Ohio, USA
| | - Yannis Hadjiyannis
- Department of Cancer Biology and Genetics, The Ohio State University Comprehensive Cancer Center, Columbus, Ohio, USA
| | - Christopher Koivisto
- Department of Biochemistry and Molecular Biology, Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina, USA
| | | | - Peggi M Angel
- Department of Cell and Molecular Pharmacology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Richard R Drake
- Department of Cell and Molecular Pharmacology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Gustavo Leone
- Department of Biochemistry and Molecular Biology, Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina, USA; Department of Biochemistry, Medical College of Wisconsin, MCW Cancer Center, Milwaukee, Wisconsin, USA
| | - Anand S Mehta
- Department of Cell and Molecular Pharmacology, Medical University of South Carolina, Charleston, South Carolina, USA.
| | - Don C Rockey
- Digestive Disease Research Center, Medical University of South Carolina, Charleston, South Carolina, USA
| |
Collapse
|
14
|
Traber MG, Head B. Vitamin E: How much is enough, too much and why! Free Radic Biol Med 2021; 177:212-225. [PMID: 34699937 DOI: 10.1016/j.freeradbiomed.2021.10.028] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 09/08/2021] [Accepted: 10/11/2021] [Indexed: 12/12/2022]
Abstract
α-Tocopherol (α-T) is a required dietary nutrient for humans and thus is a vitamin. This narrative review focuses on vitamin E structures, functions, biological determinants and its deficiency symptoms in humans. The mechanisms for the preferential α-T tissue enrichment in the human body include the α-T transfer protein (TTPA) and the preferential metabolism of non-α-T forms. Potential new α-T biomarkers, pharmacokinetic data, and whether there are better approaches to evaluate and set the α-T dietary requirement are discussed. Finally, the possible role of α-T supplements in delay of chronic diseases and the evaluation of vitamin E safety are considered.
Collapse
Affiliation(s)
- Maret G Traber
- Linus Pauling Institute, USA; School of Biological and Population Health Sciences, College of Public Health and Human Sciences, USA.
| | - Brian Head
- Linus Pauling Institute, USA; Molecular and Cell Biology Program, Oregon State University, Corvallis, OR, USA
| |
Collapse
|
15
|
Ore A, Akinloye OA, Adeogun AI, Ugbaja RN, Morifi E, Makatini M, Moepya R, Mbhele T. Buchholzia coriacea seed (wonderful kolanut) alleviates insulin resistance, steatosis, inflammation and oxidative stress in high fat diet model of fatty liver disease. J Food Biochem 2021; 46:e13836. [PMID: 34184286 DOI: 10.1111/jfbc.13836] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 05/17/2021] [Accepted: 06/07/2021] [Indexed: 12/12/2022]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a hepatic condition with multiple pathological features and it currently has no specific treatment or approved drug. Wonderful kolanut widely consumed fresh or cooked has been applied in the treatment of numerous diseases in folk medicine. In this study, we evaluate the therapeutic potentials of hydroethanolic extract of defatted Buccholzia coriacea seeds (HEBCS) in NAFLD model. HEBCS was subjected to liquid chromatography - mass spectrometry, and 30 male BALB/c mice (28 ± 2 g) were allocated to three (3) experimental groups (n = 10/group). Mice in group I were fed chow diet (CD); those in group II, high fat diet (HFD) and group III, HFD and 250 mg/kg HEBCS p.o. daily for six weeks. HEBCS alleviates HFD-induced insulin resistance and high plasma insulin and glucose levels. It further alleviates hepatic steatosis, and alters plasma lipid profile. HEBCS also protected against HFD-induced inflammation, oxidative stress and hepatocellular damage. In conclusion, HEBCS alleviated NAFLD in mice via suppression of insulin resistance, hyperlipidemia, inflammation and oxidative stress. PRACTICAL APPLICATIONS: Bioactive polyphenols and alkaloids were identified in hydroethanolic extract of defatted Buccholzia coriacea seeds (HEBCS). This study projects HEBCS as a potential therapeutic agent in the treatment of NAFLD. NAFLD is a multi-factorial condition and therefore, HEBCS is promising considering its multiple-target actions in the current model of NAFLD. HEBCS alleviates insulin resistance, metabolic dysfunction, steatosis, and inflammation in this model. There is a need to further investigate HEBCS in other models of NAFLD as a lead to future use in clinical studies.
Collapse
Affiliation(s)
- Ayokanmi Ore
- Department of Biochemistry, College of Biosciences, Federal University of Agriculture, Abeokuta, Nigeria.,Biochemistry Division, Department of Chemical Sciences, Faculty of Natural Sciences, Ajayi Crowther University, Oyo, Nigeria
| | - Oluseyi Adeboye Akinloye
- Department of Biochemistry, College of Biosciences, Federal University of Agriculture, Abeokuta, Nigeria
| | - Abideen Idowu Adeogun
- Department of Chemistry, College of Physical Sciences, Federal University of Agriculture, Abeokuta, Nigeria
| | - Regina Ngozi Ugbaja
- Department of Biochemistry, College of Biosciences, Federal University of Agriculture, Abeokuta, Nigeria
| | - Eric Morifi
- School of Chemistry, Mass Spectrometry Division, Wits University, Johannesburg, South Africa
| | - Maya Makatini
- School of Chemistry, Mass Spectrometry Division, Wits University, Johannesburg, South Africa
| | - Refilwe Moepya
- School of Chemistry, Mass Spectrometry Division, Wits University, Johannesburg, South Africa
| | - Thapelo Mbhele
- School of Chemistry, Mass Spectrometry Division, Wits University, Johannesburg, South Africa
| |
Collapse
|
16
|
Protective Effect of Cudrania tricuspidata Extract against High-Fat Diet Induced Nonalcoholic Fatty Liver Disease through Nrf-2/HO-1 Pathway. Molecules 2021; 26:molecules26092434. [PMID: 33922045 PMCID: PMC8122508 DOI: 10.3390/molecules26092434] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 04/19/2021] [Accepted: 04/20/2021] [Indexed: 12/13/2022] Open
Abstract
Nonalcoholic fatty liver disease is the most common chronic disease affecting a wide range of the world’s population and associated with obesity-induced metabolic syndrome. It is possibly emerging as a leading cause of life-threatening liver diseases for which a drug with a specific therapeutic target has not been developed yet. Previously, there have been reports on the benefits of Cudrania tricuspidata (CT) for treating obesity and diabetes via regulation of metabolic processes, such as lipogenesis, lipolysis, and inflammation. In this study, we investigated the ameliorative effect of orally administered 0.25% and 0.5% (w/w) CT mixed with high-fat diet (HFD) to C57BL/6J mice for 7 weeks. It was found that body weight, fat mass, hepatic mass, serum glucose level, and liver cholesterol levels were significantly reduced after CT treatment. In CT-treated HFD-fed mice, the mRNA expression levels of hepatic lipogenic and inflammatory cytokine-related genes were markedly reduced, whereas the expression level of epididymal lipogenic genes was increased. The mRNA expression level of beta-oxidation and Nrf-2/HO-1 genes significantly increased in CT-treated obese mice livers. We propose that CT alleviates hepatic steatosis by reducing oxidative stress and inflammation.
Collapse
|
17
|
Canbay A, Kachru N, Haas JS, Meise D, Ozbay AB, Sowa JP. Healthcare resource utilization and costs among nonalcoholic fatty liver disease patients in Germany. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:615. [PMID: 33987313 PMCID: PMC8106103 DOI: 10.21037/atm-20-7179] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Background Patients with nonalcoholic fatty liver disease (NAFLD) and nonalcoholic steatohepatitis (NASH) are associated with progression to advanced liver diseases that include compensated cirrhosis, decompensated cirrhosis, liver transplantation, and hepatocellular carcinoma (HCC). This study characterized comorbidities, healthcare resource utilization (HRU), and associated costs among NAFLD patients in Germany. Methods German healthcare claims data between 2011 and 2016 were analyzed retrospectively. Adult patients diagnosed with NAFLD and/or NASH were categorized as NAFLD, NAFLD non-progressors, compensated cirrhosis, decompensated cirrhosis, liver transplant, or HCC. Within each stage, annual all-cause HRU and costs were measured during the pre- and post-index periods. Results Among 4,580,434 patients in the database, proportion of NAFLD was 4.7% (n=215,655). Of them, 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 (52.07%, 56.46%, 57.58% vs. 27.49% for cardiovascular disease; 77.13%, 76.61%, 83.47% vs. 54.89% for hypertension; 47.20%, 53.81%, 52.89% vs. 35.21% for hyperlipidemia; 49.88%, 36.67%, 48.21% vs. 20.38% for type 2 diabetes mellitus). The mean annual numbers of post-index outpatient visits and inpatient hospitalizations were significantly higher in patients with advanced liver diseases versus non-progressors. Mean annual costs were significantly higher among patients with advanced liver diseases (compensated cirrhosis, €10,291; decompensated cirrhosis, €22,561; liver transplant, €34,089; HCC, €35,910) than non-progressors (€3,818, P<0.001, except liver transplant cohort). This trend remained consistent after adjusting for baseline demographics and comorbidities. Conclusions NAFLD patients in Germany are grossly underdiagnosed and exert substantial healthcare resource use and economic burden, particularly those with advanced liver diseases. Optimal strategies for early identification and management are needed to prevent disease progression and limit the rising costs.
Collapse
Affiliation(s)
- Ali Canbay
- Department of Internal Medicine, Ruhr-University Bochum, Bochum, Germany
| | - Nandita Kachru
- Gilead Sciences, Inc., Health Economics Outcomes Research, Foster City, CA, USA
| | | | | | - A Burak Ozbay
- Gilead Sciences, Inc., Health Economics Outcomes Research, Foster City, CA, USA
| | - Jan-Peter Sowa
- Department of Internal Medicine, Ruhr-University Bochum, Bochum, Germany
| |
Collapse
|
18
|
Briand F, Maupoint J, Brousseau E, Breyner N, Bouchet M, Costard C, Leste-Lasserre T, Petitjean M, Chen L, Chabrat A, Richard V, Burcelin R, Dubroca C, Sulpice T. Elafibranor improves diet-induced nonalcoholic steatohepatitis associated with heart failure with preserved ejection fraction in Golden Syrian hamsters. Metabolism 2021; 117:154707. [PMID: 33444606 DOI: 10.1016/j.metabol.2021.154707] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 01/04/2021] [Accepted: 01/07/2021] [Indexed: 12/11/2022]
Abstract
BACKGROUND Cardiovascular disease is the leading cause of deaths in nonalcoholic steatohepatitis (NASH) patients. Mouse models, while widely used for drug development, do not fully replicate human NASH nor integrate the associated cardiac dysfunction, i.e. heart failure with preserved ejection fraction (HFpEF). To overcome these limitations, we established a nutritional hamster model developing both NASH and HFpEF. We then evaluated the effects of the dual peroxisome proliferator activated receptor alpha/delta agonist elafibranor developed for the treatment of NASH patients. METHODS Male Golden Syrian hamsters were fed for 10 to 20 weeks with a free choice diet, which presents hamsters with a choice between control chow diet with normal drinking water or a high fat/high cholesterol diet with 10% fructose enriched drinking water. Biochemistry, histology and echocardiography analysis were performed to characterize NASH and HFpEF. Once the model was validated, elafibranor was evaluated at 15 mg/kg/day orally QD for 5 weeks. RESULTS Hamsters fed a free choice diet for up to 20 weeks developed NASH, including hepatocyte ballooning (as confirmed with cytokeratin-18 immunostaining), bridging fibrosis, and a severe diastolic dysfunction with restrictive profile, but preserved ejection fraction. Elafibranor resolved NASH, with significant reduction in ballooning and fibrosis scores, and improved diastolic dysfunction with significant reduction in E/A and E/E' ratios. CONCLUSION Our data demonstrate that the free choice diet induced NASH hamster model replicates the human phenotype and will be useful for validating novel drug candidates for the treatment of NASH and associated HFpEF.
Collapse
Affiliation(s)
- François Briand
- Physiogenex, 280 rue de l'Hers, ZAC de la Masquère, 31750 Escalquens, France.
| | - Julie Maupoint
- Cardiomedex, 280 rue de l'Hers, ZAC de la Masquère, 31750 Escalquens, France
| | - Emmanuel Brousseau
- Physiogenex, 280 rue de l'Hers, ZAC de la Masquère, 31750 Escalquens, France
| | - Natalia Breyner
- Physiogenex, 280 rue de l'Hers, ZAC de la Masquère, 31750 Escalquens, France
| | - Mélanie Bouchet
- Physiogenex, 280 rue de l'Hers, ZAC de la Masquère, 31750 Escalquens, France
| | - Clément Costard
- Cardiomedex, 280 rue de l'Hers, ZAC de la Masquère, 31750 Escalquens, France
| | | | - Mathieu Petitjean
- PharmaNest, 100 Overlook Center, FL2, Princeton, NJ 08540, United States of America
| | - Li Chen
- PharmaNest, 100 Overlook Center, FL2, Princeton, NJ 08540, United States of America
| | - Audrey Chabrat
- Sciempath Labo, 7 rue de la Gratiole, 37270 Larcay, France
| | | | - Rémy Burcelin
- Inserm U1048 CHU Rangueil, BP 84225, 31432 Toulouse Cedex 4, France
| | - Caroline Dubroca
- Cardiomedex, 280 rue de l'Hers, ZAC de la Masquère, 31750 Escalquens, France
| | - Thierry Sulpice
- Physiogenex, 280 rue de l'Hers, ZAC de la Masquère, 31750 Escalquens, France; Cardiomedex, 280 rue de l'Hers, ZAC de la Masquère, 31750 Escalquens, France
| |
Collapse
|
19
|
Abdel-Maboud M, Menshawy A, Menshawy E, Emara A, Alshandidy M, Eid M. The efficacy of vitamin E in reducing non-alcoholic fatty liver disease: a systematic review, meta-analysis, and meta-regression. Therap Adv Gastroenterol 2020; 13:1756284820974917. [PMID: 33335561 PMCID: PMC7724271 DOI: 10.1177/1756284820974917] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 10/29/2020] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Non-alcoholic fatty liver disease (NAFLD) affects up to 30% of the population. Clinical trials have questioned the role of vitamin E in the treatment of NAFLD with or without other interventions, with still no firm conclusion reached. This study aims to examine the efficiency of vitamin E alone or combined in the management of NAFLD. METHODS We performed a systematic literature search on PubMed, Scopus, Embase, Ovid, EBSCO host, Science Direct, Web of Science, and Cochrane CENTRAL for randomized controlled trials (RCTs) of the role of vitamin E alone or combined in NAFLD patients. Extracted manuscripts reported data on biochemical, histological, anthropometric, and metabolic outcomes. Baseline characteristics, settings, dosage, and frequency were also collected. RESEARCH A total of 1317 patients from 15 RCTs were included in our systematic review and meta-analysis. Vitamin E was superior at improving alanine aminotransferase (ALT), aspartate aminotransferase (AST), NAFLD activity score (NAS), and fibrosis in short- and long-term follow up in the adult population, and long-term follow up in the pediatric population. Improvements in metabolic outcomes were best noticed in pediatric patients. Results from multiple regression models showed a significant association between ALT-AST levels and vitamin E dose. AST levels had a significant effect on NAS, and patients with a baseline AST > 50 IU/l showed more promising results. Changes in weight and body mass index (BMI) were strongly associated with changes in NAS. CONCLUSION Current evidence affirms that vitamin E - whether alone or combined - improves biochemical and histological outcomes in adults and pediatric patients.
Collapse
Affiliation(s)
| | - Amr Menshawy
- Faculty of Medicine, Al-Azhar University, Cairo,
Egypt
| | | | - Amany Emara
- Faculty of Medicine, Al-Azhar University, Cairo,
Egypt
| | | | - Muhammad Eid
- Faculty of Medicine, Al-Azhar University, Cairo,
Egypt
| |
Collapse
|
20
|
Hosseinifard ES, Bavafa-Valenlia K, Saghafi-Asl M, Morshedi M. Antioxidative and Metabolic Effects of Lactobacillus plantarum, Inulin, and Their Synbiotic on the Hypothalamus and Serum of Healthy Rats. Nutr Metab Insights 2020; 13:1178638820925092. [PMID: 33177834 PMCID: PMC7592325 DOI: 10.1177/1178638820925092] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Accepted: 04/13/2020] [Indexed: 02/04/2023] Open
Abstract
Nowadays, much attention has been paid to the link between gut microbiota and brain. The beneficial metabolic effects of probiotics and prebiotics in several diseases such as diabetes and obesity have been reported. However, studies bridging the association of gut microbiome with brain function in healthy states are rare. Therefore, it was hypothesized that the administration of Lactobacillus plantarum (L plantarum) and inulin may affect serum and hypothalamic metabolic parameters as well as oxidative markers in healthy male rats. Daily L plantarum (107 CFU/mL) and inulin (5% of daily food weight) or their combination (synbiotic) was given to healthy rats. Then, serum and hypothalamic levels of leptin, insulin, and oxidative markers were measured. Administration of synbiotic for 8 weeks led to significant changes in serum levels of high-density lipoprotein cholesterol, low-density lipoprotein cholesterol, very low-density lipoprotein cholesterol, low-density lipoprotein/high-density lipoprotein ratio, triglyceride, and total cholesterol. The intake of synbiotic also resulted in a significantly reduced hypothalamic level of malondialdehyde and increased hypothalamic superoxide dismutase (SOD). Also, L plantarum could significantly increase hypothalamic SOD level. Furthermore, synbiotic administration insignificantly increased the hypothalamic and serum levels of insulin and leptin. These findings suggest that the synbiotic could significantly improve oxidative markers and lipid profile in healthy rats. Therefore, simultaneous intake of L plantarum and inulin appears to be more effective in the amelioration of metabolic and oxidative parameters.
Collapse
Affiliation(s)
- Elaheh-Sadat Hosseinifard
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Maryam Saghafi-Asl
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Nutrition Research Center, School of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Clinical Nutrition, School of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Morshedi
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Clinical Nutrition, School of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
21
|
Zhou Y, Li P, Wang X, Wu C, Fan N, Liu X, Wu L, Zhang W, Zhang W, Liu Z, Tang B. In situ visualization of peroxisomal viscosity in the liver of mice with non-alcoholic fatty liver disease by near-infrared fluorescence and photoacoustic imaging. Chem Sci 2020; 11:12149-12156. [PMID: 34094429 PMCID: PMC8163019 DOI: 10.1039/d0sc02922j] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Accepted: 10/02/2020] [Indexed: 12/15/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) can gradually develop into hepatic failure, and early diagnosis is crucial to improve treatment efficiency. The occurrence of NAFLD is closely related to lipid metabolism. Peroxisomes act as the first and main site for lipid metabolism in the hepatocytes, so abnormal lipid metabolism might directly affect peroxisomal viscosity. Herein, we developed a new near-infrared fluorescence (NIRF) and photoacoustic (PA) imaging probe (PV-1) for the real-time visualization of peroxisomal viscosity in vivo. This PV-1 encompasses the malononitrile group as the rotor, which emits strong NIRF (at 705 nm) and PA (at 680 nm) signals when rotation is hindered as viscosity increases. Through dual-mode imaging, we discovered distinctly higher viscosity in the liver of NAFLD mice for the first time. We further found the remarkable amelioration of NAFLD upon treatment with N-acetylcysteine (NAC). Therefore, we anticipate that the PV-1 imaging method is promising for the early diagnosis and prognostic evaluation of NAFLD.
Collapse
Affiliation(s)
- Yongqing Zhou
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institutes of Biomedical Sciences, Shandong Normal University Jinan 250014 People's Republic of China
| | - Ping Li
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institutes of Biomedical Sciences, Shandong Normal University Jinan 250014 People's Republic of China
| | - Xin Wang
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institutes of Biomedical Sciences, Shandong Normal University Jinan 250014 People's Republic of China
| | - Chuanchen Wu
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institutes of Biomedical Sciences, Shandong Normal University Jinan 250014 People's Republic of China
| | - Nannan Fan
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institutes of Biomedical Sciences, Shandong Normal University Jinan 250014 People's Republic of China
| | - Xiaoning Liu
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institutes of Biomedical Sciences, Shandong Normal University Jinan 250014 People's Republic of China
| | - Lijie Wu
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institutes of Biomedical Sciences, Shandong Normal University Jinan 250014 People's Republic of China
| | - Wei Zhang
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institutes of Biomedical Sciences, Shandong Normal University Jinan 250014 People's Republic of China
| | - Wen Zhang
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institutes of Biomedical Sciences, Shandong Normal University Jinan 250014 People's Republic of China
| | - Zhenzhen Liu
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institutes of Biomedical Sciences, Shandong Normal University Jinan 250014 People's Republic of China
| | - Bo Tang
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institutes of Biomedical Sciences, Shandong Normal University Jinan 250014 People's Republic of China
| |
Collapse
|
22
|
Alam S, Abrar M, Islam S, Kamal M, Hasan MJ, Khan MAS, Ahmad N. Effect of telmisartan and vitamin E on liver histopathology with non-alcoholic steatohepatitis: A randomized, open-label, noninferiority trial. JGH Open 2020; 4:663-669. [PMID: 32782954 PMCID: PMC7411541 DOI: 10.1002/jgh3.12315] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 12/04/2019] [Accepted: 02/10/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND AND AIM To compare the effect of telmisartan and vitamin E on liver histopathology of non-alcoholic steatohepatitis (NASH) patients. METHODS This noninferiority clinical trial was conducted for 1 year. Fatty liver patients with non-alcoholic fatty liver disease (NAFLD) activity score (NAS) ≥ 5 (in liver biopsy) were selected. All methods were in accordance with the Declaration of Helsinki. Patients who received telmisartan and vitamin E were denoted as Group-T and Group-E, respectively. Forty patients >18 years old were assigned and divided into two groups (20 in each group). Histological improvements were primary outcome measures. RESULTS Significant improvement in NAS score was noted in both groups (Group E [GE]: 6 ± 0.8 to 4.36 ± 1.4; P = 0.00 and Group T [GT]: 5.6 ± 0.7to 4.9 ± 1.2; P = 0.03). Fibrosis score improved from 1.6 ± 0.5 to 1.5 ± 0.5 in GE and from 1.7 ± 0.9 to 1.5 ± 0.7 in GT (P = 0.67 and 0.42, respectively). Steatosis improved in GE from 2.07 ± 0.6 to 1.14 ± 0.66 (P = 0.00) and in GT from 1.94 ± 0.57 to 1.56 ± 0.8 (P = 0.05). Lobular inflammation improved from 2.0 ± 0.4 to 1.6 ± 0.5 in GE (P = 0.02) and from 1.9 ± 0.3 to 1.8 ± 0.4 in GT (P = 0.58). Ballooning score in GE decreased from 1.9 ± 0.3 to 1.7 ± 0.5 (P = 0.03), and in GT, it reduced from 1.9 ± 0.1 to 1.5 ± 0.5 (P = 0.19). NAS improvement was similar in GE (1.6 ± 1.2) and GT (0.6 ± 1.1; P = 0.07) when controlled for weight reduction. CONCLUSION Telmisartan was similar to vitamin E in improving the histology of NASH patients.
Collapse
Affiliation(s)
- Shahinul Alam
- Department of HepatologyBangabandhu Sheikh Mujib Medical UniversityDhakaBangladesh
| | - Mushfiqul Abrar
- Department of HepatologyBangabandhu Sheikh Mujib Medical UniversityDhakaBangladesh
| | - Saiful Islam
- Department of HepatologyBangabandhu Sheikh Mujib Medical UniversityDhakaBangladesh
| | - Mohammad Kamal
- Department of PathologyBangabandhu Sheikh Mujib Medical UniversityDhakaBangladesh
| | - Mohammad J Hasan
- Department of MedicinePi Research Consultancy CenterDhakaBangladesh
| | - Md. Abdullah S Khan
- Department of MedicineShaheed Sayed Nazrul Islam Medical CollegeKishoreganjBangladesh
| | - Nooruddin Ahmad
- Department of HepatologyBangabandhu Sheikh Mujib Medical UniversityDhakaBangladesh
| |
Collapse
|
23
|
An Overview of Lipid Metabolism and Nonalcoholic Fatty Liver Disease. BIOMED RESEARCH INTERNATIONAL 2020; 2020:4020249. [PMID: 32733940 PMCID: PMC7383338 DOI: 10.1155/2020/4020249] [Citation(s) in RCA: 101] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 06/14/2020] [Accepted: 06/25/2020] [Indexed: 12/11/2022]
Abstract
The occurrence of nonalcoholic fatty liver disease (NAFLD) is associated with major abnormalities of hepatic lipid metabolism. We propose that lipid abnormalities directly or indirectly contribute to NAFLD, especially fatty acid accumulation, arachidonic acid metabolic disturbance, and ceramide overload. The effects of lipid intake and accumulation on NAFLD and NAFLD treatment are explained with theoretical and experimental details. Overall, these findings provide further understanding of lipid metabolism in NAFLD and may lead to novel therapies.
Collapse
|
24
|
|
25
|
The PPAR α/γ Agonist Saroglitazar Improves Insulin Resistance and Steatohepatitis in a Diet Induced Animal Model of Nonalcoholic Fatty Liver Disease. Sci Rep 2020; 10:9330. [PMID: 32518275 PMCID: PMC7283326 DOI: 10.1038/s41598-020-66458-z] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2019] [Accepted: 05/19/2020] [Indexed: 12/15/2022] Open
Abstract
Insulin resistance and hepatic lipid accumulation constitute the metabolic underpinning of nonalcoholic steatohepatitis (NASH). We tested the hypothesis that saroglitazar, a PPAR α/γ agonist would improve NASH in the diet-induced animal model of NAFLD. Mice received chow diet and normal water (CDNW) or high fat western diet and ad lib sugar water (WDSW). After 12 weeks, WDSW fed mice were randomized to receive (1) WDSW alone, (2) WDSW + vehicle, (3) WDSW + pioglitazone or (4) WDSW + saroglitazar for an additional 12 weeks. Compared to mice on WDSW and vehicle controls, mice receiving WDSW + saroglitazar had lower weight, lower HOMA-IR, triglycerides, total cholesterol, and ALT. Saroglitazar improved steatosis, lobular inflammation, hepatocellular ballooning and fibrosis stage. NASH resolved in all mice receiving saroglitazar. These effects were at par with or superior to pioglitazone. Molecular analyses confirmed target engagement and reduced oxidative stress, unfolded protein response and fibrogenic signaling. Transcriptomic analysis further confirmed increased PPAR-target expression and an anti-inflammatory effect with saroglitazar. Lipidomic analyses demonstrated that saroglitazar also reduced triglycerides, diglycerides, sphingomyelins and ceramides. These preclinical data provide a strong rationale for developing saroglitazar for the treatment of NASH in humans.
Collapse
|
26
|
Petta S, Ting J, Saragoni S, Degli Esposti L, Shreay S, Petroni ML, Marchesini G. Healthcare resource utilization and costs of nonalcoholic steatohepatitis patients with advanced liver disease in Italy. Nutr Metab Cardiovasc Dis 2020; 30:1014-1022. [PMID: 32423665 DOI: 10.1016/j.numecd.2020.02.016] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 02/21/2020] [Accepted: 02/24/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND AND AIMS Nonalcoholic steatohepatitis (NASH) may progress to advanced liver disease (AdvLD). This study characterized comorbidities, healthcare resource utilization (HCRU) and associated costs among hospitalized patients with AdvLD due to NASH in Italy. METHODS AND RESULTS Adult nonalcoholic fatty liver disease (NAFLD)/NASH patients from 2011 to 2017 were identified from administrative databases of Italian local health units using ICD-9-CM codes. Development of compensated cirrhosis (CC), decompensated cirrhosis (DCC), hepatocellular carcinoma (HCC), or liver transplant (LT) was identified using first diagnosis date for each severity cohort (index-date). Patients progressing to multiple disease stages were included in >1 cohort. Patients were followed from index-date until the earliest of disease progression, end of coverage, death, or end of study. Within each cohort, per member per month values were annualized to calculate all-cause HCRU or costs(€) in 2017. Of the 9,729 hospitalized NAFLD/NASH patients identified, 97% were without AdvLD, 1.3% had CC, 3.1% DCC, 0.8% HCC, 0.1% LT. Comorbidity burden was high across all cohorts. Mean annual number of inpatient services was greater in patients with AdvLD than without AdvLD. Similar trends were observed in outpatient visits and pharmacy fills. Mean total annual costs increased with disease severity, driven primarily by inpatient services costs. CONCLUSION NAFLD/NASH patients in Italy have high comorbidity burden. AdvLD patients had significantly higher costs. The higher prevalence of DCC compared to CC in this population may suggest challenges of effectively screening and identifying NAFLD/NASH patients. Early identification and effective management are needed to reduce risk of disease progression and subsequent HCRU and costs.
Collapse
Affiliation(s)
- Salvatore Petta
- Section of Gastroenterology and Hepatology, PROMISE, University of Palermo, Palermo, Italy.
| | - Jie Ting
- Gilead Sciences, Inc., Health Economics & Outcomes Research, Foster City, CA, USA
| | | | | | - Sanatan Shreay
- Gilead Sciences, Inc., Health Economics & Outcomes Research, Foster City, CA, USA
| | - Maria Letizia Petroni
- Department of Medical and Surgical Sciences, "Alma Mater" University, Bologna, Italy
| | - Giulio Marchesini
- Department of Medical and Surgical Sciences, "Alma Mater" University, Bologna, Italy
| |
Collapse
|
27
|
Vacca M, Leslie J, Virtue S, Lam BYH, Govaere O, Tiniakos D, Snow S, Davies S, Petkevicius K, Tong Z, Peirce V, Nielsen MJ, Ament Z, Li W, Kostrzewski T, Leeming DJ, Ratziu V, Allison MED, Anstee QM, Griffin JL, Oakley F, Vidal-Puig A. Bone morphogenetic protein 8B promotes the progression of non-alcoholic steatohepatitis. Nat Metab 2020; 2:514-531. [PMID: 32694734 PMCID: PMC7617436 DOI: 10.1038/s42255-020-0214-9] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Accepted: 04/24/2020] [Indexed: 12/15/2022]
Abstract
Non-alcoholic steatohepatitis (NASH) is characterized by lipotoxicity, inflammation and fibrosis, ultimately leading to end-stage liver disease. The molecular mechanisms promoting NASH are poorly understood, and treatment options are limited. Here, we demonstrate that hepatic expression of bone morphogenetic protein 8B (BMP8B), a member of the transforming growth factor beta (TGFβ)-BMP superfamily, increases proportionally to disease stage in people and animal models with NASH. BMP8B signals via both SMAD2/3 and SMAD1/5/9 branches of the TGFβ-BMP pathway in hepatic stellate cells (HSCs), promoting their proinflammatory phenotype. In vivo, the absence of BMP8B prevents HSC activation, reduces inflammation and affects the wound-healing responses, thereby limiting NASH progression. Evidence is featured in primary human 3D microtissues modelling NASH, when challenged with recombinant BMP8. Our data show that BMP8B is a major contributor to NASH progression. Owing to the near absence of BMP8B in healthy livers, inhibition of BMP8B may represent a promising new therapeutic avenue for NASH treatment.
Collapse
Affiliation(s)
- Michele Vacca
- TVP Lab, WT/MRC Institute of Metabolic Science, MRC Metabolic Diseases Unit - Metabolic Research Laboratories, University of Cambridge, Cambridge, UK.
- Department of Biochemistry and Cambridge Systems Biology Centre, University of Cambridge, Cambridge, UK.
| | - Jack Leslie
- Newcastle Fibrosis Research Group, Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Samuel Virtue
- TVP Lab, WT/MRC Institute of Metabolic Science, MRC Metabolic Diseases Unit - Metabolic Research Laboratories, University of Cambridge, Cambridge, UK
| | - Brian Y H Lam
- Yeo Group and Genomics and Transcriptomics Core, WT/MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Olivier Govaere
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Dina Tiniakos
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
- Department of Pathology, Aretaieion Hospital, Medical School, National & Kapodistrian University of Athens, Athens, Greece
| | | | - Susan Davies
- Liver Unit, Department of Medicine, Cambridge Biomedical Research Centre, Cambridge University Hospitals, Cambridge, UK
| | - Kasparas Petkevicius
- TVP Lab, WT/MRC Institute of Metabolic Science, MRC Metabolic Diseases Unit - Metabolic Research Laboratories, University of Cambridge, Cambridge, UK
| | - Zhen Tong
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - Vivian Peirce
- TVP Lab, WT/MRC Institute of Metabolic Science, MRC Metabolic Diseases Unit - Metabolic Research Laboratories, University of Cambridge, Cambridge, UK
| | | | - Zsuzsanna Ament
- Department of Biochemistry and Cambridge Systems Biology Centre, University of Cambridge, Cambridge, UK
| | - Wei Li
- Department of Medicine, University of Cambridge, Cambridge, UK
| | | | | | - Vlad Ratziu
- Sorbonne Université, Institute for Cardiometabolism and Nutrition (ICAN), Hôpital Pitié-Salpêtrière, Paris, France
| | - Michael E D Allison
- Liver Unit, Department of Medicine, Cambridge Biomedical Research Centre, Cambridge University Hospitals, Cambridge, UK
| | - Quentin M Anstee
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
- Newcastle NIHR Biomedical Research Centre, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Julian L Griffin
- Department of Biochemistry and Cambridge Systems Biology Centre, University of Cambridge, Cambridge, UK
- Biomolecular Medicine, Systems Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Fiona Oakley
- Newcastle Fibrosis Research Group, Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Antonio Vidal-Puig
- TVP Lab, WT/MRC Institute of Metabolic Science, MRC Metabolic Diseases Unit - Metabolic Research Laboratories, University of Cambridge, Cambridge, UK.
- Welcome Trust Sanger Institute, Hinxton, UK.
- Cambridge University Nanjing Centre of Technology and Innovation, Jiangbei Area, Nanjing, P R China.
| |
Collapse
|
28
|
BEKTAS A, ULUSOY M, DAĞALP K. URSODEOKSİKOLİK ASİT, DİYABETİK OLMAYAN, ALKOL DIŞI KARACİĞER YAĞLANMASI TEDAVİSİNDE YER ALMALI MI? KONURALP TIP DERGISI 2020. [DOI: 10.18521/ktd.527978] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
29
|
De Chiara F, Thomsen KL, Habtesion A, Jones H, Davies N, Gracia-Sancho J, Manicardi N, Hall A, Andreola F, Paish HL, Reed LH, Watson AA, Leslie J, Oakley F, Rombouts K, Mookerjee RP, Mann J, Jalan R. Ammonia Scavenging Prevents Progression of Fibrosis in Experimental Nonalcoholic Fatty Liver Disease. Hepatology 2020; 71:874-892. [PMID: 31378982 DOI: 10.1002/hep.30890] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2019] [Accepted: 07/07/2019] [Indexed: 01/10/2023]
Abstract
BACKGROUND AND AIMS In nonalcoholic fatty liver disease (NAFLD), fibrosis is the most important factor contributing to NAFLD-associated morbidity and mortality. Prevention of progression and reduction in fibrosis are the main aims of treatment. Even in early stages of NAFLD, hepatic and systemic hyperammonemia is evident. This is due to reduced urea synthesis; and as ammonia is known to activate hepatic stellate cells, we hypothesized that ammonia may be involved in the progression of fibrosis in NAFLD. APPROACH AND RESULTS In a high-fat, high-cholesterol diet-induced rodent model of NAFLD, we observed a progressive stepwise reduction in the expression and activity of urea cycle enzymes resulting in hyperammonemia, evidence of hepatic stellate cell activation, and progressive fibrosis. In primary, cultured hepatocytes and precision-cut liver slices we demonstrated increased gene expression of profibrogenic markers after lipid and/or ammonia exposure. Lowering of ammonia with the ammonia scavenger ornithine phenylacetate prevented hepatocyte cell death and significantly reduced the development of fibrosis both in vitro in the liver slices and in vivo in a rodent model. The prevention of fibrosis in the rodent model was associated with restoration of urea cycle enzyme activity and function, reduced hepatic ammonia, and markers of inflammation. CONCLUSIONS The results of this study suggest that hepatic steatosis results in hyperammonemia, which is associated with progression of hepatic fibrosis. Reduction of ammonia levels prevented progression of fibrosis, providing a potential treatment for NAFLD.
Collapse
Affiliation(s)
- Francesco De Chiara
- UCL Institute of Liver and Digestive Health, University College London, London, UK
| | - Karen Louise Thomsen
- UCL Institute of Liver and Digestive Health, University College London, London, UK.,Department of Hepatology & Gastroenterology, Aarhus University Hospital, Aarhus, Denmark
| | - Abeba Habtesion
- UCL Institute of Liver and Digestive Health, University College London, London, UK
| | - Helen Jones
- UCL Institute of Liver and Digestive Health, University College London, London, UK
| | - Nathan Davies
- UCL Institute of Liver and Digestive Health, University College London, London, UK
| | - Jordi Gracia-Sancho
- Liver Vascular Biology Research Group, IDIBAPS Biomedical Research Institute & CIBEREHD, Barcelona, Spain
| | - Nicolò Manicardi
- Liver Vascular Biology Research Group, IDIBAPS Biomedical Research Institute & CIBEREHD, Barcelona, Spain
| | - Andrew Hall
- UCL Institute of Liver and Digestive Health, University College London, London, UK
| | - Fausto Andreola
- UCL Institute of Liver and Digestive Health, University College London, London, UK
| | - Hannah L Paish
- Newcastle Fibrosis Research Group, Institute of Cellular Medicine, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Lee H Reed
- Newcastle Fibrosis Research Group, Institute of Cellular Medicine, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Abigail A Watson
- Newcastle Fibrosis Research Group, Institute of Cellular Medicine, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Jack Leslie
- Newcastle Fibrosis Research Group, Institute of Cellular Medicine, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Fiona Oakley
- Newcastle Fibrosis Research Group, Institute of Cellular Medicine, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Krista Rombouts
- UCL Institute of Liver and Digestive Health, University College London, London, UK
| | | | - Jelena Mann
- Newcastle Fibrosis Research Group, Institute of Cellular Medicine, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Rajiv Jalan
- UCL Institute of Liver and Digestive Health, University College London, London, UK
| |
Collapse
|
30
|
Violet PC, Ebenuwa IC, Wang Y, Niyyati M, Padayatty SJ, Head B, Wilkins K, Chung S, Thakur V, Ulatowski L, Atkinson J, Ghelfi M, Smith S, Tu H, Bobe G, Liu CY, Herion DW, Shamburek RD, Manor D, Traber MG, Levine M. Vitamin E sequestration by liver fat in humans. JCI Insight 2020; 5:133309. [PMID: 31821172 PMCID: PMC7030816 DOI: 10.1172/jci.insight.133309] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 11/26/2019] [Indexed: 12/27/2022] Open
Abstract
BACKGROUNDWe hypothesized that obesity-associated hepatosteatosis is a pathophysiological chemical depot for fat-soluble vitamins and altered normal physiology. Using α-tocopherol (vitamin E) as a model vitamin, pharmacokinetics and kinetics principles were used to determine whether excess liver fat sequestered α-tocopherol in women with obesity-associated hepatosteatosis versus healthy controls.METHODSCustom-synthesized deuterated α-tocopherols (d3- and d6-α-tocopherols) were administered to hospitalized healthy women and women with hepatosteatosis under investigational new drug guidelines. Fluorescently labeled α-tocopherol was custom-synthesized for cell studies.RESULTSIn healthy subjects, 85% of intravenous d6-α-tocopherol disappeared from the circulation within 20 minutes but reappeared within minutes and peaked at 3-4 hours; d3- and d6-α-tocopherols localized to lipoproteins. Lipoprotein redistribution occurred only in vivo within 1 hour, indicating a key role of the liver in uptake and re-release. Compared with healthy subjects who received 2 mg, subjects with hepatosteatosis had similar d6-α-tocopherol entry rates into liver but reduced initial release rates (P < 0.001). Similarly, pharmacokinetics parameters were reduced in hepatosteatosis subjects, indicating reduced hepatic d6-α-tocopherol output. Reductions in kinetics and pharmacokinetics parameters in hepatosteatosis subjects who received 2 mg were echoed by similar reductions in healthy subjects when comparing 5- and 2-mg doses. In vitro, fluorescent-labeled α-tocopherol localized to lipid in fat-loaded hepatocytes, indicating sequestration.CONCLUSIONSThe unique role of the liver in vitamin E physiology is dysregulated by excess liver fat. Obesity-associated hepatosteatosis may produce unrecognized hepatic vitamin E sequestration, which might subsequently drive liver disease. Our findings raise the possibility that hepatosteatosis may similarly alter hepatic physiology of other fat-soluble vitamins.TRIAL REGISTRATIONClinicalTrials.gov, NCT00862433.FUNDINGNational Institute of Diabetes and Digestive and Kidney Diseases and NIH grants DK053213-13, DK067494, and DK081761.
Collapse
Affiliation(s)
- Pierre-Christian Violet
- Molecular and Clinical Nutrition Section, Intramural Research Program, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, Maryland, USA
| | - Ifechukwude C. Ebenuwa
- Molecular and Clinical Nutrition Section, Intramural Research Program, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, Maryland, USA
| | - Yu Wang
- Molecular and Clinical Nutrition Section, Intramural Research Program, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, Maryland, USA
| | - Mahtab Niyyati
- Molecular and Clinical Nutrition Section, Intramural Research Program, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, Maryland, USA
| | - Sebastian J. Padayatty
- Molecular and Clinical Nutrition Section, Intramural Research Program, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, Maryland, USA
| | - Brian Head
- Linus Pauling Institute, Oregon State University, Corvallis, Oregon, USA
| | - Kenneth Wilkins
- Office of the Director, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, Maryland, USA
| | - Stacey Chung
- Department of Pharmacology and Department of Nutrition, School of Medicine, Case Western Reserve University and the Case Comprehensive Cancer Center, Cleveland, Ohio, USA
| | - Varsha Thakur
- Department of Pharmacology and Department of Nutrition, School of Medicine, Case Western Reserve University and the Case Comprehensive Cancer Center, Cleveland, Ohio, USA
| | - Lynn Ulatowski
- Department of Pharmacology and Department of Nutrition, School of Medicine, Case Western Reserve University and the Case Comprehensive Cancer Center, Cleveland, Ohio, USA
| | - Jeffrey Atkinson
- Department of Chemistry, Brock University, Saint Catharines, Ontario, Canada
| | - Mikel Ghelfi
- Department of Chemistry, Brock University, Saint Catharines, Ontario, Canada
| | - Sheila Smith
- Molecular and Clinical Nutrition Section, Intramural Research Program, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, Maryland, USA
| | - Hongbin Tu
- Molecular and Clinical Nutrition Section, Intramural Research Program, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, Maryland, USA
| | - Gerd Bobe
- Linus Pauling Institute, Oregon State University, Corvallis, Oregon, USA
| | | | - David W. Herion
- Clinical Research Informatics, Clinical Center, NIH, Bethesda, Maryland, USA
| | - Robert D. Shamburek
- Cardiovascular Branch, Intramural Research Program, National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland, USA
| | - Danny Manor
- Department of Pharmacology and Department of Nutrition, School of Medicine, Case Western Reserve University and the Case Comprehensive Cancer Center, Cleveland, Ohio, USA
| | - Maret G. Traber
- Linus Pauling Institute, Oregon State University, Corvallis, Oregon, USA
| | - Mark Levine
- Molecular and Clinical Nutrition Section, Intramural Research Program, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, Maryland, USA
| |
Collapse
|
31
|
Chen YC, Chen HJ, Huang BM, Chen YC, Chang CF. Polyphenol-Rich Extracts from Toona sinensis Bark and Fruit Ameliorate Free Fatty Acid-Induced Lipogenesis through AMPK and LC3 Pathways. J Clin Med 2019; 8:E1664. [PMID: 31614650 PMCID: PMC6832244 DOI: 10.3390/jcm8101664] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 09/26/2019] [Accepted: 10/03/2019] [Indexed: 01/18/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a chronic liver disease found worldwide. The present study aimed to evaluate the mechanisms of inhibiting lipid accumulation in free fatty acid (FFA)-treated HepG2 cells caused by bark and fruit extracts of Toona sinensis (TSB and TSF). FFA induced lipid and triglyceride (TG) accumulation, which was attenuated by TSB and TSF. TSB and/or TSF promoted phosphorylation of AMP-activated protein kinase (AMPK) and acetyl-coA carboxylase and peroxisome proliferator-activated receptor alpha upregulation. Furthermore, TSB and TSF suppressed FFA-induced liver X receptor, sterol regulatory element-binding transcription protein 1, fatty acid synthase, and stearoyl-CoA desaturase 1 protein expression. Moreover, TSB and/or TSF induced phosphorylation of Unc-51 like autophagy-activating kinase and microtubule-associated protein 1A/1B-light chain 3 expressions. Therefore, TSB and TSF relieve lipid accumulation by attenuating lipogenic protein expression, activating the AMPK pathway, and upregulating the autophagic flux to enhance lipid metabolism. Moreover, TSB and TSF reduced TG contents, implying the therapeutic use of TSB and TSF in NAFLD.
Collapse
Affiliation(s)
- Yung-Chia Chen
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan.
- Department of Anatomy, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan.
| | - Hsin-Ju Chen
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan.
- Department of Anatomy, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan.
| | - Bu-Miin Huang
- Department of Anatomy, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan.
| | - Yu-Chi Chen
- Department of Urology, E-Da Hospital, Kaohsiung 82445, Taiwan.
- Department of Urology, E-Da Cancer Hospital, Kaohsiung 40402, Taiwan.
| | - Chi-Fen Chang
- Department of Anatomy, School of Medicine, China Medical University, Taichung 40401, Taiwan.
| |
Collapse
|
32
|
Alam S, Jahid Hasan M, Khan MAS, Alam M, Hasan N. Effect of Weight Reduction on Histological Activity and Fibrosis of Lean Nonalcoholic Steatohepatitis Patient. J Transl Int Med 2019; 7:106-114. [PMID: 31637181 PMCID: PMC6795050 DOI: 10.2478/jtim-2019-0023] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND AND OBJECTIVES Weight reduction has evidenced benefit on attenuation of histological activity and fibrosis of nonalcoholic steatohepatitis (NASH), but there is scarcity of data for lean NASH subgroup. We have designed this study to compare the effects of weight reduction on histological activity and fibrosis of lean and non-lean NASH. METHODS We have included 20 lean and 20 non-lean histologically proven NASH patients. BMI < 25 kg/m2 was defined as non-lean. Informed consent was taken from each subject. All methods were carried out in accordance with the Declaration of Helsinki. Moderate exercise along with dietary restriction was advised for both groups for weight reduction. After 1 year, 16 non-lean and 15 lean had completed second liver biopsy. RESULTS Age, sex, alanine transaminase (ALT), aspartate aminotransferase (AST), gamma-glutamyltrasferase (GGT), Homeostasis model assessment insulin resistance (HOMA-IR), triglyceride and high density lipoprotein (HDL) was similar in both groups. Steatosis, ballooning, lobular inflammation, nonalcoholic fatty liver disease activity score (NAS) and fibrosis was similar in the two groups. In lean/non-lean group, any amount of weight reduction, ≥ 5% weight reduction and ≥ 7% weight reduction was found in respectively 8/11, 5/6 and 2/6 patients. In both lean and non-lean groups, weight reduction of any amount was associated with significant reduction of steatosis, ballooning and NAS, except lobular inflammation and fibrosis. In both groups, weight reduction of ≥ 5% was associated with significant reduction in NAS only. However, significant improvement in NAS was noted with ≥ 7% weight reduction in non-lean group only. CONCLUSION Smaller amount of weight reduction had the good benefit of improvement in all the segments of histological activity in both lean and non-lean NASH.
Collapse
Affiliation(s)
- Shahinul Alam
- Department of Hepatology, Bangabandhu Sheikh Mujib Medical University, Shahbag, Dhaka, Bangladesh
| | - Mohammad Jahid Hasan
- Department of Medicine, Dr. Sirajul Islam Medical College & Hospital, Dhaka, Bangladesh
| | | | - Mahabubul Alam
- Department of Hepatology, Bangabandhu Sheikh Mujib Medical University, Shahbag, Dhaka, Bangladesh
| | - Nazmul Hasan
- Department of Hepatology, Bangabandhu Sheikh Mujib Medical University, Shahbag, Dhaka, Bangladesh
| |
Collapse
|
33
|
Houghton D, Wilcox MD, Brownlee IA, Chater PI, Seal CJ, Pearson JP. Acceptability of alginate enriched bread and its effect on fat digestion in humans. Food Hydrocoll 2019; 93:395-401. [PMID: 32226189 PMCID: PMC7086458 DOI: 10.1016/j.foodhyd.2019.02.027] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Lifestyle interventions and physical activity remain the cornerstone of obesity management, as pharmacological therapies (orlistat) are associated with gastrointestinal (GI) side effects. Combining orlistat with fibers can reduce side effects, improving compliance. Therefore, a fiber that inhibits lipase without side effects could help treat obesity. The aims of the present work were to assess whether alginate enriched bread could inhibit fat digestion, and assess the acceptability of alginate bread and its effect on GI wellbeing. A double-blind, randomised, controlled cross-over pilot study (NCT03350958) assessed the impact of an alginate bread meal on; lipid content in ileal effluent and circulating triacylglycerol levels. This was compared against the same meal with non-enriched (control) bread. GI wellbeing and acceptability of alginate bread was compared to control bread through daily wellbeing questionnaires and food diaries (NCT03477981). Control bread followed by alginate bread were consumed for two weeks respectively. Consumption of alginate bread reduced circulating triacylglycerol compared to control (2% reduction in AUC) and significantly increased lipid content in ileal effluent (3.8 g ± 1.6 after 210 min). There were no significant changes to GI wellbeing when comparing alginate bread to control bread. A significant increase in the feeling of fullness occurred with alginate bread compared to baseline and the first week of control bread consumption. This study showed that sustained consumption of alginate enriched bread does not alter GI wellbeing and can decrease lipolysis, increasing lipid leaving the small intestine. Further studies are required to demonstrate that reduced fat digestion through the action of alginate can reduce fat mass or body weight. Alginate can be incorporated into a highly acceptable loaf at 4%. Sustained (two weeks) consumption of alginate bread did not affect GI wellbeing. Consumption of alginate bread decreases circulating triglyceride after the meal. Consumption of alginate bread increases lipid leaving the ileum after the meal.
Collapse
Affiliation(s)
- David Houghton
- Institute for Cell and Molecular Biosciences, Newcastle University, Medical School, Framlington Place, Newcastle upon Tyne, UK
| | - Matthew D Wilcox
- Institute for Cell and Molecular Biosciences, Newcastle University, Medical School, Framlington Place, Newcastle upon Tyne, UK
| | - Iain A Brownlee
- Institute for Cell and Molecular Biosciences, Newcastle University, Medical School, Framlington Place, Newcastle upon Tyne, UK
| | - Peter I Chater
- Institute for Cell and Molecular Biosciences, Newcastle University, Medical School, Framlington Place, Newcastle upon Tyne, UK
| | - Chris J Seal
- Human Nutrition Research Centre, Institute of Cellular Medicine, Newcastle University, M2.054 Leech Building, Newcastle upon Tyne, NE2 4HH, UK
| | - Jeffrey P Pearson
- Institute for Cell and Molecular Biosciences, Newcastle University, Medical School, Framlington Place, Newcastle upon Tyne, UK
| |
Collapse
|
34
|
Povsic M, Wong OY, Perry R, Bottomley J. A Structured Literature Review of the Epidemiology and Disease Burden of Non-Alcoholic Steatohepatitis (NASH). Adv Ther 2019; 36:1574-1594. [PMID: 31065991 PMCID: PMC6824389 DOI: 10.1007/s12325-019-00960-3] [Citation(s) in RCA: 105] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Non-Alcoholic Steatohepatitis (NASH) is a chronic, progressive disease characterized by fatty liver and liver cell injury, advancing to fibrosis, cirrhosis and hepatocellular carcinoma (HCC). Diagnosis involves liver biopsy; however, as a result of its high cost and invasiveness, NASH remains underdiagnosed, and accurate burden of disease (BoD) data are lacking. Our aim was to understand the epidemiological and BoD landscape in NASH and identify knowledge gaps. METHODS The Ovid search engine was used to conduct a structured review, following quality systematic principles. It included publications that reported on epidemiology, quality of life (QoL) and BoD outcomes in NASH adults. Searches were limited to English language studies published between January 2007 and September 2017. Additional grey literature searches were conducted. A total of 53 references were selected; 38 were peer-reviewed and 15 were grey literature sources. RESULTS NASH is estimated to affect 3-5% of the global population, most suffering from several comorbidities. Advancing fibrosis drives clinical outcomes, with approximately 20% of patients developing cirrhosis and/or HCC, the latter being a leading cause of death in NASH. A recent model predicted the 15-year survival of advanced fibrosis patients at F3 and F4 as 51.0% and 28.4%, respectively. The limited data consistently show that NASH patients experience significantly poorer QoL and higher costs compared to non-NASH patients. CONCLUSION This first broad-ranging examination of NASH literature revealed a paucity of evidence, with poor-quality, small studies found. The overwhelming impact of NASH and its patient and healthcare burden is evident. Further evidence is needed to improve our understanding of NASH, especially as fibrosis stages advance. FUNDING Gilead Science Inc.
Collapse
|
35
|
Mansour A, Hekmatdoost A, Mirmiran P. What are the main areas of focus to prevent or treat non-alcoholic fatty liver disease? J Dig Dis 2019; 20:271-277. [PMID: 30968561 DOI: 10.1111/1751-2980.12751] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 02/26/2019] [Accepted: 04/07/2019] [Indexed: 12/11/2022]
Abstract
Recently, a growing body of information has accumulated to suggest that nutritional status and food compounds impact on the development or progression of non-alcoholic fatty liver disease (NAFLD). The best strategy to prevent and treat NAFLD is to modify diet and lifestyle by maintaining a healthy weight, following a well-balanced diet with appropriate energy intake and increasing physical activity or strength training. Here we review the literatures and discuss existing and potential therapeutic strategies for the prevention and management of NAFLD, emphasizing the description of nutritional status and its clinical impact on the outcomes of NAFLD.
Collapse
Affiliation(s)
- Asieh Mansour
- Student Research Committee, Faculty of Nutrition Sciences and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Department of Clinical Nutrition and Dietetics, Faculty of Nutrition Sciences and Food Technology, National Nutrition and Food Technology, Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Azita Hekmatdoost
- Department of Clinical Nutrition and Dietetics, Faculty of Nutrition Sciences and Food Technology, National Nutrition and Food Technology, Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Parvin Mirmiran
- Department of Clinical Nutrition and Dietetics, Faculty of Nutrition Sciences and Food Technology, National Nutrition and Food Technology, Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
36
|
Povsic M, Oliver L, Jiandani NR, Perry R, Bottomley J. A structured literature review of interventions used in the management of nonalcoholic steatohepatitis (NASH). Pharmacol Res Perspect 2019; 7:e00485. [PMID: 31149341 PMCID: PMC6536401 DOI: 10.1002/prp2.485] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 04/18/2019] [Accepted: 04/26/2019] [Indexed: 12/18/2022] Open
Abstract
Nonalcoholic steatohepatitis (NASH) is a chronic, progressive disease, that can advance to fibrosis, cirrhosis, and hepatocellular carcinoma. Despite being a leading cause of liver transplantation, there are no approved pharmacological treatments. Our aim was to identify literature on management options in NASH. Our structured review of interventions treating NASH patients from English language publications between 1 January 2007 and 25 September 2017 elicited 48 eligible references. Lifestyle management was identified as the mainstay of NASH therapy. Vitamin E and pioglitazone reported reductions in steatosis; however, although recommended for some, no therapies are indicated in NASH. Multiple investigational treatments reported efficacy in mild-to-moderate fibrosis in Phase II/III NASH trials. Lifestyle management, although the focus of clinical guidelines, is insufficient for patients progressing to advanced fibrosis. With no clear guidelines for patients requiring interventions beyond lifestyle modification, long-term outcomes data are needed, particularly in patients with moderate-to-severe fibrosis.
Collapse
|
37
|
Oh S, Oshida N, Someya N, Maruyama T, Isobe T, Okamoto Y, Kim T, Kim B, Shoda J. Whole-body vibration for patients with nonalcoholic fatty liver disease: a 6-month prospective study. Physiol Rep 2019; 7:e14062. [PMID: 31087530 PMCID: PMC6513769 DOI: 10.14814/phy2.14062] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 03/13/2019] [Accepted: 03/16/2019] [Indexed: 01/14/2023] Open
Abstract
Physical exercise has demonstrated benefits for managing nonalcoholic fatty liver disease (NAFLD). However, in daily life maintaining exercise without help may be difficult. A whole-body vibration device (WBV) has been recently introduced as an exercise modality that may be suitable for patients who have difficulty engaging in exercise. We tested WBV in patients with NAFLD and estimated its effectiveness. We studied the effects of a 6-month WBV program on hepatic steatosis and its underlying pathophysiology in 25 patients with NAFLD. Seventeen patients with NAFLD were designated as a control group. After WBV exercise, body weight in the study group decreased by only 2.5% compared with the control group. However, we found significant increases in muscle area (+2.6%) and strength (+20.5%) and decreases in fat mass (-6.8%). The hepatic (-9.9%) and visceral (-6.2%) fat content also significantly decreased (P < 0.05). There was substantial lowering of hepatic stiffness (-15.7%), along with improvements in the levels of inflammatory markers; tumor necrosis factor alpha (-50.9%), adiponectin (+12.0%), ferritin (-33.2%), and high-sensitivity C-reactive protein (-43.0%) (P < 0.05). These results suggest that WBV is an exercise option for patients with NAFLD that is effective, efficient, and convenient.
Collapse
Affiliation(s)
- Sechang Oh
- The Center of Sports Medicine and Health SciencesTsukuba University HospitalTsukubaIbarakiJapan
- Faculty of MedicineUniversity of TsukubaTsukubaIbarakiJapan
| | - Natsumi Oshida
- The Center of Sports Medicine and Health SciencesTsukuba University HospitalTsukubaIbarakiJapan
- Graduate School of Comprehensive Human SciencesUniversity of TsukubaTsukubaIbarakiJapan
| | - Noriko Someya
- Graduate School of Comprehensive Human SciencesUniversity of TsukubaTsukubaIbarakiJapan
| | - Tsuyoshi Maruyama
- Department of RehabilitationUniversity of Tsukuba HospitalTsukubaIbarakiJapan
| | - Tomonori Isobe
- Faculty of MedicineUniversity of TsukubaTsukubaIbarakiJapan
- Department of Diagnostic RadiologyUniversity of Tsukuba HospitalTsukubaIbarakiJapan
| | - Yoshikazu Okamoto
- Faculty of MedicineUniversity of TsukubaTsukubaIbarakiJapan
- Department of Diagnostic RadiologyUniversity of Tsukuba HospitalTsukubaIbarakiJapan
| | - Taeho Kim
- The Center of Sports Medicine and Health SciencesTsukuba University HospitalTsukubaIbarakiJapan
| | - Bokun Kim
- Faculty of Sports Health CareInje UniversityGimhaeGyeongsangnamdoRepublic of Korea
| | - Junichi Shoda
- The Center of Sports Medicine and Health SciencesTsukuba University HospitalTsukubaIbarakiJapan
- Faculty of MedicineUniversity of TsukubaTsukubaIbarakiJapan
| |
Collapse
|
38
|
James G, Reisberg S, Lepik K, Galwey N, Avillach P, Kolberg L, Mägi R, Esko T, Alexander M, Waterworth D, Loomis AK, Vilo J. An exploratory phenome wide association study linking asthma and liver disease genetic variants to electronic health records from the Estonian Biobank. PLoS One 2019; 14:e0215026. [PMID: 30978214 PMCID: PMC6461350 DOI: 10.1371/journal.pone.0215026] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Accepted: 03/25/2019] [Indexed: 12/22/2022] Open
Abstract
The Estonian Biobank, governed by the Institute of Genomics at the University of Tartu (Biobank), has stored genetic material/DNA and continuously collected data since 2002 on a total of 52,274 individuals representing ~5% of the Estonian adult population and is increasing. To explore the utility of data available in the Biobank, we conducted a phenome-wide association study (PheWAS) in two areas of interest to healthcare researchers; asthma and liver disease. We used 11 asthma and 13 liver disease-associated single nucleotide polymorphisms (SNPs), identified from published genome-wide association studies, to test our ability to detect established associations. We confirmed 2 asthma and 5 liver disease associated variants at nominal significance and directionally consistent with published results. We found 2 associations that were opposite to what was published before (rs4374383:AA increases risk of NASH/NAFLD, rs11597086 increases ALT level). Three SNP-diagnosis pairs passed the phenome-wide significance threshold: rs9273349 and E06 (thyroiditis, p = 5.50x10-8); rs9273349 and E10 (type-1 diabetes, p = 2.60x10-7); and rs2281135 and K76 (non-alcoholic liver diseases, including NAFLD, p = 4.10x10-7). We have validated our approach and confirmed the quality of the data for these conditions. Importantly, we demonstrate that the extensive amount of genetic and medical information from the Estonian Biobank can be successfully utilized for scientific research.
Collapse
Affiliation(s)
- Glen James
- AstraZeneca, Global Medical Affairs, Cambridge, United Kingdom
| | - Sulev Reisberg
- Institute of Computer Science, University of Tartu, Tartu, Estonia
- STACC, Tartu, Estonia
- Quretec, Tartu, Estonia
| | - Kaido Lepik
- Institute of Computer Science, University of Tartu, Tartu, Estonia
| | - Nicholas Galwey
- GlaxoSmithKline, Research and Development, Stevenage, United Kingdom
| | - Paul Avillach
- Department of Biomedical Informatics, Harvard Medical School, Boston, United States of America
- Department of Medical Informatics, Erasmus University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Liis Kolberg
- Institute of Computer Science, University of Tartu, Tartu, Estonia
| | - Reedik Mägi
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Tõnu Esko
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Myriam Alexander
- GlaxoSmithKline, Research and Development, Stevenage, United Kingdom
| | - Dawn Waterworth
- GlaxoSmithKline, Genetics, Collegeville, PA, United States of America
| | - A. Katrina Loomis
- Pfizer Worldwide Research and Development, Groton, CT, United States of America
| | - Jaak Vilo
- Institute of Computer Science, University of Tartu, Tartu, Estonia
| |
Collapse
|
39
|
Reisberg S, Galwey N, Avillach P, Sahlqvist AS, Kolberg L, Mägi R, Esko T, Vilo J, James G. Comparison of variation in frequency for SNPs associated with asthma or liver disease between Estonia, HapMap populations and the 1000 genome project populations. Int J Immunogenet 2019; 46:49-58. [PMID: 30659741 DOI: 10.1111/iji.12413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Accepted: 12/04/2018] [Indexed: 11/28/2022]
Abstract
Allele-specific analyses to understand frequency differences across populations, particularly populations not well studied, are important to help identify variants that may have a functional effect on disease mechanisms and phenotypic predisposition, facilitating new Genome-Wide Association Studies (GWAS). We aimed to compare the allele frequency of 11 asthma-associated and 16 liver disease-associated single nucleotide polymorphisms (SNPs) between the Estonian, HapMap and 1000 genome project populations. When comparing EGCUT with HapMap populations, the largest difference in allele frequencies was observed with the Maasai population in Kinyawa, Kenya, with 12 SNP variants reporting statistical significance. Similarly, when comparing EGCUT with 1000 genomes project populations, the largest difference in allele frequencies was observed with pooled African populations with 22 SNP variants reporting statistical significance. For 11 asthma-associated and 16 liver disease-associated SNPs, Estonians are genetically similar to other European populations but significantly different from African populations. Understanding differences in genetic architecture between ethnic populations is important to facilitate new GWAS targeted at underserved ethnic groups to enable novel genetic findings to aid the development of new therapies to reduce morbidity and mortality.
Collapse
Affiliation(s)
- Sulev Reisberg
- Institute of Computer Science, University of Tartu, Tartu, Estonia.,Software Technology and Applications Competence Center, Tartu, Estonia.,Quretec Ltd, Tartu, Estonia
| | | | - Paul Avillach
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA.,Department of Medical Informatics, Erasmus University Medical Center, Rotterdam, Netherlands
| | | | - Liis Kolberg
- Institute of Computer Science, University of Tartu, Tartu, Estonia
| | - Reedik Mägi
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Tõnu Esko
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Jaak Vilo
- Institute of Computer Science, University of Tartu, Tartu, Estonia
| | - Glen James
- Research and Development, GlaxoSmithKline, Stevenage, UK
| |
Collapse
|
40
|
Peng Z, Li X, Xing D, Du X, Wang Z, Liu G, Li X. Nobiletin alleviates palmitic acid‑induced NLRP3 inflammasome activation in a sirtuin 1‑dependent manner in AML‑12 cells. Mol Med Rep 2018; 18:5815-5822. [DOI: 10.3892/mmr.2018.9615] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 09/11/2018] [Indexed: 11/05/2022] Open
Affiliation(s)
- Zhicheng Peng
- Key Laboratory of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, Jilin 130062, P.R. China
| | - Xiaobing Li
- Key Laboratory of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, Jilin 130062, P.R. China
| | - Dongmei Xing
- Department of Basic Veterinary Medicine, Animal Medicine College, Hunan�Agriculture University, Changsha, Hunan 410128, P.R. China
| | - Xiliang Du
- Key Laboratory of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, Jilin 130062, P.R. China
| | - Zhe Wang
- Key Laboratory of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, Jilin 130062, P.R. China
| | - Guowen Liu
- Key Laboratory of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, Jilin 130062, P.R. China
| | - Xinwei Li
- Key Laboratory of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, Jilin 130062, P.R. China
| |
Collapse
|
41
|
Al-Muzafar HM, Amin KA. Thiazolidinedione induces a therapeutic effect on hepatosteatosis by regulating stearoyl-CoA desaturase-1, lipase activity, leptin and resistin. Exp Ther Med 2018; 16:2938-2948. [PMID: 30214514 PMCID: PMC6125847 DOI: 10.3892/etm.2018.6563] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2017] [Accepted: 04/06/2018] [Indexed: 02/06/2023] Open
Abstract
Hepatosteatosis is a disease present worldwide, which presents a number of health problems. Recently, thiazolidinedione (TZD) has been used as a therapy for lipid disorders. The present study demonstrates the potential of TZD as a treatment for hepatosteatosis and its mechanism of action, particularly focusing on its role in lipid metabolism. A total of 60 (80-90 g) rats were divided into three groups: A normal group with a standard diet, a high-fat, high-carbohydrate diet (HFCD) group or a HFCD+TZD group (n=20/group). The HFCD induced hepatosteatosis over a period of 12 weeks and the HFCD+TZD group were administered TZD in weeks 13-16. Blood and tissue samples were collected to measure hepatic function, the lipid profile, metabolism and hormone biomarkers, including serum triglyceride (TG), lipoprotein lipase (LPL), stearoyl-CoA desaturase (SCD-1), leptin and resistin. The HFCD-fed rats exhibited a significant increase in serum TG, total cholesterol, low-density lipoproteins, alanine transaminase and bilirubin compared with the normal group as well as a significant decrease in high-density lipoprotein. In addition, serum leptin and resistin were significantly elevated in the HFCD group compared with the normal group. The administration of TZD significantly increased SCD-1 activity and significantly inhibited LPL activity. It also attenuated the changes in the lipid profiles and normalized serum leptin and resistin levels. The results of the present study indicated that HFCD induced lipid abnormalities associated with hypertriglyceridemia, hypercholesterolemia and hepatosteatosis. These changes resulted from disruption to leptin and resistin, which may be due to alterations in LPL and SCD-1 activity. TZD mitigated the effects of HFCD-induced hepatosteatosis, indicating a possible regulatory effect of TZD in the development of hepatosteatosis. The authors suggest that the manipulation of SCD-1 and lipase by TZD may be useful as a treatment for hepatosteatosis.
Collapse
Affiliation(s)
- Hessah Mohammed Al-Muzafar
- Department of Chemistry and Biochemistry, College of Science, Imam Abdulrahman Bin Faisal University, Dammam 31441, Saudi Arabia
| | - Kamal Adel Amin
- Department of Chemistry and Biochemistry, College of Science, Imam Abdulrahman Bin Faisal University, Dammam 31441, Saudi Arabia
| |
Collapse
|
42
|
Sayari S, Neishaboori H, Jameshorani M. Combined effects of synbiotic and sitagliptin versus sitagliptin alone in patients with nonalcoholic fatty liver disease. Clin Mol Hepatol 2018; 24:331-338. [PMID: 29890569 PMCID: PMC6166102 DOI: 10.3350/cmh.2018.0006] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Accepted: 02/20/2018] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND/AIMS Non-alcoholic fatty liver disease (NAFLD) is one of the most prevalent chronic liver diseases in recent years. The aim of this study was to evaluate the effects of sitagliptin with and without a synbiotic supplement in the treatment of patients with NAFLD. METHODS In total, 138 NAFLD patients aged 18-60 years were enrolled in the study. Patients were randomized to one of the following treatments for 16 weeks: Group I (n=68), sitagliptin 50 mg daily plus placebo (one capsule per day) or group II (n=70) sitagliptin 50 mg daily plus synbiotic (one capsule per day). Changes in fasting blood glucose (FBS), liver enzymes, lipid profile, and body mass index were compared between the groups. RESULTS The mean change in FBS with sitagliptin-placebo from baseline was -10.47±5.77 mg/dL, and that with sitagliptin-synbiotic was -13.52±4.16 mg/dL. There was a significant difference between the groups (P<0.001). The mean change in cholesterol (Chol) was -8.34±28.83 mg/dL with sitagliptin-placebo and -21.25±15.50 mg/dL with sitagliptinsynbiotic. There was a significant difference between the two groups (P=0.029). The administration of sitagliptin-placebo induced an increase of 6.13±27.04 mg/dL in low density lipoprotein (LDL), whereas sitagliptin-synbiotic induced a decrease of 14.92±15.85 mg/dL in LDL. A significant difference was observed between the two groups (P<0.001). On the other hand, in the sitagliptin-synbiotic group, there was significant improvement in aspartate aminotransferase (AST) level compared to the sitagliptin-placebo group (P=0.018). CONCLUSION Sitagliptin-synbiotic produced greater improvement in FBS, AST, Chol, and LDL compared to sitagliptin alone in patients with NAFLD.
Collapse
Affiliation(s)
- Saba Sayari
- Zanjan Metabolic Disease Research Center, Vali-e-Asr Hospital, Zanjan University of Medical Science, Zanjan, Iran
| | - Hassan Neishaboori
- Zanjan Metabolic Disease Research Center, Vali-e-Asr Hospital, Zanjan University of Medical Science, Zanjan, Iran
| | - Maryam Jameshorani
- Zanjan Metabolic Disease Research Center, Vali-e-Asr Hospital, Zanjan University of Medical Science, Zanjan, Iran
| |
Collapse
|
43
|
Hypoxic Signaling and Cholesterol Lipotoxicity in Fatty Liver Disease Progression. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:2548154. [PMID: 29955245 PMCID: PMC6000860 DOI: 10.1155/2018/2548154] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/25/2018] [Revised: 04/30/2018] [Accepted: 05/14/2018] [Indexed: 02/06/2023]
Abstract
Cholesterol is the only lipid whose absorption in the gastrointestinal tract is limited by gate-keeping transporters and efflux mechanisms, preventing its rapid absorption and accumulation in the liver and blood vessels. In this review, I explored the current data regarding cholesterol accumulation in liver cells and key mechanisms in cholesterol-induced fatty liver disease associated with the activation of deleterious hypoxic and nitric oxide signal transduction pathways. Although nonalcoholic fatty liver disease (NAFLD) affects both obese and nonobese individuals, the mechanism of NAFLD progression in lean individuals with healthy metabolism is puzzling. Lean NAFLD individuals exhibit normal metabolic responses, implying that liver damage is not associated with impaired metabolism per se and that direct lipotoxic effects are crucial for disease progression. Several redox and oxidant signaling pathways involving cholesterol are at play in fatty liver disease development. These include impairment of the mitochondrial and lysosomal function by cholesterol loading of the inner-cell membranes; formation of cholesterol crystals and hepatocyte degradation; and crown-like structures surrounding degrading hepatocytes, activating Kupffer cells, and evoking inflammation. The current review focuses on the induction of liver inflammation, fibrosis, and steatosis by free cholesterol via the hypoxia-inducible factor 1α (HIF-1α), a main oxygen-sensing transcription factor involved in all stages of NAFLD. Cholesterol loading in hepatocytes can result in chronic HIF-1α activity because of the decreased oxygen availability and excessive production of nitric oxide and mitochondrial reactive oxygen species.
Collapse
|
44
|
Borrelli A, Bonelli P, Tuccillo FM, Goldfine ID, Evans JL, Buonaguro FM, Mancini A. Role of gut microbiota and oxidative stress in the progression of non-alcoholic fatty liver disease to hepatocarcinoma: Current and innovative therapeutic approaches. Redox Biol 2018; 15:467-479. [PMID: 29413959 PMCID: PMC5975181 DOI: 10.1016/j.redox.2018.01.009] [Citation(s) in RCA: 196] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Revised: 01/10/2018] [Accepted: 01/17/2018] [Indexed: 02/07/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) represents the most common chronic liver disease in industrialized countries. NAFLD progresses through the inflammatory phase of non-alcoholic steatohepatitis (NASH) to fibrosis and cirrhosis, with some cases developing liver failure or hepatocellular carcinoma (HCC). Liver biopsy remains the gold standard approach to a definitive diagnosis of NAFLD and the distinction between simple steatosis and NASH. The pathogenesis of NASH is still not clear. Several theories have been proposed ranging from the "Two Hit Theory" to the "Multiple Hit Theory". However, the general consensus is that the gut microbiota, oxidative stress, and mitochondrial damage play key roles in the pathogenesis of NASH. The interaction between the gut epithelia and some commensal bacteria induces the rapid generation of reactive oxygen species (ROS). The main goal of any therapy addressing NASH is to reverse or prevent progression to liver fibrosis/cirrhosis. This problem represents the first "Achilles' heel" of the new molecules being evaluated in most ongoing clinical trials. The second is the inability of these molecules to reach the mitochondria, the primary sites of energy production and ROS generation. Recently, a variety of non-pharmacological and pharmacological treatment approaches for NASH have been evaluated including vitamin E, the thiazolidinediones, and novel molecules related to NASH pathogenesis (including obeticholic acid and elafibranor). Recently, a new isoform of human manganese superoxide dismutase (MnSOD) was isolated and obtained in a synthetic recombinant form designated rMnSOD. This protein has been shown to be a powerful antioxidant capable of mediating ROS dismutation, penetrating biological barriers via its uncleaved leader peptide, and reducing portal hypertension and fibrosis in rats affected by liver cirrhosis. Based on these distinctive characteristics, it can be hypothesized that this novel recombinant protein (rMnSOD) potentially represents a new and highly efficient adjuvant therapy to counteract the progression from NASH to HCC.
Collapse
Affiliation(s)
- Antonella Borrelli
- Molecular Biology and Viral Oncology Unit, Istituto Nazionale Tumori IRCCS "Fondazione G Pascale", 80131 Napoli, Italy.
| | - Patrizia Bonelli
- Molecular Biology and Viral Oncology Unit, Istituto Nazionale Tumori IRCCS "Fondazione G Pascale", 80131 Napoli, Italy
| | - Franca Maria Tuccillo
- Molecular Biology and Viral Oncology Unit, Istituto Nazionale Tumori IRCCS "Fondazione G Pascale", 80131 Napoli, Italy
| | | | | | - Franco Maria Buonaguro
- Molecular Biology and Viral Oncology Unit, Istituto Nazionale Tumori IRCCS "Fondazione G Pascale", 80131 Napoli, Italy
| | - Aldo Mancini
- Leadhexa Biotechnologies Inc., Belvedere, CA, USA
| |
Collapse
|
45
|
Mahamid M, Mahroum N, Bragazzi NL, Shalaata K, Yavne Y, Adawi M, Amital H, Watad A. Folate and B12 Levels Correlate with Histological Severity in NASH Patients. Nutrients 2018; 10:440. [PMID: 29614799 PMCID: PMC5946225 DOI: 10.3390/nu10040440] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 03/27/2018] [Accepted: 03/30/2018] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The correlation between abnormal vitamin serum levels and chronic liver disease has been previously described in literature. However, the association between the severity of folate serum levels (B9), vitamin B12 and nonalcoholic steatohepatitis (NASH) has not been widely evaluated. Therefore, the aim of this study was to investigate the existence of such a correlation in a cohort of NASH patients. METHODS All patients aged 18 years and older who were diagnosed with biopsy-proven NASH at the EMMS hospital in Nazareth during the years 2015-2017 were enrolled in this study. Data regarding demographic, clinical and laboratory parameters was collected. Patients with other liver diseases were excluded. RESULTS Eighty-three NASH patients were enrolled during the study period. The mean age was 41 ± 11 years and the majority of patients were male. Mean values of folate and B12 were 9.85 ± 10.90 ng/mL and 387.53 ± 205.50 pg/mL, respectively. Half of the patients were presented with a grade 1 steatosis (43.4%), a grade 2 fibrosis (50.6%) and a grade 3 activity score (55.4%). The fibrosis grade was significantly correlated with low folate levels on multivariate analysis (p-value < 0.01). Similarly, low B12 levels were significantly associated with a higher fibrosis grade and NASH activity (p-value < 0.001 and p-value < 0.05 respectively). CONCLUSION Our study demonstrated a statistically significant correlation between low levels of folate and vitamin B12 with the histological severity of NASH. These findings could have diagnostic and therapeutic implications for patient management and follow-up.
Collapse
Affiliation(s)
- Mahmud Mahamid
- Endoscopy Unit, Nazareth Hospital EMMS, 16100 Nazareth, Israel.
- Azrieli Faculty of Medicine, Bar-Ilan University, 13195 Safed, Israel.
| | - Naim Mahroum
- Department of Medicine 'B', Sheba Medical Center, Tel-Hashomer, Israel, Sackler Faculty of Medicine, Tel-Aviv University, 52621 Tel-Aviv, Israel.
| | - Nicola Luigi Bragazzi
- School of Public Health, Department of Health Sciences, University of Genoa, 16132 Genoa, Italy.
| | - Kasem Shalaata
- Azrieli Faculty of Medicine, Bar-Ilan University, 13195 Safed, Israel.
- Internal Medicine Department, Nazareth Hospital EMMS, 16100 Nazareth, Israel.
| | - Yarden Yavne
- Department of Medicine 'B', Sheba Medical Center, Tel-Hashomer, Israel, Sackler Faculty of Medicine, Tel-Aviv University, 52621 Tel-Aviv, Israel.
| | | | - Howard Amital
- Department of Medicine 'B', Sheba Medical Center, Tel-Hashomer, Israel, Sackler Faculty of Medicine, Tel-Aviv University, 52621 Tel-Aviv, Israel.
| | - Abdulla Watad
- Department of Medicine 'B', Sheba Medical Center, Tel-Hashomer, Israel, Sackler Faculty of Medicine, Tel-Aviv University, 52621 Tel-Aviv, Israel.
| |
Collapse
|
46
|
Affiliation(s)
| | - Piero Pingitore
- University of Gothenburg, Sweden Department of Molecular and Clinical Medicine
| |
Collapse
|
47
|
Narayanankutty A, Palliyil DM, Kuruvilla K, Raghavamenon AC. Virgin coconut oil reverses hepatic steatosis by restoring redox homeostasis and lipid metabolism in male Wistar rats. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2018; 98:1757-1764. [PMID: 28862329 DOI: 10.1002/jsfa.8650] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Revised: 07/28/2017] [Accepted: 08/27/2017] [Indexed: 06/07/2023]
Abstract
BACKGROUND Hepatosteatosis, a form of nonalcoholic fatty liver disease (NAFLD), is being increasingly recognized as a major health burden worldwide. Insulin resistance, dyslipidemia and imbalances in adipokine/cytokine interplay are reported to be involved in the onset and progression of this disease. Use of dietary nutraceuticals in prevention and treatment of NAFLD is emerging. Virgin coconut oil (VCO), a fermented product of fresh coconut kernel, has been shown to impede the development of hepatosteatosis in rats. This study analyzes the potential of VCO to reverse the already developed hepatosteatosis condition. RESULTS Hyperglycemia, reduced glucose tolerance, dyslipidemia, and hepatic macrovesicles in high-fructose-diet-fed rats (4 weeks) confirmed the development of hepatosteatosis. Natural reversion in these parameters was observed upon shifting to normal diet in untreated control animals. Administration of VCO, however, increased this natural reversion by improving high-density lipoprotein cholesterol level (53.5%) and reducing hepatic and serum triacylglycerols (78.0 and 51.7%). Increased hepatic glutathione level (P < 0.01), antioxidant enzyme activities (P < 0.05) and reduced lipid peroxidation were also noticed in these animals. These observations were in concordance with reduced liver enzyme activities (P < 0.01) and restoration of altered hepatic architecture. CONCLUSION The study indicates that VCO can be used as a nutraceutical against hepatosteatosis. © 2017 Society of Chemical Industry.
Collapse
Affiliation(s)
| | - Devika Mukundan Palliyil
- Amala Cancer Research Centre, Amala Nagar P O, Thrissur, Kerala, India
- Department of Zoology, Vimala College, Thrissur, Kerala, India
| | - Kezia Kuruvilla
- Department of Zoology, Vimala College, Thrissur, Kerala, India
| | | |
Collapse
|
48
|
Cassard AM, Gérard P, Perlemuter G. Microbiota, Liver Diseases, and Alcohol. BUGS AS DRUGS 2018:187-212. [DOI: 10.1128/9781555819705.ch8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
Affiliation(s)
- Anne-Marie Cassard
- INSERM U996 Inflammation, Chemokines and Immunopathology, DHU Hepatinov, Univ Paris-Sud; Université Paris-Saclay; 92140 Clamart France
| | - Philippe Gérard
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay; 78350 Jouyen-Josas France
| | - Gabriel Perlemuter
- INSERM U996 Inflammation, Chemokines and Immunopathology, DHU Hepatinov, Univ Paris-Sud; Université Paris-Saclay; 92140 Clamart France
- AP-HP, Hepatogastroenterology and Nutrition, Hôpital Antoine-Béclère; Clamart France
| |
Collapse
|
49
|
|
50
|
Zhang J, Zhang H, Deng X, Zhang Y, Xu K. Baicalin protects AML-12 cells from lipotoxicity via the suppression of ER stress and TXNIP/NLRP3 inflammasome activation. Chem Biol Interact 2017; 278:189-196. [DOI: 10.1016/j.cbi.2017.10.010] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Revised: 07/28/2017] [Accepted: 10/09/2017] [Indexed: 02/08/2023]
|