51
|
Shang Y, Widman L, Hagström H. Nonalcoholic Fatty Liver Disease and Risk of Dementia: A Population-Based Cohort Study. Neurology 2022; 99:e574-e582. [PMID: 35831178 PMCID: PMC9442617 DOI: 10.1212/wnl.0000000000200853] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 04/28/2022] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND AND OBJECTIVES Nonalcoholic fatty liver disease (NAFLD) and dementia share common risk factors including metabolic disorders. However, whether NAFLD is associated with dementia risk is unclear. We investigated the association between NAFLD and dementia risk as well as the role of cardiovascular complications including heart disease and stroke. METHODS In this population-based matched cohort study, we identified all Swedish patients aged 65 years or older with NAFLD identified from the National Patient Register (NPR) between 1987 and 2016. These were matched with up to 10 reference individuals from the general population on age, sex, and municipality at the year of diagnosis. Incident dementia diagnosis was derived from the NPR or the Cause of Death Register until 2016. Adjusted hazard ratios (aHRs) and 95% CIs were estimated with Cox regression models. RESULTS A total of 2,898 patients with NAFLD and 28,357 matched controls were identified (median age at entry, interquartile range [IQR], 70 [8]; 55.1% female). During a median follow-up of 5.5 years (IQR: 8.5 years), 145 (5.0%) patients with NAFLD and 1,291 (4.6%) reference individuals were diagnosed with dementia. Compared with the reference individuals, patients with NAFLD had higher rates of dementia (aHR 1.38, 95% CI 1.10-1.72) and vascular dementia (aHR 1.44, 95% CI 0.96-2.23, p = 0.07). Comorbid NAFLD and either heart disease (aHR 1.50 95% 1.08-2.05) or stroke (aHR 2.60 95% CI 1.95-3.47) conferred a greater risk of dementia. DISCUSSION NAFLD had a modest association with increased rates of dementia. This was stronger among patients with NAFLD diagnosed with cardiovascular comorbidities. CLASSIFICATION OF EVIDENCE This study provides Class II evidence that nonalcoholic fatty liver disease is associated with the development of vascular and nonvascular dementia.
Collapse
Affiliation(s)
- Ying Shang
- From the Department of Medicine (Y.S., H.H.), Huddinge, Karolinska Institutet; Division of Biostatistics (L.W.), Institute of Environmental Medicine, Karolinska Institutet; Division of Hepatology (H.H.), Department of Upper GI, Karolinska University Hospital; and Clinical Epidemiology Unit (H.H.), Department of Medicine, Karolinska Institutet, Stockholm, Sweden.
| | - Linnea Widman
- From the Department of Medicine (Y.S., H.H.), Huddinge, Karolinska Institutet; Division of Biostatistics (L.W.), Institute of Environmental Medicine, Karolinska Institutet; Division of Hepatology (H.H.), Department of Upper GI, Karolinska University Hospital; and Clinical Epidemiology Unit (H.H.), Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Hannes Hagström
- From the Department of Medicine (Y.S., H.H.), Huddinge, Karolinska Institutet; Division of Biostatistics (L.W.), Institute of Environmental Medicine, Karolinska Institutet; Division of Hepatology (H.H.), Department of Upper GI, Karolinska University Hospital; and Clinical Epidemiology Unit (H.H.), Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
52
|
Cazac GD, Lăcătușu CM, Mihai C, Grigorescu ED, Onofriescu A, Mihai BM. New Insights into Non-Alcoholic Fatty Liver Disease and Coronary Artery Disease: The Liver-Heart Axis. Life (Basel) 2022; 12:1189. [PMID: 36013368 PMCID: PMC9410285 DOI: 10.3390/life12081189] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 07/28/2022] [Accepted: 07/29/2022] [Indexed: 12/17/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) represents the hepatic expression of the metabolic syndrome and is the most prevalent liver disease. NAFLD is associated with liver-related and extrahepatic morbi-mortality. Among extrahepatic complications, cardiovascular disease (CVD) is the primary cause of mortality in patients with NAFLD. The most frequent clinical expression of CVD is the coronary artery disease (CAD). Epidemiological data support a link between CAD and NAFLD, underlain by pathogenic factors, such as the exacerbation of insulin resistance, genetic phenotype, oxidative stress, atherogenic dyslipidemia, pro-inflammatory mediators, and gut microbiota. A thorough assessment of cardiovascular risk and identification of all forms of CVD, especially CAD, are needed in all patients with NAFLD regardless of their metabolic status. Therefore, this narrative review aims to examine the available data on CAD seen in patients with NAFLD, to outline the main directions undertaken by the CVD risk assessment and the multiple putative underlying mechanisms implicated in the relationship between CAD and NAFLD, and to raise awareness about this underestimated association between two major, frequent and severe diseases.
Collapse
Affiliation(s)
- Georgiana-Diana Cazac
- Unit of Diabetes, Nutrition and Metabolic Diseases, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania
- Clinical Center of Diabetes, Nutrition and Metabolic Diseases, “St. Spiridon” County Clinical Emergency Hospital, 700111 Iași, Romania
| | - Cristina-Mihaela Lăcătușu
- Unit of Diabetes, Nutrition and Metabolic Diseases, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania
- Clinical Center of Diabetes, Nutrition and Metabolic Diseases, “St. Spiridon” County Clinical Emergency Hospital, 700111 Iași, Romania
| | - Cătălina Mihai
- Institute of Gastroenterology and Hepatology, “Sf. Spiridon” Emergency Hospital, 700111 Iași, Romania
- Unit of Medical Semiology and Gastroenterology, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Elena-Daniela Grigorescu
- Unit of Diabetes, Nutrition and Metabolic Diseases, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania
| | - Alina Onofriescu
- Unit of Diabetes, Nutrition and Metabolic Diseases, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania
- Clinical Center of Diabetes, Nutrition and Metabolic Diseases, “St. Spiridon” County Clinical Emergency Hospital, 700111 Iași, Romania
| | - Bogdan-Mircea Mihai
- Unit of Diabetes, Nutrition and Metabolic Diseases, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania
- Clinical Center of Diabetes, Nutrition and Metabolic Diseases, “St. Spiridon” County Clinical Emergency Hospital, 700111 Iași, Romania
| |
Collapse
|
53
|
Galiano Rus S, Ortiz García de la Foz V, Arias-Loste MT, Iruzubieta P, Gómez-Revuelta M, Juncal-Ruiz M, Crespo J, Crespo-Facorro B, Vázquez-Bourgon J. Elevated risk of liver steatosis in first-episode psychosis patients: Results from a 3-year prospective study. Schizophr Res 2022; 246:30-38. [PMID: 35696859 DOI: 10.1016/j.schres.2022.06.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 01/20/2022] [Accepted: 06/01/2022] [Indexed: 12/09/2022]
Abstract
BACKGROUND Cardiometabolic disorders are largely responsible for excess mortality in schizophrenia. Non-alcoholic fatty liver disease (NAFLD) is increasingly relevant in the development of metabolic risk factors that have been associated with antipsychotic treatment. We aimed to assess the incidence of NAFLD and metabolic disturbances during the first 3 years of antipsychotic treatment in patients with first episode of psychosis (FEP), and compare it with the incidence in a control group. METHODS Data were obtained from patients with psychosis (n = 160) and healthy controls (n = 66) included in the Cantabria's clinical and research program on FEP (PAFIP) from 2012 to 2018. Fatty Liver Index (FLI) was used to estimate the amount of fat in the liver. FLI has been validated for the diagnosis of NAFLD against different standards such as liver ultrasound and biopsy. FLI and metabolic parameters were registered at baseline, 3 months and then yearly for 3 years. RESULTS At the end of the follow-up (3-years), 21.9 % of patients with psychosis developed a FLI ≥ 60, suggestive of liver steatosis, compared to only a 3 % of subjects within the control group (X2 = 12.120; p < 0.001). In the FEP patients group, developing a FLI ≥ 60 was statistically associated with significant increments in metabolic parameters, and with Metabolic Syndrome (MetS) (X2 = 16.151; p < 0.001) and high blood pressure (X2 = 10.654; p = 0.001). CONCLUSIONS Having a first episode of non-affective psychosis was significantly associated with developing liver steatosis (FLI ≥ 60) in the first three years after initiating antipsychotic treatment. The results highlight the importance of early screening the emergence of NAFLD in schizophrenia patients.
Collapse
Affiliation(s)
- Sara Galiano Rus
- Department of Psychiatry, University Hospital Marqués de Valdecilla, School of Medicine, University of Cantabria, Institute of Biomedical Research Valdecilla (IDIVAL), Santander, Spain
| | - Víctor Ortiz García de la Foz
- Department of Psychiatry, University Hospital Marqués de Valdecilla, School of Medicine, University of Cantabria, Institute of Biomedical Research Valdecilla (IDIVAL), Santander, Spain
| | - María Teresa Arias-Loste
- Gastroenterology and Hepatology Department, University Hospital Marqués de Valdecilla, School of Medicine, University of Cantabria, Institute of Biomedical Research Valdecilla (IDIVAL), Santander, Spain; Centro de Investigación Biomédica en Red en Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain
| | - Paula Iruzubieta
- Gastroenterology and Hepatology Department, University Hospital Marqués de Valdecilla, School of Medicine, University of Cantabria, Institute of Biomedical Research Valdecilla (IDIVAL), Santander, Spain; Centro de Investigación Biomédica en Red en Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain
| | - Marcos Gómez-Revuelta
- Department of Psychiatry, University Hospital Marqués de Valdecilla, School of Medicine, University of Cantabria, Institute of Biomedical Research Valdecilla (IDIVAL), Santander, Spain
| | - María Juncal-Ruiz
- Department of Psychiatry, School of Medicine (UC), Sierrallana Hospital, Instituto of Biomedical Research Valdecilla (IDIVAL), Santander, Spain
| | - Javier Crespo
- Gastroenterology and Hepatology Department, University Hospital Marqués de Valdecilla, School of Medicine, University of Cantabria, Institute of Biomedical Research Valdecilla (IDIVAL), Santander, Spain; Centro de Investigación Biomédica en Red en Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain
| | - Benedicto Crespo-Facorro
- Department of Psychiatry, School of Medicine, University Hospital Virgen del Rocio-IBIS, Sevilla, Spain; Centro de Investigación Biomédica en Red en Salud Mental (CIBERSAM), Madrid, Spain
| | - Javier Vázquez-Bourgon
- Department of Psychiatry, University Hospital Marqués de Valdecilla, School of Medicine, University of Cantabria, Institute of Biomedical Research Valdecilla (IDIVAL), Santander, Spain; Centro de Investigación Biomédica en Red en Salud Mental (CIBERSAM), Madrid, Spain.
| |
Collapse
|
54
|
Hagström H, Kechagias S, Ekstedt M. Risk for hepatic and extra-hepatic outcomes in nonalcoholic fatty liver disease. J Intern Med 2022; 292:177-189. [PMID: 34118091 DOI: 10.1111/joim.13343] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is defined by presence of steatosis in more than 5% of liver cells. The gold standard for diagnosis is liver biopsy, but this is seldom achieved due to costs and risk for side effects, and that is why the diagnosis is mostly made based on a combination of radiology and exclusion of other liver diseases. Disease severity staging can be noninvasively achieved with radiological exams such as elastography or blood-based markers that usually have lower sensitivity and specificity. NAFLD is today the most common chronic liver disease globally with a prevalence estimated to be 25%. Fortunately, for many persons NAFLD is an incidental finding with a good prognosis. Whilst a major focus has been on liver-related outcomes in NAFLD, there has recently been an increased interest in extrahepatic consequences of NAFLD. The most commonly studied outcomes include cardiovascular disease and cancer. The risk of adverse outcomes generally differs according to the baseline fibrosis stage. There is a five-time higher risk of liver-related events in NAFLD patients with fibrosis stage 3 as compared to those with no or little fibrosis. Meanwhile, the presence of nonalcoholic steatohepatitis (NASH) does not seem to influence prognosis in addition to fibrosis stage. Patients with NAFLD clearly have a higher risk for cardiovascular outcomes compared to the general population, with a recent meta-analysis indicating a 37% increased hazard for cardiovascular events as opposed to individuals without NAFLD. The risk of liver cancer is increased, which is mostly driven by presence of cirrhosis, but the increased risk is present also in patients without cirrhosis, and to a greater extent than for other chronic liver disease. Around one-third of patients with NAFLD and liver cancer do not have cirrhosis. Additionally, the risk of extrahepatic malignancies is thought to be moderately increased, with most evidence for a link between NAFLD and colorectal cancer where the risk is approximately 50% higher compared to patients without NAFLD. A particularly salient point is if NAFLD can be considered an independent risk factor for outcomes. Many studies have not been able to adjust for key confounders, or suffer from different forms of bias. The clinical problem is nevertheless to identify persons with an increased risk for adverse hepatic and extrahepatic outcomes. We here discuss the evidence linking NAFLD to severe hepatic and extrahepatic outcomes.
Collapse
Affiliation(s)
- Hannes Hagström
- Division of Hepatology, Department of Upper GI diseases, Karolinska University Hospital, Stockholm, Sweden.,Clinical Epidemiology Division, Department of Medicine, Karolinska Institutet, Solna, Stockholm, Sweden.,Department of Medicine, Karolinska Institutet, Huddinge, Stockholm, Sweden
| | - Stergios Kechagias
- Division of Diagnostics and Specialist Medicine, Department of Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden
| | - Mattias Ekstedt
- Division of Diagnostics and Specialist Medicine, Department of Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden
| |
Collapse
|
55
|
Esteban JPG, Asgharpour A. Evaluation of liver transplant candidates with non-alcoholic steatohepatitis. Transl Gastroenterol Hepatol 2022; 7:24. [PMID: 35892057 PMCID: PMC9257540 DOI: 10.21037/tgh.2020.03.04] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 02/03/2020] [Indexed: 11/07/2023] Open
Abstract
Non-alcoholic steatohepatitis (NASH) is anticipated to become the leading indication for liver transplantation (LT) in the United States in the near future. LT is indicated in patients with NASH-related cirrhosis who have medically refractory hepatic decompensation, synthetic dysfunction, and hepatocellular carcinoma (HCC) meeting certain criteria. The objective of LT evaluation is to determine which patient will derive the most benefit from LT with the least risk, thus maximizing the societal benefits of a limited resource. LT evaluation is a multidisciplinary undertaking involving several specialists, assessment tools, and diagnostic testing. Although the steps involved in LT evaluation are relatively similar across different liver diseases, patients with NASH-related cirrhosis have unique demographic and clinical features that affect transplant outcomes and influence their LT evaluation. LT candidates with NASH should be assessed for metabolic syndrome and obesity, malnutrition and sarcopenia, frailty, and cardiovascular disease. Interventions that treat cardiometabolic co-morbidities and improve patients' nutrition and functionality should be considered in order to improve patient outcomes in the waitlist and after LT.
Collapse
Affiliation(s)
- James Philip G Esteban
- Division of Gastroenterology and Hepatology, Medical College of Wisconsin, Milwaukee, WI, USA
- Division of Liver Diseases, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Amon Asgharpour
- Division of Liver Diseases, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
56
|
Fatty Liver as Potential Biomarker of Atherosclerotic Damage in Familial Combined Hyperlipidemia. Biomedicines 2022; 10:biomedicines10081770. [PMID: 35892670 PMCID: PMC9332610 DOI: 10.3390/biomedicines10081770] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 07/20/2022] [Accepted: 07/20/2022] [Indexed: 11/22/2022] Open
Abstract
Familial combined hyperlipidemia (FCH) is a very common inherited lipid disorder, characterized by a high risk of developing cardiovascular (CV) disease and metabolic complications, including insulin resistance (IR) and type 2 diabetes mellitus (T2DM). The prevalence of non-alcoholic fatty liver disease (NAFLD) is increased in FCH patients, especially in those with IR or T2DM. However, it is unknown how precociously metabolic and cardiovascular complications appear in FCH patients. We aimed to evaluate the prevalence of NAFLD and to assess CV risk in newly diagnosed insulin-sensitive FCH patients. From a database including 16,504 patients, 110 insulin-sensitive FCH patients were selected by general practitioners and referred to the Lipid Center. Lipid profile, fasting plasma glucose and insulin were determined by standard methods. Based on the results of the hospital screening, 96 patients were finally included (mean age 52.2 ± 9.8 years; 44 males, 52 females). All participants underwent carotid ultrasound to assess carotid intima media thickness (cIMT), presence or absence of plaque, and pulse wave velocity (PWV). Liver steatosis was assessed by both hepatic steatosis index (HSI) and abdomen ultrasound (US). Liver fibrosis was non-invasively assessed by transient elastography (TE) and by fibrosis 4 score (FIB-4) index. Carotid plaque was found in 44 out of 96 (45.8%) patients, liver steatosis was found in 68 out of 96 (70.8%) and in 41 out of 96 (42.7%) patients by US examination and HSI, respectively. Overall, 72 subjects (75%) were diagnosed with steatosis by either ultrasound or HSI, while 24 (25%) had steatosis excluded (steatosis excluded by both US and HSI). Patients with liver steatosis had a significantly higher body mass index (BMI) compared to those without (p < 0.05). Steatosis correlated with fasting insulin (p < 0.05), liver stiffness (p < 0.05), BMI (p < 0.001), and inversely with high-density lipoprotein cholesterol (p < 0.05). Fibrosis assessed by TE was significantly associated with BMI (p < 0.001) and cIMT (p < 0.05); fibrosis assessed by FIB-4 was significantly associated with sex (p < 0.05), cIMT (p < 0.05), and atherosclerotic plaque (p < 0.05). The presence of any grade of liver fibrosis was significantly associated with atherosclerotic plaque in the multivariable model, independent of alcohol habit, sex, HSI score, and liver stiffness by TE (OR 6.863, p < 0.001). In our cohort of newly diagnosed, untreated, insulin-sensitive FCH patients we found a high prevalence of liver steatosis. Indeed, the risk of atherosclerotic plaque was significantly increased in patients with liver fibrosis, suggesting a possible connection between liver disease and CV damage in dyslipidemic patients beyond the insulin resistance hypothesis.
Collapse
|
57
|
The establishment of public health policies and the burden of non-alcoholic fatty liver disease in the Americas. Lancet Gastroenterol Hepatol 2022; 7:552-559. [PMID: 35430032 DOI: 10.1016/s2468-1253(22)00008-5] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 01/12/2022] [Accepted: 01/13/2022] [Indexed: 11/17/2022]
|
58
|
Hassen G, Singh A, Belete G, Jain N, De la Hoz I, Camacho-Leon GP, Dargie NK, Carrera KG, Alemu T, Jhaveri S, Solomon N. Nonalcoholic Fatty Liver Disease: An Emerging Modern-Day Risk Factor for Cardiovascular Disease. Cureus 2022; 14:e25495. [PMID: 35783879 PMCID: PMC9242599 DOI: 10.7759/cureus.25495] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 05/30/2022] [Indexed: 11/05/2022] Open
|
59
|
Galatou E, Mourelatou E, Hatziantoniou S, Vizirianakis IS. Nonalcoholic Steatohepatitis (NASH) and Atherosclerosis: Explaining Their Pathophysiology, Association and the Role of Incretin-Based Drugs. Antioxidants (Basel) 2022; 11:1060. [PMID: 35739957 PMCID: PMC9220192 DOI: 10.3390/antiox11061060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 05/23/2022] [Accepted: 05/24/2022] [Indexed: 11/17/2022] Open
Abstract
Nonalcoholic steatohepatitis (NASH) is the most severe manifestation of nonalcoholic fatty liver disease (NAFLD), a common complication of type 2 diabetes, and may lead to cirrhosis and hepatocellular carcinoma. Oxidative stress and liver cell damage are the major triggers of the severe hepatic inflammation that characterizes NASH, which is highly correlated with atherosclerosis and coronary artery disease. Regarding drug therapy, research on the role of GLP-1 analogues and DPP4 inhibitors, novel classes of antidiabetic drugs, is growing. In this review, we outline the association between NASH and atherosclerosis, the underlying molecular mechanisms, and the effects of incretin-based drugs, especially GLP-1 RAs, for the therapeutic management of these conditions.
Collapse
Affiliation(s)
- Eleftheria Galatou
- Department of Life & Health Sciences, School of Sciences and Engineering, University of Nicosia, 2417 Nicosia, Cyprus;
| | - Elena Mourelatou
- Department of Life & Health Sciences, School of Sciences and Engineering, University of Nicosia, 2417 Nicosia, Cyprus;
| | - Sophia Hatziantoniou
- Laboratory of Pharmaceutical Technology, Department of Pharmacy, School of Health Sciences, University of Patras, 26504 Patras, Greece;
| | - Ioannis S. Vizirianakis
- Department of Life & Health Sciences, School of Sciences and Engineering, University of Nicosia, 2417 Nicosia, Cyprus;
- Laboratory of Pharmacology, School of Pharmacy, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| |
Collapse
|
60
|
Comprehensive Review and Updates on Holistic Approach Towards Non-Alcoholic Fatty Liver Disease Management with Cardiovascular Disease. Curr Atheroscler Rep 2022; 24:515-532. [PMID: 35507280 DOI: 10.1007/s11883-022-01027-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/01/2022] [Indexed: 02/06/2023]
Abstract
PURPOSE OF REVIEW The global prevalence of non-alcoholic fatty liver disease (NAFLD) presents an unmet need in treating these, often asymptomatic, individuals. In this review, we summarised NAFLD management and described recent developments in non-alcoholic steatohepatitis (NASH) therapeutics that can shape the future of NAFLD. RECENT FINDINGS A multi-disciplinary effort in promoting sustainable lifestyle measures is paramount, with the goal of either limiting energy surplus alone or in combination with targeting downstream pathways of inflammation and fibrosis. Several antidiabetic medications like PPAR-γ agonist and glucagon-like peptide receptor agonists have beneficial effects on the metabolic profile as well as NASH histology. Vitamin E has shown promise in specific groups of patients with the haptoglobin2 allele protein. Newer drugs have demonstrated promising results in NASH resolution and fibrosis improvement such as obeticholic acid, resmetirom, aramchol, efruxifermin, aldafermin and lanifibranor. Apart from discussing the results of late stage clinical trials and the possible challenges in managing these patients with limited approved therapies, we also discussed the specific management of comorbidities (diabetes, hypertension, hyperlipidaemia, cardiovascular diseases) in NAFLD patients. Treatment strategy needs to target improvements in liver-related outcomes and cardiometabolic profile.
Collapse
|
61
|
Kaya E, Yilmaz Y. Metabolic-associated Fatty Liver Disease (MAFLD): A Multi-systemic Disease Beyond the Liver. J Clin Transl Hepatol 2022; 10:329-338. [PMID: 35528971 PMCID: PMC9039705 DOI: 10.14218/jcth.2021.00178] [Citation(s) in RCA: 68] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 08/19/2021] [Accepted: 09/17/2021] [Indexed: 02/05/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a multisystemic clinical condition that presents with a wide spectrum of extrahepatic manifestations, such as obesity, type 2 diabetes mellitus, metabolic syndrome, cardiovascular diseases, chronic kidney disease, extrahepatic malignancies, cognitive disorders, and polycystic ovarian syndrome. Among NAFLD patients, the most common mortality etiology is cardiovascular disorders, followed by extrahepatic malignancies, diabetes mellitus, and liver-related complications. Furthermore, the severity of extrahepatic diseases is parallel to the severity of NAFLD. In clinical practice, awareness of the associations of concomitant diseases is of major importance for initiating prompt and timely screening and multidisciplinary management of the disease spectrum. In 2020, a consensus from 22 countries redefined the disease as metabolic (dysfunction)-associated fatty liver disease (MAFLD), which resulted in the redefinition of the corresponding population. Although the patients diagnosed with MAFLD and NAFLD mostly overlap, the MAFLD and NAFLD populations are not identical. In this review, we compared the associations of key extrahepatic diseases between NAFLD and MAFLD.
Collapse
Affiliation(s)
- Eda Kaya
- Department of Internal Medicine, Ruhr University Bochum, University Hospital Knappschaftskrankenhaus Bochum, Bochum, Germany
| | - Yusuf Yilmaz
- Department of Gastroenterology, School of Medicine, Marmara University, Istanbul, Turkey
- Liver Research Unit, Institute of Gastroenterology, Marmara University, Istanbul, Turkey
| |
Collapse
|
62
|
Tang Y, Li H, Chen C. Non-coding RNA-Associated Therapeutic Strategies in Atherosclerosis. Front Cardiovasc Med 2022; 9:889743. [PMID: 35548442 PMCID: PMC9081650 DOI: 10.3389/fcvm.2022.889743] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 03/21/2022] [Indexed: 12/14/2022] Open
Abstract
Atherosclerosis has been the main cause of disability and mortality in the world, resulting in a heavy medical burden for all countries. It is widely known to be a kind of chronic inflammatory disease in the blood walls, of which the key pathogenesis is the accumulation of immunologic cells in the lesion, foam cells formation, and eventually plaque rupture causing ischemia of various organs. Non-coding RNAs (ncRNAs) play a vital role in regulating the physiologic and pathophysiologic processes in cells. More and more studies have revealed that ncRNAs also participated in the development of atherosclerosis and regulated cellular phenotypes such as endothelial dysfunction, leukocyte recruitment, foam cells formation, and vascular smooth muscle cells phenotype-switching and apoptosis. Given the broad functions of ncRNAs in atherogenesis, they have become potential therapeutic targets. Apart from that, ncRNAs have become powerful blueprints to design new drugs. For example, RNA interference drugs were inspired by small interfering RNAs that exist in normal cellular physiologic processes and behave as negative regulators of specific proteins. For instance, inclisiran is a kind of RNAi drug targeting PCKS9 mRNA, which can lower the level of LDL-C and treat atherosclerosis. We introduce some recent research progresses on ncRNAs related to atherosclerotic pathophysiologic process and the current clinical trials of RNA drugs pointed at atherosclerosis.
Collapse
Affiliation(s)
- Yuyan Tang
- Division of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
| | - Huaping Li
- Division of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
- *Correspondence: Huaping Li
| | - Chen Chen
- Division of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
- Chen Chen
| |
Collapse
|
63
|
Elsaid MI, Bridges JF, Li N, Rustgi VK. Metabolic Syndrome Severity Predicts Mortality in Nonalcoholic Fatty Liver Disease. GASTRO HEP ADVANCES 2022; 1:445-456. [PMID: 39131673 PMCID: PMC11308509 DOI: 10.1016/j.gastha.2022.02.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 02/02/2022] [Indexed: 08/13/2024]
Abstract
Background and Aims Previous studies have examined the effects of metabolic syndrome (MetS) presence rather than the severity on mortality risk in nonalcoholic fatty liver disease (NAFLD). We used the MetS severity score, a validated gender- and race-specific measure, to assess the relationship between MetS severity and mortality risk in NAFLD. Methods The study included 10,638 adults aged between 20 and 74 years who participated in the Third National Health and Nutrition Examination Survey. NAFLD was defined as mild, moderate, or severe hepatic steatosis on ultrasound without excessive alcohol intake and other liver diseases. Adjusted Cox proportional models were used to test the association between the MetS severity score and mortality risk related to all-cause, heart disease, diabetes, and hypertension. Results The median MetS severity score was significantly higher in NAFLD (0.49 [69th] vs -0.23 [41st]). An increase in the MetS severity corresponded to a linear rise in biomarkers for cardiovascular disease, insulin resistance, lipid abnormalities, and liver and kidney problems. The MetS severity score was a significant predictor for all-cause and cause-specific adjusted mortalities. A quartile increase in MetS severity score was associated with higher mortality risks from all-causes adjusted hazard ratio (aHR) 1.36 (95% confidence interval [CI]: 1.17-1.57), heart disease aHR 1.70 (95% CI: 1.17-2.47), diabetes aHR 3.64 (95% CI: 2.27-5.83), and hypertension aHR 1.87 (95% CI: 1.14-3.04). A higher MetS severity score was also associated with nonlinear increased risks of mortality in all adjusted models. Conclusion The MetS severity score is a clinically accessible tool that can be used to identify and monitor NAFLD patients at the highest risk of mortality.
Collapse
Affiliation(s)
- Mohamed I. Elsaid
- Department of Biomedical Informatics, College of Medicine, The Ohio State University, Columbus, Ohio
- Secondary Data Core, Center for Biostatistics, College of Medicine, The Ohio State University, Columbus, Ohio
| | - John F.P. Bridges
- Secondary Data Core, Center for Biostatistics, College of Medicine, The Ohio State University, Columbus, Ohio
- Department of Surgery, College of Medicine, The Ohio State University, Columbus, Ohio
| | - Na Li
- Division of Gastroenterology, Hepatology, & Nutrition, College of Medicine, The Ohio State University, Columbus, Ohio
| | - Vinod K. Rustgi
- Division of Gastroenterology and Hepatology, Rutgers Robert Wood Johnson Medical School, New Brunswick, New Jersey
- Center for Liver Diseases and Masses, Rutgers Robert Wood Johnson Medical School, New Brunswick, New Jersey
| |
Collapse
|
64
|
Carter J, Heseltine TD, Meah MN, Tzolos E, Kwiecinski J, Doris M, McElhinney P, Moss AJ, Adamson PD, Hunter A, Alam S, Shah ASV, Pawade T, Wang C, Weir-McCall JR, Roditi G, van Beek EJR, Nicol ED, Shaw LJ, Berman DS, Slomka PJ, Mills NL, Dweck MR, Newby DE, Murray SW, Dey D, Williams MC. Hepatosteatosis and Atherosclerotic Plaque at Coronary CT Angiography. Radiol Cardiothorac Imaging 2022; 4:e210260. [PMID: 35506136 PMCID: PMC9059242 DOI: 10.1148/ryct.210260] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 03/01/2022] [Accepted: 03/29/2022] [Indexed: 01/22/2023]
Abstract
Purpose To assess the association between nonalcoholic fatty liver disease (NAFLD) and quantitative atherosclerotic plaque at CT. Materials and Methods In this post hoc analysis of the prospective Scottish Computed Tomography of the HEART trial (November 2010 to September 2014), hepatosteatosis and coronary artery calcium score were measured at noncontrast CT. Presence of stenoses, visually assessed high-risk plaque, and quantitative plaque burden were assessed at coronary CT angiography. Multivariable models were constructed to assess the impact of hepatosteatosis and cardiovascular risk factors on coronary artery disease. Results Images from 1726 participants (mean age, 58 years ± 9 [SD]; 974 men) were included. Participants with hepatosteatosis (155 of 1726, 9%) had a higher body mass index, more hypertension and diabetes mellitus, and higher cardiovascular risk scores (P < .001 for all) compared with those without hepatosteatosis. They had increased coronary artery calcium scores (median, 43 Agatston units [AU] [interquartile range, 0-273] vs 19 AU [0-225], P = .046), more nonobstructive disease (48% vs 37%, P = .02), and higher low-attenuation plaque burden (5.11% [0-7.16] vs 4.07% [0-6.84], P = .04). However, these associations were not independent of cardiovascular risk factors. Over a median of 4.7 years, there was no evidence of a difference in myocardial infarction between those with and without hepatosteatosis (1.9% vs 2.4%, P = .92). Conclusion Hepatosteatosis at CT was associated with an increased prevalence of coronary artery disease at CT, but this was not independent of the presence of cardiovascular risk factors.Keywords: CT, Cardiac, Nonalcoholic Fatty Liver Disease, Coronary Artery Disease, Hepatosteatosis, Plaque QuantificationClinical trial registration no. NCT01149590 Supplemental material is available for this article. © RSNA, 2022See also commentary by Abohashem and Blankstein in this issue.
Collapse
Affiliation(s)
- Jessica Carter
- From the University/BHF Centre for Cardiovascular Science, University
of Edinburgh, Chancellor’s Building, 49 Little France Crescent,
Edinburgh, Scotland EH16 SUF, (J.C., M.N.M., E.T., J.K., M.D., A.J.M., P.D.A.,
A.H., S.A., A.S.V.S., T.P., C.W., N.L.M., M.R.D., D.E.N., M.C.W.); Liverpool
Centre for Cardiovascular Science, Liverpool, England (T.D.H., S.W.M.);
Department of Interventional Cardiology and Angiology, Institute of Cardiology,
Warsaw, Poland (J.K.); Biomedical Imaging Research Institute and Division of
Artificial Intelligence in Medicine, Cedars-Sinai Medical Center, Los Angeles,
Calif (P.M., D.S.B., P.J.S., D.D.); Christchurch Heart Institute, University of
Otago, Christchurch, New Zealand (P.D.A.); Department of Radiology, University
of Cambridge, Cambridge, England (J.R.W.M.); Institute of Cardiovascular and
Medical Sciences, University of Glasgow, Glasgow, Scotland (G.R.); Edinburgh
Imaging Facility QMRI, University of Edinburgh, Edinburgh, Scotland (E.J.R.v.B.,
M.R.D., D.E.N., M.C.W.); Royal Brompton and Harefield NHS Foundation Trust
Departments of Cardiology and Radiology, London, England and the National Heart
and Lung Institute, Faculty of Medicine, Imperial College, London, England
(E.D.N.); and Icahn School of Medicine, Weill Cornell Medical College, New York,
NY (L.J.S.)
| | - Thomas D. Heseltine
- From the University/BHF Centre for Cardiovascular Science, University
of Edinburgh, Chancellor’s Building, 49 Little France Crescent,
Edinburgh, Scotland EH16 SUF, (J.C., M.N.M., E.T., J.K., M.D., A.J.M., P.D.A.,
A.H., S.A., A.S.V.S., T.P., C.W., N.L.M., M.R.D., D.E.N., M.C.W.); Liverpool
Centre for Cardiovascular Science, Liverpool, England (T.D.H., S.W.M.);
Department of Interventional Cardiology and Angiology, Institute of Cardiology,
Warsaw, Poland (J.K.); Biomedical Imaging Research Institute and Division of
Artificial Intelligence in Medicine, Cedars-Sinai Medical Center, Los Angeles,
Calif (P.M., D.S.B., P.J.S., D.D.); Christchurch Heart Institute, University of
Otago, Christchurch, New Zealand (P.D.A.); Department of Radiology, University
of Cambridge, Cambridge, England (J.R.W.M.); Institute of Cardiovascular and
Medical Sciences, University of Glasgow, Glasgow, Scotland (G.R.); Edinburgh
Imaging Facility QMRI, University of Edinburgh, Edinburgh, Scotland (E.J.R.v.B.,
M.R.D., D.E.N., M.C.W.); Royal Brompton and Harefield NHS Foundation Trust
Departments of Cardiology and Radiology, London, England and the National Heart
and Lung Institute, Faculty of Medicine, Imperial College, London, England
(E.D.N.); and Icahn School of Medicine, Weill Cornell Medical College, New York,
NY (L.J.S.)
| | - Mohammed N. Meah
- From the University/BHF Centre for Cardiovascular Science, University
of Edinburgh, Chancellor’s Building, 49 Little France Crescent,
Edinburgh, Scotland EH16 SUF, (J.C., M.N.M., E.T., J.K., M.D., A.J.M., P.D.A.,
A.H., S.A., A.S.V.S., T.P., C.W., N.L.M., M.R.D., D.E.N., M.C.W.); Liverpool
Centre for Cardiovascular Science, Liverpool, England (T.D.H., S.W.M.);
Department of Interventional Cardiology and Angiology, Institute of Cardiology,
Warsaw, Poland (J.K.); Biomedical Imaging Research Institute and Division of
Artificial Intelligence in Medicine, Cedars-Sinai Medical Center, Los Angeles,
Calif (P.M., D.S.B., P.J.S., D.D.); Christchurch Heart Institute, University of
Otago, Christchurch, New Zealand (P.D.A.); Department of Radiology, University
of Cambridge, Cambridge, England (J.R.W.M.); Institute of Cardiovascular and
Medical Sciences, University of Glasgow, Glasgow, Scotland (G.R.); Edinburgh
Imaging Facility QMRI, University of Edinburgh, Edinburgh, Scotland (E.J.R.v.B.,
M.R.D., D.E.N., M.C.W.); Royal Brompton and Harefield NHS Foundation Trust
Departments of Cardiology and Radiology, London, England and the National Heart
and Lung Institute, Faculty of Medicine, Imperial College, London, England
(E.D.N.); and Icahn School of Medicine, Weill Cornell Medical College, New York,
NY (L.J.S.)
| | - Evangelos Tzolos
- From the University/BHF Centre for Cardiovascular Science, University
of Edinburgh, Chancellor’s Building, 49 Little France Crescent,
Edinburgh, Scotland EH16 SUF, (J.C., M.N.M., E.T., J.K., M.D., A.J.M., P.D.A.,
A.H., S.A., A.S.V.S., T.P., C.W., N.L.M., M.R.D., D.E.N., M.C.W.); Liverpool
Centre for Cardiovascular Science, Liverpool, England (T.D.H., S.W.M.);
Department of Interventional Cardiology and Angiology, Institute of Cardiology,
Warsaw, Poland (J.K.); Biomedical Imaging Research Institute and Division of
Artificial Intelligence in Medicine, Cedars-Sinai Medical Center, Los Angeles,
Calif (P.M., D.S.B., P.J.S., D.D.); Christchurch Heart Institute, University of
Otago, Christchurch, New Zealand (P.D.A.); Department of Radiology, University
of Cambridge, Cambridge, England (J.R.W.M.); Institute of Cardiovascular and
Medical Sciences, University of Glasgow, Glasgow, Scotland (G.R.); Edinburgh
Imaging Facility QMRI, University of Edinburgh, Edinburgh, Scotland (E.J.R.v.B.,
M.R.D., D.E.N., M.C.W.); Royal Brompton and Harefield NHS Foundation Trust
Departments of Cardiology and Radiology, London, England and the National Heart
and Lung Institute, Faculty of Medicine, Imperial College, London, England
(E.D.N.); and Icahn School of Medicine, Weill Cornell Medical College, New York,
NY (L.J.S.)
| | - Jacek Kwiecinski
- From the University/BHF Centre for Cardiovascular Science, University
of Edinburgh, Chancellor’s Building, 49 Little France Crescent,
Edinburgh, Scotland EH16 SUF, (J.C., M.N.M., E.T., J.K., M.D., A.J.M., P.D.A.,
A.H., S.A., A.S.V.S., T.P., C.W., N.L.M., M.R.D., D.E.N., M.C.W.); Liverpool
Centre for Cardiovascular Science, Liverpool, England (T.D.H., S.W.M.);
Department of Interventional Cardiology and Angiology, Institute of Cardiology,
Warsaw, Poland (J.K.); Biomedical Imaging Research Institute and Division of
Artificial Intelligence in Medicine, Cedars-Sinai Medical Center, Los Angeles,
Calif (P.M., D.S.B., P.J.S., D.D.); Christchurch Heart Institute, University of
Otago, Christchurch, New Zealand (P.D.A.); Department of Radiology, University
of Cambridge, Cambridge, England (J.R.W.M.); Institute of Cardiovascular and
Medical Sciences, University of Glasgow, Glasgow, Scotland (G.R.); Edinburgh
Imaging Facility QMRI, University of Edinburgh, Edinburgh, Scotland (E.J.R.v.B.,
M.R.D., D.E.N., M.C.W.); Royal Brompton and Harefield NHS Foundation Trust
Departments of Cardiology and Radiology, London, England and the National Heart
and Lung Institute, Faculty of Medicine, Imperial College, London, England
(E.D.N.); and Icahn School of Medicine, Weill Cornell Medical College, New York,
NY (L.J.S.)
| | - Mhairi Doris
- From the University/BHF Centre for Cardiovascular Science, University
of Edinburgh, Chancellor’s Building, 49 Little France Crescent,
Edinburgh, Scotland EH16 SUF, (J.C., M.N.M., E.T., J.K., M.D., A.J.M., P.D.A.,
A.H., S.A., A.S.V.S., T.P., C.W., N.L.M., M.R.D., D.E.N., M.C.W.); Liverpool
Centre for Cardiovascular Science, Liverpool, England (T.D.H., S.W.M.);
Department of Interventional Cardiology and Angiology, Institute of Cardiology,
Warsaw, Poland (J.K.); Biomedical Imaging Research Institute and Division of
Artificial Intelligence in Medicine, Cedars-Sinai Medical Center, Los Angeles,
Calif (P.M., D.S.B., P.J.S., D.D.); Christchurch Heart Institute, University of
Otago, Christchurch, New Zealand (P.D.A.); Department of Radiology, University
of Cambridge, Cambridge, England (J.R.W.M.); Institute of Cardiovascular and
Medical Sciences, University of Glasgow, Glasgow, Scotland (G.R.); Edinburgh
Imaging Facility QMRI, University of Edinburgh, Edinburgh, Scotland (E.J.R.v.B.,
M.R.D., D.E.N., M.C.W.); Royal Brompton and Harefield NHS Foundation Trust
Departments of Cardiology and Radiology, London, England and the National Heart
and Lung Institute, Faculty of Medicine, Imperial College, London, England
(E.D.N.); and Icahn School of Medicine, Weill Cornell Medical College, New York,
NY (L.J.S.)
| | - Priscilla McElhinney
- From the University/BHF Centre for Cardiovascular Science, University
of Edinburgh, Chancellor’s Building, 49 Little France Crescent,
Edinburgh, Scotland EH16 SUF, (J.C., M.N.M., E.T., J.K., M.D., A.J.M., P.D.A.,
A.H., S.A., A.S.V.S., T.P., C.W., N.L.M., M.R.D., D.E.N., M.C.W.); Liverpool
Centre for Cardiovascular Science, Liverpool, England (T.D.H., S.W.M.);
Department of Interventional Cardiology and Angiology, Institute of Cardiology,
Warsaw, Poland (J.K.); Biomedical Imaging Research Institute and Division of
Artificial Intelligence in Medicine, Cedars-Sinai Medical Center, Los Angeles,
Calif (P.M., D.S.B., P.J.S., D.D.); Christchurch Heart Institute, University of
Otago, Christchurch, New Zealand (P.D.A.); Department of Radiology, University
of Cambridge, Cambridge, England (J.R.W.M.); Institute of Cardiovascular and
Medical Sciences, University of Glasgow, Glasgow, Scotland (G.R.); Edinburgh
Imaging Facility QMRI, University of Edinburgh, Edinburgh, Scotland (E.J.R.v.B.,
M.R.D., D.E.N., M.C.W.); Royal Brompton and Harefield NHS Foundation Trust
Departments of Cardiology and Radiology, London, England and the National Heart
and Lung Institute, Faculty of Medicine, Imperial College, London, England
(E.D.N.); and Icahn School of Medicine, Weill Cornell Medical College, New York,
NY (L.J.S.)
| | - Alastair J. Moss
- From the University/BHF Centre for Cardiovascular Science, University
of Edinburgh, Chancellor’s Building, 49 Little France Crescent,
Edinburgh, Scotland EH16 SUF, (J.C., M.N.M., E.T., J.K., M.D., A.J.M., P.D.A.,
A.H., S.A., A.S.V.S., T.P., C.W., N.L.M., M.R.D., D.E.N., M.C.W.); Liverpool
Centre for Cardiovascular Science, Liverpool, England (T.D.H., S.W.M.);
Department of Interventional Cardiology and Angiology, Institute of Cardiology,
Warsaw, Poland (J.K.); Biomedical Imaging Research Institute and Division of
Artificial Intelligence in Medicine, Cedars-Sinai Medical Center, Los Angeles,
Calif (P.M., D.S.B., P.J.S., D.D.); Christchurch Heart Institute, University of
Otago, Christchurch, New Zealand (P.D.A.); Department of Radiology, University
of Cambridge, Cambridge, England (J.R.W.M.); Institute of Cardiovascular and
Medical Sciences, University of Glasgow, Glasgow, Scotland (G.R.); Edinburgh
Imaging Facility QMRI, University of Edinburgh, Edinburgh, Scotland (E.J.R.v.B.,
M.R.D., D.E.N., M.C.W.); Royal Brompton and Harefield NHS Foundation Trust
Departments of Cardiology and Radiology, London, England and the National Heart
and Lung Institute, Faculty of Medicine, Imperial College, London, England
(E.D.N.); and Icahn School of Medicine, Weill Cornell Medical College, New York,
NY (L.J.S.)
| | - Philip D. Adamson
- From the University/BHF Centre for Cardiovascular Science, University
of Edinburgh, Chancellor’s Building, 49 Little France Crescent,
Edinburgh, Scotland EH16 SUF, (J.C., M.N.M., E.T., J.K., M.D., A.J.M., P.D.A.,
A.H., S.A., A.S.V.S., T.P., C.W., N.L.M., M.R.D., D.E.N., M.C.W.); Liverpool
Centre for Cardiovascular Science, Liverpool, England (T.D.H., S.W.M.);
Department of Interventional Cardiology and Angiology, Institute of Cardiology,
Warsaw, Poland (J.K.); Biomedical Imaging Research Institute and Division of
Artificial Intelligence in Medicine, Cedars-Sinai Medical Center, Los Angeles,
Calif (P.M., D.S.B., P.J.S., D.D.); Christchurch Heart Institute, University of
Otago, Christchurch, New Zealand (P.D.A.); Department of Radiology, University
of Cambridge, Cambridge, England (J.R.W.M.); Institute of Cardiovascular and
Medical Sciences, University of Glasgow, Glasgow, Scotland (G.R.); Edinburgh
Imaging Facility QMRI, University of Edinburgh, Edinburgh, Scotland (E.J.R.v.B.,
M.R.D., D.E.N., M.C.W.); Royal Brompton and Harefield NHS Foundation Trust
Departments of Cardiology and Radiology, London, England and the National Heart
and Lung Institute, Faculty of Medicine, Imperial College, London, England
(E.D.N.); and Icahn School of Medicine, Weill Cornell Medical College, New York,
NY (L.J.S.)
| | - Amanda Hunter
- From the University/BHF Centre for Cardiovascular Science, University
of Edinburgh, Chancellor’s Building, 49 Little France Crescent,
Edinburgh, Scotland EH16 SUF, (J.C., M.N.M., E.T., J.K., M.D., A.J.M., P.D.A.,
A.H., S.A., A.S.V.S., T.P., C.W., N.L.M., M.R.D., D.E.N., M.C.W.); Liverpool
Centre for Cardiovascular Science, Liverpool, England (T.D.H., S.W.M.);
Department of Interventional Cardiology and Angiology, Institute of Cardiology,
Warsaw, Poland (J.K.); Biomedical Imaging Research Institute and Division of
Artificial Intelligence in Medicine, Cedars-Sinai Medical Center, Los Angeles,
Calif (P.M., D.S.B., P.J.S., D.D.); Christchurch Heart Institute, University of
Otago, Christchurch, New Zealand (P.D.A.); Department of Radiology, University
of Cambridge, Cambridge, England (J.R.W.M.); Institute of Cardiovascular and
Medical Sciences, University of Glasgow, Glasgow, Scotland (G.R.); Edinburgh
Imaging Facility QMRI, University of Edinburgh, Edinburgh, Scotland (E.J.R.v.B.,
M.R.D., D.E.N., M.C.W.); Royal Brompton and Harefield NHS Foundation Trust
Departments of Cardiology and Radiology, London, England and the National Heart
and Lung Institute, Faculty of Medicine, Imperial College, London, England
(E.D.N.); and Icahn School of Medicine, Weill Cornell Medical College, New York,
NY (L.J.S.)
| | - Shirjel Alam
- From the University/BHF Centre for Cardiovascular Science, University
of Edinburgh, Chancellor’s Building, 49 Little France Crescent,
Edinburgh, Scotland EH16 SUF, (J.C., M.N.M., E.T., J.K., M.D., A.J.M., P.D.A.,
A.H., S.A., A.S.V.S., T.P., C.W., N.L.M., M.R.D., D.E.N., M.C.W.); Liverpool
Centre for Cardiovascular Science, Liverpool, England (T.D.H., S.W.M.);
Department of Interventional Cardiology and Angiology, Institute of Cardiology,
Warsaw, Poland (J.K.); Biomedical Imaging Research Institute and Division of
Artificial Intelligence in Medicine, Cedars-Sinai Medical Center, Los Angeles,
Calif (P.M., D.S.B., P.J.S., D.D.); Christchurch Heart Institute, University of
Otago, Christchurch, New Zealand (P.D.A.); Department of Radiology, University
of Cambridge, Cambridge, England (J.R.W.M.); Institute of Cardiovascular and
Medical Sciences, University of Glasgow, Glasgow, Scotland (G.R.); Edinburgh
Imaging Facility QMRI, University of Edinburgh, Edinburgh, Scotland (E.J.R.v.B.,
M.R.D., D.E.N., M.C.W.); Royal Brompton and Harefield NHS Foundation Trust
Departments of Cardiology and Radiology, London, England and the National Heart
and Lung Institute, Faculty of Medicine, Imperial College, London, England
(E.D.N.); and Icahn School of Medicine, Weill Cornell Medical College, New York,
NY (L.J.S.)
| | - Anoop S. V. Shah
- From the University/BHF Centre for Cardiovascular Science, University
of Edinburgh, Chancellor’s Building, 49 Little France Crescent,
Edinburgh, Scotland EH16 SUF, (J.C., M.N.M., E.T., J.K., M.D., A.J.M., P.D.A.,
A.H., S.A., A.S.V.S., T.P., C.W., N.L.M., M.R.D., D.E.N., M.C.W.); Liverpool
Centre for Cardiovascular Science, Liverpool, England (T.D.H., S.W.M.);
Department of Interventional Cardiology and Angiology, Institute of Cardiology,
Warsaw, Poland (J.K.); Biomedical Imaging Research Institute and Division of
Artificial Intelligence in Medicine, Cedars-Sinai Medical Center, Los Angeles,
Calif (P.M., D.S.B., P.J.S., D.D.); Christchurch Heart Institute, University of
Otago, Christchurch, New Zealand (P.D.A.); Department of Radiology, University
of Cambridge, Cambridge, England (J.R.W.M.); Institute of Cardiovascular and
Medical Sciences, University of Glasgow, Glasgow, Scotland (G.R.); Edinburgh
Imaging Facility QMRI, University of Edinburgh, Edinburgh, Scotland (E.J.R.v.B.,
M.R.D., D.E.N., M.C.W.); Royal Brompton and Harefield NHS Foundation Trust
Departments of Cardiology and Radiology, London, England and the National Heart
and Lung Institute, Faculty of Medicine, Imperial College, London, England
(E.D.N.); and Icahn School of Medicine, Weill Cornell Medical College, New York,
NY (L.J.S.)
| | - Tania Pawade
- From the University/BHF Centre for Cardiovascular Science, University
of Edinburgh, Chancellor’s Building, 49 Little France Crescent,
Edinburgh, Scotland EH16 SUF, (J.C., M.N.M., E.T., J.K., M.D., A.J.M., P.D.A.,
A.H., S.A., A.S.V.S., T.P., C.W., N.L.M., M.R.D., D.E.N., M.C.W.); Liverpool
Centre for Cardiovascular Science, Liverpool, England (T.D.H., S.W.M.);
Department of Interventional Cardiology and Angiology, Institute of Cardiology,
Warsaw, Poland (J.K.); Biomedical Imaging Research Institute and Division of
Artificial Intelligence in Medicine, Cedars-Sinai Medical Center, Los Angeles,
Calif (P.M., D.S.B., P.J.S., D.D.); Christchurch Heart Institute, University of
Otago, Christchurch, New Zealand (P.D.A.); Department of Radiology, University
of Cambridge, Cambridge, England (J.R.W.M.); Institute of Cardiovascular and
Medical Sciences, University of Glasgow, Glasgow, Scotland (G.R.); Edinburgh
Imaging Facility QMRI, University of Edinburgh, Edinburgh, Scotland (E.J.R.v.B.,
M.R.D., D.E.N., M.C.W.); Royal Brompton and Harefield NHS Foundation Trust
Departments of Cardiology and Radiology, London, England and the National Heart
and Lung Institute, Faculty of Medicine, Imperial College, London, England
(E.D.N.); and Icahn School of Medicine, Weill Cornell Medical College, New York,
NY (L.J.S.)
| | - Chengjia Wang
- From the University/BHF Centre for Cardiovascular Science, University
of Edinburgh, Chancellor’s Building, 49 Little France Crescent,
Edinburgh, Scotland EH16 SUF, (J.C., M.N.M., E.T., J.K., M.D., A.J.M., P.D.A.,
A.H., S.A., A.S.V.S., T.P., C.W., N.L.M., M.R.D., D.E.N., M.C.W.); Liverpool
Centre for Cardiovascular Science, Liverpool, England (T.D.H., S.W.M.);
Department of Interventional Cardiology and Angiology, Institute of Cardiology,
Warsaw, Poland (J.K.); Biomedical Imaging Research Institute and Division of
Artificial Intelligence in Medicine, Cedars-Sinai Medical Center, Los Angeles,
Calif (P.M., D.S.B., P.J.S., D.D.); Christchurch Heart Institute, University of
Otago, Christchurch, New Zealand (P.D.A.); Department of Radiology, University
of Cambridge, Cambridge, England (J.R.W.M.); Institute of Cardiovascular and
Medical Sciences, University of Glasgow, Glasgow, Scotland (G.R.); Edinburgh
Imaging Facility QMRI, University of Edinburgh, Edinburgh, Scotland (E.J.R.v.B.,
M.R.D., D.E.N., M.C.W.); Royal Brompton and Harefield NHS Foundation Trust
Departments of Cardiology and Radiology, London, England and the National Heart
and Lung Institute, Faculty of Medicine, Imperial College, London, England
(E.D.N.); and Icahn School of Medicine, Weill Cornell Medical College, New York,
NY (L.J.S.)
| | - Jonathan R. Weir-McCall
- From the University/BHF Centre for Cardiovascular Science, University
of Edinburgh, Chancellor’s Building, 49 Little France Crescent,
Edinburgh, Scotland EH16 SUF, (J.C., M.N.M., E.T., J.K., M.D., A.J.M., P.D.A.,
A.H., S.A., A.S.V.S., T.P., C.W., N.L.M., M.R.D., D.E.N., M.C.W.); Liverpool
Centre for Cardiovascular Science, Liverpool, England (T.D.H., S.W.M.);
Department of Interventional Cardiology and Angiology, Institute of Cardiology,
Warsaw, Poland (J.K.); Biomedical Imaging Research Institute and Division of
Artificial Intelligence in Medicine, Cedars-Sinai Medical Center, Los Angeles,
Calif (P.M., D.S.B., P.J.S., D.D.); Christchurch Heart Institute, University of
Otago, Christchurch, New Zealand (P.D.A.); Department of Radiology, University
of Cambridge, Cambridge, England (J.R.W.M.); Institute of Cardiovascular and
Medical Sciences, University of Glasgow, Glasgow, Scotland (G.R.); Edinburgh
Imaging Facility QMRI, University of Edinburgh, Edinburgh, Scotland (E.J.R.v.B.,
M.R.D., D.E.N., M.C.W.); Royal Brompton and Harefield NHS Foundation Trust
Departments of Cardiology and Radiology, London, England and the National Heart
and Lung Institute, Faculty of Medicine, Imperial College, London, England
(E.D.N.); and Icahn School of Medicine, Weill Cornell Medical College, New York,
NY (L.J.S.)
| | - Giles Roditi
- From the University/BHF Centre for Cardiovascular Science, University
of Edinburgh, Chancellor’s Building, 49 Little France Crescent,
Edinburgh, Scotland EH16 SUF, (J.C., M.N.M., E.T., J.K., M.D., A.J.M., P.D.A.,
A.H., S.A., A.S.V.S., T.P., C.W., N.L.M., M.R.D., D.E.N., M.C.W.); Liverpool
Centre for Cardiovascular Science, Liverpool, England (T.D.H., S.W.M.);
Department of Interventional Cardiology and Angiology, Institute of Cardiology,
Warsaw, Poland (J.K.); Biomedical Imaging Research Institute and Division of
Artificial Intelligence in Medicine, Cedars-Sinai Medical Center, Los Angeles,
Calif (P.M., D.S.B., P.J.S., D.D.); Christchurch Heart Institute, University of
Otago, Christchurch, New Zealand (P.D.A.); Department of Radiology, University
of Cambridge, Cambridge, England (J.R.W.M.); Institute of Cardiovascular and
Medical Sciences, University of Glasgow, Glasgow, Scotland (G.R.); Edinburgh
Imaging Facility QMRI, University of Edinburgh, Edinburgh, Scotland (E.J.R.v.B.,
M.R.D., D.E.N., M.C.W.); Royal Brompton and Harefield NHS Foundation Trust
Departments of Cardiology and Radiology, London, England and the National Heart
and Lung Institute, Faculty of Medicine, Imperial College, London, England
(E.D.N.); and Icahn School of Medicine, Weill Cornell Medical College, New York,
NY (L.J.S.)
| | - Edwin J. R. van Beek
- From the University/BHF Centre for Cardiovascular Science, University
of Edinburgh, Chancellor’s Building, 49 Little France Crescent,
Edinburgh, Scotland EH16 SUF, (J.C., M.N.M., E.T., J.K., M.D., A.J.M., P.D.A.,
A.H., S.A., A.S.V.S., T.P., C.W., N.L.M., M.R.D., D.E.N., M.C.W.); Liverpool
Centre for Cardiovascular Science, Liverpool, England (T.D.H., S.W.M.);
Department of Interventional Cardiology and Angiology, Institute of Cardiology,
Warsaw, Poland (J.K.); Biomedical Imaging Research Institute and Division of
Artificial Intelligence in Medicine, Cedars-Sinai Medical Center, Los Angeles,
Calif (P.M., D.S.B., P.J.S., D.D.); Christchurch Heart Institute, University of
Otago, Christchurch, New Zealand (P.D.A.); Department of Radiology, University
of Cambridge, Cambridge, England (J.R.W.M.); Institute of Cardiovascular and
Medical Sciences, University of Glasgow, Glasgow, Scotland (G.R.); Edinburgh
Imaging Facility QMRI, University of Edinburgh, Edinburgh, Scotland (E.J.R.v.B.,
M.R.D., D.E.N., M.C.W.); Royal Brompton and Harefield NHS Foundation Trust
Departments of Cardiology and Radiology, London, England and the National Heart
and Lung Institute, Faculty of Medicine, Imperial College, London, England
(E.D.N.); and Icahn School of Medicine, Weill Cornell Medical College, New York,
NY (L.J.S.)
| | - Edward D. Nicol
- From the University/BHF Centre for Cardiovascular Science, University
of Edinburgh, Chancellor’s Building, 49 Little France Crescent,
Edinburgh, Scotland EH16 SUF, (J.C., M.N.M., E.T., J.K., M.D., A.J.M., P.D.A.,
A.H., S.A., A.S.V.S., T.P., C.W., N.L.M., M.R.D., D.E.N., M.C.W.); Liverpool
Centre for Cardiovascular Science, Liverpool, England (T.D.H., S.W.M.);
Department of Interventional Cardiology and Angiology, Institute of Cardiology,
Warsaw, Poland (J.K.); Biomedical Imaging Research Institute and Division of
Artificial Intelligence in Medicine, Cedars-Sinai Medical Center, Los Angeles,
Calif (P.M., D.S.B., P.J.S., D.D.); Christchurch Heart Institute, University of
Otago, Christchurch, New Zealand (P.D.A.); Department of Radiology, University
of Cambridge, Cambridge, England (J.R.W.M.); Institute of Cardiovascular and
Medical Sciences, University of Glasgow, Glasgow, Scotland (G.R.); Edinburgh
Imaging Facility QMRI, University of Edinburgh, Edinburgh, Scotland (E.J.R.v.B.,
M.R.D., D.E.N., M.C.W.); Royal Brompton and Harefield NHS Foundation Trust
Departments of Cardiology and Radiology, London, England and the National Heart
and Lung Institute, Faculty of Medicine, Imperial College, London, England
(E.D.N.); and Icahn School of Medicine, Weill Cornell Medical College, New York,
NY (L.J.S.)
| | - Leslee J. Shaw
- From the University/BHF Centre for Cardiovascular Science, University
of Edinburgh, Chancellor’s Building, 49 Little France Crescent,
Edinburgh, Scotland EH16 SUF, (J.C., M.N.M., E.T., J.K., M.D., A.J.M., P.D.A.,
A.H., S.A., A.S.V.S., T.P., C.W., N.L.M., M.R.D., D.E.N., M.C.W.); Liverpool
Centre for Cardiovascular Science, Liverpool, England (T.D.H., S.W.M.);
Department of Interventional Cardiology and Angiology, Institute of Cardiology,
Warsaw, Poland (J.K.); Biomedical Imaging Research Institute and Division of
Artificial Intelligence in Medicine, Cedars-Sinai Medical Center, Los Angeles,
Calif (P.M., D.S.B., P.J.S., D.D.); Christchurch Heart Institute, University of
Otago, Christchurch, New Zealand (P.D.A.); Department of Radiology, University
of Cambridge, Cambridge, England (J.R.W.M.); Institute of Cardiovascular and
Medical Sciences, University of Glasgow, Glasgow, Scotland (G.R.); Edinburgh
Imaging Facility QMRI, University of Edinburgh, Edinburgh, Scotland (E.J.R.v.B.,
M.R.D., D.E.N., M.C.W.); Royal Brompton and Harefield NHS Foundation Trust
Departments of Cardiology and Radiology, London, England and the National Heart
and Lung Institute, Faculty of Medicine, Imperial College, London, England
(E.D.N.); and Icahn School of Medicine, Weill Cornell Medical College, New York,
NY (L.J.S.)
| | - Daniel S. Berman
- From the University/BHF Centre for Cardiovascular Science, University
of Edinburgh, Chancellor’s Building, 49 Little France Crescent,
Edinburgh, Scotland EH16 SUF, (J.C., M.N.M., E.T., J.K., M.D., A.J.M., P.D.A.,
A.H., S.A., A.S.V.S., T.P., C.W., N.L.M., M.R.D., D.E.N., M.C.W.); Liverpool
Centre for Cardiovascular Science, Liverpool, England (T.D.H., S.W.M.);
Department of Interventional Cardiology and Angiology, Institute of Cardiology,
Warsaw, Poland (J.K.); Biomedical Imaging Research Institute and Division of
Artificial Intelligence in Medicine, Cedars-Sinai Medical Center, Los Angeles,
Calif (P.M., D.S.B., P.J.S., D.D.); Christchurch Heart Institute, University of
Otago, Christchurch, New Zealand (P.D.A.); Department of Radiology, University
of Cambridge, Cambridge, England (J.R.W.M.); Institute of Cardiovascular and
Medical Sciences, University of Glasgow, Glasgow, Scotland (G.R.); Edinburgh
Imaging Facility QMRI, University of Edinburgh, Edinburgh, Scotland (E.J.R.v.B.,
M.R.D., D.E.N., M.C.W.); Royal Brompton and Harefield NHS Foundation Trust
Departments of Cardiology and Radiology, London, England and the National Heart
and Lung Institute, Faculty of Medicine, Imperial College, London, England
(E.D.N.); and Icahn School of Medicine, Weill Cornell Medical College, New York,
NY (L.J.S.)
| | - Piotr J. Slomka
- From the University/BHF Centre for Cardiovascular Science, University
of Edinburgh, Chancellor’s Building, 49 Little France Crescent,
Edinburgh, Scotland EH16 SUF, (J.C., M.N.M., E.T., J.K., M.D., A.J.M., P.D.A.,
A.H., S.A., A.S.V.S., T.P., C.W., N.L.M., M.R.D., D.E.N., M.C.W.); Liverpool
Centre for Cardiovascular Science, Liverpool, England (T.D.H., S.W.M.);
Department of Interventional Cardiology and Angiology, Institute of Cardiology,
Warsaw, Poland (J.K.); Biomedical Imaging Research Institute and Division of
Artificial Intelligence in Medicine, Cedars-Sinai Medical Center, Los Angeles,
Calif (P.M., D.S.B., P.J.S., D.D.); Christchurch Heart Institute, University of
Otago, Christchurch, New Zealand (P.D.A.); Department of Radiology, University
of Cambridge, Cambridge, England (J.R.W.M.); Institute of Cardiovascular and
Medical Sciences, University of Glasgow, Glasgow, Scotland (G.R.); Edinburgh
Imaging Facility QMRI, University of Edinburgh, Edinburgh, Scotland (E.J.R.v.B.,
M.R.D., D.E.N., M.C.W.); Royal Brompton and Harefield NHS Foundation Trust
Departments of Cardiology and Radiology, London, England and the National Heart
and Lung Institute, Faculty of Medicine, Imperial College, London, England
(E.D.N.); and Icahn School of Medicine, Weill Cornell Medical College, New York,
NY (L.J.S.)
| | - Nicholas L. Mills
- From the University/BHF Centre for Cardiovascular Science, University
of Edinburgh, Chancellor’s Building, 49 Little France Crescent,
Edinburgh, Scotland EH16 SUF, (J.C., M.N.M., E.T., J.K., M.D., A.J.M., P.D.A.,
A.H., S.A., A.S.V.S., T.P., C.W., N.L.M., M.R.D., D.E.N., M.C.W.); Liverpool
Centre for Cardiovascular Science, Liverpool, England (T.D.H., S.W.M.);
Department of Interventional Cardiology and Angiology, Institute of Cardiology,
Warsaw, Poland (J.K.); Biomedical Imaging Research Institute and Division of
Artificial Intelligence in Medicine, Cedars-Sinai Medical Center, Los Angeles,
Calif (P.M., D.S.B., P.J.S., D.D.); Christchurch Heart Institute, University of
Otago, Christchurch, New Zealand (P.D.A.); Department of Radiology, University
of Cambridge, Cambridge, England (J.R.W.M.); Institute of Cardiovascular and
Medical Sciences, University of Glasgow, Glasgow, Scotland (G.R.); Edinburgh
Imaging Facility QMRI, University of Edinburgh, Edinburgh, Scotland (E.J.R.v.B.,
M.R.D., D.E.N., M.C.W.); Royal Brompton and Harefield NHS Foundation Trust
Departments of Cardiology and Radiology, London, England and the National Heart
and Lung Institute, Faculty of Medicine, Imperial College, London, England
(E.D.N.); and Icahn School of Medicine, Weill Cornell Medical College, New York,
NY (L.J.S.)
| | - Marc R. Dweck
- From the University/BHF Centre for Cardiovascular Science, University
of Edinburgh, Chancellor’s Building, 49 Little France Crescent,
Edinburgh, Scotland EH16 SUF, (J.C., M.N.M., E.T., J.K., M.D., A.J.M., P.D.A.,
A.H., S.A., A.S.V.S., T.P., C.W., N.L.M., M.R.D., D.E.N., M.C.W.); Liverpool
Centre for Cardiovascular Science, Liverpool, England (T.D.H., S.W.M.);
Department of Interventional Cardiology and Angiology, Institute of Cardiology,
Warsaw, Poland (J.K.); Biomedical Imaging Research Institute and Division of
Artificial Intelligence in Medicine, Cedars-Sinai Medical Center, Los Angeles,
Calif (P.M., D.S.B., P.J.S., D.D.); Christchurch Heart Institute, University of
Otago, Christchurch, New Zealand (P.D.A.); Department of Radiology, University
of Cambridge, Cambridge, England (J.R.W.M.); Institute of Cardiovascular and
Medical Sciences, University of Glasgow, Glasgow, Scotland (G.R.); Edinburgh
Imaging Facility QMRI, University of Edinburgh, Edinburgh, Scotland (E.J.R.v.B.,
M.R.D., D.E.N., M.C.W.); Royal Brompton and Harefield NHS Foundation Trust
Departments of Cardiology and Radiology, London, England and the National Heart
and Lung Institute, Faculty of Medicine, Imperial College, London, England
(E.D.N.); and Icahn School of Medicine, Weill Cornell Medical College, New York,
NY (L.J.S.)
| | - David E. Newby
- From the University/BHF Centre for Cardiovascular Science, University
of Edinburgh, Chancellor’s Building, 49 Little France Crescent,
Edinburgh, Scotland EH16 SUF, (J.C., M.N.M., E.T., J.K., M.D., A.J.M., P.D.A.,
A.H., S.A., A.S.V.S., T.P., C.W., N.L.M., M.R.D., D.E.N., M.C.W.); Liverpool
Centre for Cardiovascular Science, Liverpool, England (T.D.H., S.W.M.);
Department of Interventional Cardiology and Angiology, Institute of Cardiology,
Warsaw, Poland (J.K.); Biomedical Imaging Research Institute and Division of
Artificial Intelligence in Medicine, Cedars-Sinai Medical Center, Los Angeles,
Calif (P.M., D.S.B., P.J.S., D.D.); Christchurch Heart Institute, University of
Otago, Christchurch, New Zealand (P.D.A.); Department of Radiology, University
of Cambridge, Cambridge, England (J.R.W.M.); Institute of Cardiovascular and
Medical Sciences, University of Glasgow, Glasgow, Scotland (G.R.); Edinburgh
Imaging Facility QMRI, University of Edinburgh, Edinburgh, Scotland (E.J.R.v.B.,
M.R.D., D.E.N., M.C.W.); Royal Brompton and Harefield NHS Foundation Trust
Departments of Cardiology and Radiology, London, England and the National Heart
and Lung Institute, Faculty of Medicine, Imperial College, London, England
(E.D.N.); and Icahn School of Medicine, Weill Cornell Medical College, New York,
NY (L.J.S.)
| | - Scott W. Murray
- From the University/BHF Centre for Cardiovascular Science, University
of Edinburgh, Chancellor’s Building, 49 Little France Crescent,
Edinburgh, Scotland EH16 SUF, (J.C., M.N.M., E.T., J.K., M.D., A.J.M., P.D.A.,
A.H., S.A., A.S.V.S., T.P., C.W., N.L.M., M.R.D., D.E.N., M.C.W.); Liverpool
Centre for Cardiovascular Science, Liverpool, England (T.D.H., S.W.M.);
Department of Interventional Cardiology and Angiology, Institute of Cardiology,
Warsaw, Poland (J.K.); Biomedical Imaging Research Institute and Division of
Artificial Intelligence in Medicine, Cedars-Sinai Medical Center, Los Angeles,
Calif (P.M., D.S.B., P.J.S., D.D.); Christchurch Heart Institute, University of
Otago, Christchurch, New Zealand (P.D.A.); Department of Radiology, University
of Cambridge, Cambridge, England (J.R.W.M.); Institute of Cardiovascular and
Medical Sciences, University of Glasgow, Glasgow, Scotland (G.R.); Edinburgh
Imaging Facility QMRI, University of Edinburgh, Edinburgh, Scotland (E.J.R.v.B.,
M.R.D., D.E.N., M.C.W.); Royal Brompton and Harefield NHS Foundation Trust
Departments of Cardiology and Radiology, London, England and the National Heart
and Lung Institute, Faculty of Medicine, Imperial College, London, England
(E.D.N.); and Icahn School of Medicine, Weill Cornell Medical College, New York,
NY (L.J.S.)
| | - Damini Dey
- From the University/BHF Centre for Cardiovascular Science, University
of Edinburgh, Chancellor’s Building, 49 Little France Crescent,
Edinburgh, Scotland EH16 SUF, (J.C., M.N.M., E.T., J.K., M.D., A.J.M., P.D.A.,
A.H., S.A., A.S.V.S., T.P., C.W., N.L.M., M.R.D., D.E.N., M.C.W.); Liverpool
Centre for Cardiovascular Science, Liverpool, England (T.D.H., S.W.M.);
Department of Interventional Cardiology and Angiology, Institute of Cardiology,
Warsaw, Poland (J.K.); Biomedical Imaging Research Institute and Division of
Artificial Intelligence in Medicine, Cedars-Sinai Medical Center, Los Angeles,
Calif (P.M., D.S.B., P.J.S., D.D.); Christchurch Heart Institute, University of
Otago, Christchurch, New Zealand (P.D.A.); Department of Radiology, University
of Cambridge, Cambridge, England (J.R.W.M.); Institute of Cardiovascular and
Medical Sciences, University of Glasgow, Glasgow, Scotland (G.R.); Edinburgh
Imaging Facility QMRI, University of Edinburgh, Edinburgh, Scotland (E.J.R.v.B.,
M.R.D., D.E.N., M.C.W.); Royal Brompton and Harefield NHS Foundation Trust
Departments of Cardiology and Radiology, London, England and the National Heart
and Lung Institute, Faculty of Medicine, Imperial College, London, England
(E.D.N.); and Icahn School of Medicine, Weill Cornell Medical College, New York,
NY (L.J.S.)
| | - Michelle C. Williams
- From the University/BHF Centre for Cardiovascular Science, University
of Edinburgh, Chancellor’s Building, 49 Little France Crescent,
Edinburgh, Scotland EH16 SUF, (J.C., M.N.M., E.T., J.K., M.D., A.J.M., P.D.A.,
A.H., S.A., A.S.V.S., T.P., C.W., N.L.M., M.R.D., D.E.N., M.C.W.); Liverpool
Centre for Cardiovascular Science, Liverpool, England (T.D.H., S.W.M.);
Department of Interventional Cardiology and Angiology, Institute of Cardiology,
Warsaw, Poland (J.K.); Biomedical Imaging Research Institute and Division of
Artificial Intelligence in Medicine, Cedars-Sinai Medical Center, Los Angeles,
Calif (P.M., D.S.B., P.J.S., D.D.); Christchurch Heart Institute, University of
Otago, Christchurch, New Zealand (P.D.A.); Department of Radiology, University
of Cambridge, Cambridge, England (J.R.W.M.); Institute of Cardiovascular and
Medical Sciences, University of Glasgow, Glasgow, Scotland (G.R.); Edinburgh
Imaging Facility QMRI, University of Edinburgh, Edinburgh, Scotland (E.J.R.v.B.,
M.R.D., D.E.N., M.C.W.); Royal Brompton and Harefield NHS Foundation Trust
Departments of Cardiology and Radiology, London, England and the National Heart
and Lung Institute, Faculty of Medicine, Imperial College, London, England
(E.D.N.); and Icahn School of Medicine, Weill Cornell Medical College, New York,
NY (L.J.S.)
| |
Collapse
|
65
|
Koo BK, Lim S. Metabolic Syndrome and Metabolic Dysfunction‐Associated Fatty Liver Disease. CLINICAL OBESITY IN ADULTS AND CHILDREN 2022:159-177. [DOI: 10.1002/9781119695257.ch13] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
66
|
Ale-Ebrahim M, Rahmani R, Faryabi K, Mohammadifar N, Mortazavi P, Karkhaneh L. Atheroprotective and hepatoprotective effects of trans-chalcone through modification of eNOS/AMPK/KLF-2 pathway and regulation of COX-2, Ang-II, and PDGF mRNA expression in NMRI mice fed HCD. Mol Biol Rep 2022; 49:3433-3443. [PMID: 35190927 DOI: 10.1007/s11033-022-07174-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Accepted: 01/19/2022] [Indexed: 02/06/2023]
Abstract
BACKGROUND The effects of trans-chalcone on atherosclerosis and NAFLD have been investigated. However, the underlying molecular mechanisms of these effects are not completely understood. This study aimed to deduce the impacts of trans-chalcone on the eNOS/AMPK/KLF-2 pathway in the heart tissues and the expression of Ang-II, PDFG, and COX-2 genes in liver sections of NMRI mice fed HCD. METHODS AND RESULTS Thirty-two male mice were divided into four groups (n = 8): control group; fed normal diet. HCD group; fed HCD (consisted of 2% cholesterol) (12 weeks). TCh groups; received HCD (12 weeks) besides co-treated with trans-chalcone (20 mg/kg and 40 mg/kg b.w. dosages respectively) for 4 weeks. Finally, the blood samples were collected to evaluate the biochemical parameters. Histopathological observations of aorta and liver sections were performed by H&E staining. The real-time PCR method was used for assessing the expression of the aforementioned genes. Histopathological examination demonstrated atheroma plaque formation and fatty liver in mice fed HCD which were accomplished with alteration in biochemical factors and Real-time PCR outcomes. Administration of trans-chalcone significantly modulated the serum of biochemical parameters. These effects were accompanied by significant increasing the expression of eNOS, AMPK, KLF-2 genes in heart sections and significant decrease in COX-2, Ang-II, and PDGF mRNA expression in liver sections. CONCLUSION Our findings propose that the atheroprotective and hepatoprotective effects of trans-chalcone may be attributed to the activation of the eNOS/AMPK/KLF-2 pathway and down-regulation of Ang-II, PDFG, and COX-2 genes, respectively.
Collapse
Affiliation(s)
- Mahsa Ale-Ebrahim
- Department of Physiology, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Raziyeh Rahmani
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Kousar Faryabi
- Department of Microbiology, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Niloofar Mohammadifar
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Pejman Mortazavi
- Department of Veterinary Pathology, Faculty of Specialized Veterinary, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Leyla Karkhaneh
- Department of Physiology, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| |
Collapse
|
67
|
Maevskaya M, Kotovskaya Y, Ivashkin V, Tkacheva O, Troshina E, Shestakova M, Breder V, Geyvandova N, Doschitsin V, Dudinskaya E, Ershova E, Kodzoeva K, Komshilova K, Korochanskaya N, Mayorov A, Mishina E, Nadinskaya M, Nikitin I, Pogosova N, Tarzimanova A, Shamkhalova M. The National Consensus statement on the management of adult patients with non-alcoholic fatty liver disease and main comorbidities. TERAPEVT ARKH 2022; 94:216-253. [PMID: 36286746 DOI: 10.26442/00403660.2022.02.201363] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Indexed: 12/15/2022]
Abstract
The National Consensus was prepared with the participation of the National Medical Association for the Study of the Multimorbidity, Russian Scientific Liver Society, Russian Association of Endocrinologists, Russian Association of Gerontologists and Geriatricians, National Society for Preventive Cardiology, Professional Foundation for the Promotion of Medicine Fund PROFMEDFORUM.
The aim of the multidisciplinary consensus is a detailed analysis of the course of non-alcoholic fatty liver disease (NAFLD) and the main associated conditions. The definition of NAFLD is given, its prevalence is described, methods for diagnosing its components such as steatosis, inflammation and fibrosis are described.
The association of NAFLD with a number of cardio-metabolic diseases (arterial hypertension, atherosclerosis, thrombotic complications, type 2 diabetes mellitus (T2DM), obesity, dyslipidemia, etc.), chronic kidney disease (CKD) and the risk of developing hepatocellular cancer (HCC) were analyzed. The review of non-drug methods of treatment of NAFLD and modern opportunities of pharmacotherapy are presented.
The possibilities of new molecules in the treatment of NAFLD are considered: agonists of nuclear receptors, antagonists of pro-inflammatory molecules, etc. The positive properties and disadvantages of currently used drugs (vitamin E, thiazolidinediones, etc.) are described. Special attention is paid to the multi-target ursodeoxycholic acid (UDCA) molecule in the complex treatment of NAFLD as a multifactorial disease. Its anti-inflammatory, anti-oxidant and cytoprotective properties, the ability to reduce steatosis an independent risk factor for the development of cardiovascular pathology, reduce inflammation and hepatic fibrosis through the modulation of autophagy are considered.
The ability of UDCA to influence glucose and lipid homeostasis and to have an anticarcinogenic effect has been demonstrated. The Consensus statement has advanced provisions for practitioners to optimize the diagnosis and treatment of NAFLD and related common pathogenetic links of cardio-metabolic diseases.
Collapse
|
68
|
Dalbeni A, Castelli M, Zoncapè M, Minuz P, Sacerdoti D. Platelets in Non-alcoholic Fatty Liver Disease. Front Pharmacol 2022; 13:842636. [PMID: 35250588 PMCID: PMC8895200 DOI: 10.3389/fphar.2022.842636] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 02/03/2022] [Indexed: 12/17/2022] Open
Abstract
Non alcoholic steatohepatitis (NASH) is the inflammatory reaction of the liver to excessive accumulation of lipids in the hepatocytes. NASH can progress to cirrhosis and hepatocellular carcinoma (HCC). Fatty liver is the hepatic manifestation of metabolic syndrome. A subclinical inflammatory state is present in patients with metabolic alterations like insulin resistance, type-2 diabetes, obesity, hyperlipidemia, and hypertension. Platelets participate in immune cells recruitment and cytokines-induced liver damage. It is hypothesized that lipid toxicity cause accumulation of platelets in the liver, platelet adhesion and activation, which primes the immunoinflammatory reaction and activation of stellate cells. Recent data suggest that antiplatelet drugs may interrupt this cascade and prevent/improve NASH. They may also improve some metabolic alterations. The pathophysiology of inflammatory liver disease and the implication of platelets are discussed in details.
Collapse
Affiliation(s)
- Andrea Dalbeni
- Division of General Medicine C, Department of Medicine, University and Azienda Ospedaliera Universitaria Integrata of Verona, Verona, Italy
- Liver Unit, Department of Medicine, University and Azienda Ospedaliera Universitaria Integrata of Verona, Verona, Italy
| | - Marco Castelli
- Division of General Medicine C, Department of Medicine, University and Azienda Ospedaliera Universitaria Integrata of Verona, Verona, Italy
| | - Mirko Zoncapè
- Division of General Medicine C, Department of Medicine, University and Azienda Ospedaliera Universitaria Integrata of Verona, Verona, Italy
- Liver Unit, Department of Medicine, University and Azienda Ospedaliera Universitaria Integrata of Verona, Verona, Italy
| | - Pietro Minuz
- Division of General Medicine C, Department of Medicine, University and Azienda Ospedaliera Universitaria Integrata of Verona, Verona, Italy
- *Correspondence: Pietro Minuz,
| | - David Sacerdoti
- Liver Unit, Department of Medicine, University and Azienda Ospedaliera Universitaria Integrata of Verona, Verona, Italy
| |
Collapse
|
69
|
Mingrone G, van Baar AC, Devière J, Hopkins D, Moura E, Cercato C, Rajagopalan H, Lopez-Talavera JC, White K, Bhambhani V, Costamagna G, Haidry R, Grecco E, Galvao Neto M, Aithal G, Repici A, Hayee B, Haji A, Morris AJ, Bisschops R, Chouhan MD, Sakai NS, Bhatt DL, Sanyal AJ, Bergman JJGHM. Safety and efficacy of hydrothermal duodenal mucosal resurfacing in patients with type 2 diabetes: the randomised, double-blind, sham-controlled, multicentre REVITA-2 feasibility trial. Gut 2022; 71:254-264. [PMID: 33597157 PMCID: PMC8761999 DOI: 10.1136/gutjnl-2020-323608] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 12/26/2020] [Accepted: 01/30/2021] [Indexed: 12/19/2022]
Abstract
OBJECTIVE Hydrothermal duodenal mucosal resurfacing (DMR) is a safe, outpatient endoscopic procedure. REVITA-2, a double-blind, superiority randomised controlled trial, investigates safety and efficacy of DMR using the single catheter Revita system (Revita DMR (catheter and system)), on glycaemic control and liver fat content in type 2 diabetes (T2D). DESIGN Eligible patients (haemoglobin A1c (HbA1c) 59-86 mmol/mol, body mass index≥24 and ≤40 kg/m2, fasting insulin >48.6 pmol/L, ≥1 oral antidiabetic medication) enrolled in Europe and Brazil. Primary endpoints were safety, change from baseline in HbA1c at 24 weeks, and liver MRI proton-density fat fraction (MRI-PDFF) at 12 weeks. RESULTS Overall mITT (DMR n=56; sham n=52), 24 weeks post DMR, median (IQR) HbA1c change was -10.4 (18.6) mmol/mol in DMR group versus -7.1 (16.4) mmol/mol in sham group (p=0.147). In patients with baseline liver MRI-PDFF >5% (DMR n=48; sham n=43), 12-week post-DMR liver-fat change was -5.4 (5.6)% in DMR group versus -2.9 (6.2)% in sham group (p=0.096). Results from prespecified interaction testing and clinical parameter assessment showed heterogeneity between European (DMR n=39; sham n=37) and Brazilian (DMR n=17; sham n=16) populations (p=0.063); therefore, results were stratified by region. In European mITT, 24 weeks post DMR, median (IQR) HbA1c change was -6.6 mmol/mol (17.5 mmol/mol) versus -3.3 mmol/mol (10.9 mmol/mol) post-sham (p=0.033); 12-week post-DMR liver-fat change was -5.4% (6.1%) versus -2.2% (4.3%) post-sham (p=0.035). Brazilian mITT results trended towards DMR benefit in HbA1c, but not liver fat, in context of a large sham effect. In overall PP, patients with high baseline fasting plasma glucose ((FPG)≥10 mmol/L) had significantly greater reductions in HbA1c post-DMR versus sham (p=0.002). Most adverse events were mild and transient. CONCLUSIONS DMR is safe and exerts beneficial disease-modifying metabolic effects in T2D with or without non-alcoholic liver disease, particularly in patients with high FPG. TRIAL REGISTRATION NUMBER NCT02879383.
Collapse
Affiliation(s)
- Geltrude Mingrone
- Division of Obesity and Metabolic Diseases, Fondazione Policlinico Universitario A. Gemelli IRCCS-Università Cattolica del Sacro Cuore, Rome, Italy
- Department of Diabetes, School of Life Course Sciences, King's College London, London, UK
| | - Annieke Cg van Baar
- Departments of Gastroenterology and Hepatology, Academic Medical Center Amsterdam, Amsterdam, The Netherlands
| | - Jacques Devière
- Department of Gastroenterology, Erasme University Hospital, Brussels, Belgium
| | - David Hopkins
- Institute of Diabetes, Endocrinology and Obesity, King's Health Partners, London, UK
| | - Eduardo Moura
- Department of Gastroenterology, Universidade de Sao Paulo Medical School, Sao Paulo, Brazil
| | - Cintia Cercato
- Obesity Unit, Department of Endocrinology, Hospital das Clínicas, University of Sao Paulo, Sao Paulo, Brazil
| | - Harith Rajagopalan
- Research and Development, Fractyl Laboratories, Inc, Lexington, Massachusetts, USA
| | | | - Kelly White
- Research and Development, Fractyl Laboratories, Inc, Lexington, Massachusetts, USA
| | - Vijeta Bhambhani
- Research and Development, Fractyl Laboratories, Inc, Lexington, Massachusetts, USA
| | - Guido Costamagna
- Digestive Endoscopy Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS-Università Cattolica del Sacro Cuore, Rome, Italy
| | - Rehan Haidry
- Department of Gastroenterology, King's College Hospital, London, UK
| | - Eduardo Grecco
- Department of Surgery, ABC University, Medical School, São Paulo, Brazil
| | - Manoel Galvao Neto
- Department of Surgery, ABC University, Medical School, São Paulo, Brazil
| | - Guruprasad Aithal
- NIHR Nottingham Biomedical Research Centre, Ottingham University Hospitals NHS Trust and the University of Nottingham, Nottingham, UK
| | - Alessandro Repici
- Humanitas Clinical and Research Center - IRCCS, Rozzano, Italy
- Humanitas University, Department of Biomedical Sciences, Milan, Italy
| | - Bu'Hussain Hayee
- Department of Gastroenterology, King's College Hospital, London, UK
| | - Amyn Haji
- King's Institute of Therapeutic Endoscopy, King's College Hospital, London, UK
| | - A John Morris
- Department of Gastroenterology, Glasgow Royal Infirmary, Glasgow, UK
| | - Raf Bisschops
- Gastroenterology Department, University of Leuven, Leuven, Belgium
| | - Manil D Chouhan
- Division of Medicine, University College London Center for Medical Imaging, London, UK
| | - Naomi S Sakai
- Division of Medicine, University College London Center for Medical Imaging, London, UK
| | - Deepak L Bhatt
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Arun J Sanyal
- Division of Gastroenterology, Hepatology and Nutrition, Department of Internal Medicine, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA
| | - J J G H M Bergman
- Departments of Gastroenterology and Hepatology, Academic Medical Center Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
70
|
Nelidova AV, Livzan MA, Nikolaev NA, Krolevets TS. Cardiovascular Diseases and Non-Alcoholic Fatty Liver Disease: Relationship and Pathogenetic Aspects of Pharmacotherapy. RATIONAL PHARMACOTHERAPY IN CARDIOLOGY 2022; 17:880-888. [DOI: 10.20996/1819-6446-2021-12-14] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/17/2024] Open
Abstract
The association of non-alcoholic fatty liver disease (NAFLD) and cardiovascular risk is currently one of the actively studied areas. The incidence of non-alcoholic fatty liver disease continues to grow worldwide. In the structure of mortality rate of patients with non-alcoholic fatty liver disease, the first place is occupied by cardiovascular events: stroke and myocardial infarction. Studies have shown that the presence of severe liver fibrosis (F3-4) in NAFLD not only increases the risk of cardiovascular diseases (CVD), but also increases the risk of overall mortality by 69% due to mortality from cardiovascular causes. The degree of increased risk is associated with the degree of activity of non-alcoholic steatohepatitis (NASH). Despite the large number of works on this topic, we do not have a clear opinion on the impact on cardiovascular risk, interaction and the contribution of various factors, as well as algorithms for managing patients with non-alcoholic fatty liver disease to reduce the risk of cardiovascular diseases. This article describes the pathogenetic factors of formation of cardiovascular risks in patients with non-alcoholic fatty liver disease, proposed the idea of stratification of cardiovascular risks in these patients, taking into account changes in the structure of the liver (fibrosis) and function (clinical and biochemical activity) and also it describes the main directions of drug therapy, taking into account the common pathogenetic mechanisms for non-alcoholic fatty liver disease and cardiovascular diseases. The role of obesity, local fat depots, adipokines, and endothelial dysfunction as the leading pathogenetic factors of increased cardiovascular risk in patients with NAFLD is discussed. Among pathogenetically justified drugs in conditions of poly and comorbidity, hypolipidemic (statins, fibrates), angiotensin II receptor antagonists, beta-blockers, etc. can be considered. According to numerous studies, it becomes obvious that the assessment of cardiovascular risks in patients with NAFLD will probably allow prescribing cardiological drugs, selecting individualized therapy regimens, taking into account the form of NAFLD, and on the other hand, building curation taking into account the identified cardiovascular risks.
Collapse
|
71
|
Sharma D, Gotlieb N, Farkouh ME, Patel K, Xu W, Bhat M. Machine Learning Approach to Classify Cardiovascular Disease in Patients With Nonalcoholic Fatty Liver Disease in the UK Biobank Cohort. J Am Heart Assoc 2022; 11:e022576. [PMID: 34927450 PMCID: PMC9075189 DOI: 10.1161/jaha.121.022576] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Accepted: 11/12/2021] [Indexed: 12/18/2022]
Abstract
Background Nonalcoholic fatty liver disease (NAFLD) is the most prevalent liver disease worldwide. Cardiovascular disease (CVD) is the leading cause of mortality among patients with NAFLD. The aim of our study was to develop a machine learning algorithm integrating clinical, lifestyle, and genetic risk factors to identify CVD in patients with NAFLD. Methods and Results We created a cohort of patients with NAFLD from the UK Biobank, diagnosed according to proton density fat fraction from magnetic resonance imaging data sets. A total of 400 patients with NAFLD with subclinical atherosclerosis or clinical CVD, defined by disease codes, constituted cases and 446 NAFLD cases with no CVD constituted controls. We evaluated 7 different supervised machine learning approaches on clinical, lifestyle, and genetic variables for identifying CVD in patients with NAFLD. The most significant clinical and lifestyle variables observed by the predictive modeling were age (59 years [54.00-63.00 years]), hypertension (145 mm Hg [134.0-156.0 mm Hg] and 85 mm Hg [79.00-93.00 mm Hg]), waist circumference (98 cm [95.00-105.00 cm]), and sedentary lifestyle, defined as time spent watching TV >4 h/d. In the genetic data, single-nucleotide polymorphisms in IL16 and ANKLE1 gene were most significant. Our proposed ensemble-based integrative machine learning model achieved an area under the curve of 0.849 using the random forest modeling for CVD prediction. Conclusions We propose a machine learning algorithm that identifies CVD in patients with NAFLD through integration of significant clinical, lifestyle, and genetic risk factors. These patients with NAFLD at higher risk of CVD should be flagged for screening and aggressive treatment of their cardiometabolic risk factors to prevent cardiovascular morbidity and mortality.
Collapse
Affiliation(s)
- Divya Sharma
- Department of BiostatisticsPrincess Margaret Cancer CentreUniversity Health NetworkTorontoOntarioCanada
| | - Neta Gotlieb
- Division of Adult GastroenterologyUniversity Health NetworkToronto General HospitalTorontoOntarioCanada
| | - Michael E. Farkouh
- Peter Munk Cardiac Centre, Heart and Stroke Richard Lewar CentreUniversity of TorontoOntarioCanada
| | - Keyur Patel
- Division of GastroenterologyUniversity Health NetworkToronto General HospitalTorontoOntarioCanada
| | - Wei Xu
- Department of BiostatisticsPrincess Margaret Cancer CentreUniversity Health NetworkTorontoOntarioCanada
- Biostatistics DivisionDalla Lana School of Public HealthUniversity of TorontoOntarioCanada
| | - Mamatha Bhat
- Department of MedicineMulti‐Organ Transplant ProgramToronto General HospitalTorontoOntarioCanada
| |
Collapse
|
72
|
Weinstein G, O’Donnell A, Davis-Plourde K, Zelber-Sagi S, Ghosh S, DeCarli CS, Thibault EG, Sperling RA, Johnson KA, Beiser AS, Seshadri S. Non-Alcoholic Fatty Liver Disease, Liver Fibrosis, and Regional Amyloid-β and Tau Pathology in Middle-Aged Adults: The Framingham Study. J Alzheimers Dis 2022; 86:1371-1383. [PMID: 35213373 PMCID: PMC11323287 DOI: 10.3233/jad-215409] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND Liver steatosis and fibrosis are emerging as risk factors for multiple extrahepatic health conditions; however, their relationship with Alzheimer's disease pathology is unclear. OBJECTIVE To examine whether non-alcoholic fatty liver disease (NAFLD) and FIB-4, a non-invasive index of advanced fibrosis, are associated with brain amyloid-β (Aβ) and tau pathology. METHODS The study sample included Framingham Study participants from the Offspring and Third generation cohorts who attended exams 9 (2011-2014) and 2 (2008-2011), respectively. Participants underwent 11C-Pittsburgh Compound-B amyloid and 18F-Flortaucipir tau positron emission tomography (PET) imaging and abdomen computed tomography, or had information on all components of the FIB-4 index. Linear regression models were used to assess the relationship of NAFLD and FIB-4 with regional tau and Aβ, adjusting for potential confounders and multiple comparisons. RESULTS Of the subsample with NAFLD information (N = 169; mean age 52±9 y; 57% males), 57 (34%) had NAFLD. Of the subsample with information on liver fibrosis (N = 177; mean age 50±10 y; 51% males), 34 (19%) had advanced fibrosis (FIB-4 > 1.3). Prevalent NAFLD was not associated with Aβ or tau PET. However, FIB-4 index was significantly associated with increased rhinal tau (β= 1.03±0.33, p = 0.002). Among individuals with prevalent NAFLD, FIB-4 was related to inferior temporal, parahippocampal gyrus, entorhinal and rhinal tau (β= 2.01±0.47, p < 0.001; β= 1.60±0.53, p = 0.007, and β= 1.59±0.47, p = 0.003 and β= 1.60±0.42, p = 0.001, respectively) and to Aβ deposition overall and in the inferior temporal and parahippocampal regions (β= 1.93±0.47, p < 0.001; β= 1.59±0.38, p < 0.001, and β= 1.52±0.54, p = 0.008, respectively). CONCLUSION This study suggests a possible association between liver fibrosis and early Alzheimer's disease pathology, independently of cardio-metabolic risk factors.
Collapse
Affiliation(s)
| | - Adrienne O’Donnell
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
- The Framingham Study, Framingham, MA, USA
| | - Kendra Davis-Plourde
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
- The Framingham Study, Framingham, MA, USA
| | - Shira Zelber-Sagi
- School of Public Health, University of Haifa, Haifa, Israel
- Liver Unit, Department of Gastroenterology, Tel-Aviv Medical Center, Tel-Aviv, Israel
| | - Saptaparni Ghosh
- The Framingham Study, Framingham, MA, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA
| | - Charles S. DeCarli
- Department of Neurology, School of Medicine & Imaging of Dementia and Aging Laboratory, Center for Neuroscience, University of California Davis, Davis, CA, USA
| | - Emma G. Thibault
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Reisa A. Sperling
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Keith A. Johnson
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Alexa S. Beiser
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
- The Framingham Study, Framingham, MA, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA
| | - Sudha Seshadri
- The Framingham Study, Framingham, MA, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA
- Glenn Biggs Institute for Alzheimer’s and Neurodegenerative Diseases, University of Texas Health Sciences Center, San Antonio, TX, USA
| |
Collapse
|
73
|
Ma W, Wu W, Wen W, Xu F, Han D, Lyu J, Huang Y. Association of NAFLD with cardiovascular disease and all-cause mortality: a large-scale prospective cohort study based on UK Biobank. Ther Adv Chronic Dis 2022; 13:20406223221122478. [PMID: 36159632 PMCID: PMC9493675 DOI: 10.1177/20406223221122478] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 08/03/2022] [Indexed: 12/02/2022] Open
Abstract
Objective Nonalcoholic fatty liver disease (NAFLD) is considered as the hepatic manifestation of metabolic syndrome, sharing the similar cardiometabolic risk factors with cardiovascular disease (CVD). Whether NAFLD by itself is associated with increased cardiovascular events and death remain an issue to debate. This study aimed to further investigate the association between NAFLD and adverse CVD outcomes. Methods Participants were followed up until the end of 2020 in current analysis. NAFLD is defined using fatty liver index (FLI). Cox proportional hazard model was used to analyze the association between NAFLD and all-cause mortality, major adverse cardiovascular events (MACEs), CVD mortality, fatal/nonfatal acute myocardial infarction (AMI), and fatal/nonfatal stroke. C-index was calculated to evaluate the model enhancement when adding NAFLD factor. Results After screening the data of 502,492 participants in the original cohort, 215,245 eligible participants were included in this study for MACEs outcome. Compared with non-NAFLD participants, the multivariable adjusted hazard ratios of NAFLD group was 1.25 (1.14-1.36) for MACEs; 1.14 (1.08-1.20) for all-cause mortality; 1.61(1.42-1.82) for CVD mortality; 1.58(1.19-2.11) for AMI mortality; and 1.18 (0.85-1.64) for stroke mortality. When adding FLI, C-index of NAFLD model improved for all-cause mortality, MACEs, and CVD mortality compared with that in the traditional CVD risk factor model. Conclusion NAFLD is an independent risk factor for all-cause mortality and adverse CVD outcomes. Based on the traditional CVD risk factor model, additionally screening NAFLD could improve the prediction efficiency for adverse CVD outcomes.
Collapse
Affiliation(s)
- Wen Ma
- Department of Cardiology, Shunde Hospital, Southern Medical University, Foshan, P.R. China.,Department of Clinical Research, The First Affiliated Hospital, Jinan University, Guangzhou, P.R. China.,School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an, P.R. China
| | - Wentao Wu
- School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an, P.R. China
| | - Weixing Wen
- Department of Cardiology, Shunde Hospital, Southern Medical University, Foshan, P.R. China
| | - Fengshuo Xu
- School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an, P.R. China
| | - Didi Han
- School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an, P.R. China
| | - Jun Lyu
- Department of Clinical Research, The First Affiliated Hospital, Jinan University, Guangzhou 510630, P.R. China.,Guangdong Provincial Key Laboratory of Traditional Chinese Medicine Informatization (2021B1212040007), Guangzhou, P.R. China
| | - Yuli Huang
- Department of Cardiology, Shunde Hospital, Southern Medical University, Jiazi Road, Lunjiao, Shunde, Foshan 528300, P.R. China.,The George Institute for Global Health, Faculty of Medicine, University of New South Wales Sydney, Sydney, NSW, Australia
| |
Collapse
|
74
|
Wong MYZ, Yap JJL, Sultana R, Cheah M, Goh GBB, Yeo KK. Association between non-alcoholic fatty liver disease and subclinical atherosclerosis in Western and Asian cohorts: an updated meta-analysis. Open Heart 2021; 8:openhrt-2021-001850. [PMID: 34933963 PMCID: PMC8693165 DOI: 10.1136/openhrt-2021-001850] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 10/25/2021] [Indexed: 11/16/2022] Open
Abstract
Background Non-alcoholic fatty liver disease (NAFLD) is a well-established risk factor for cardiovascular disease, with ethnic and regional differences noted. With the recent surge of research within this field, we re-examine the evidence associating NAFLD with subclinical atherosclerosis, and investigate potential regional differences. Methods This is a systematic review and meta-analysis. PubMed and EMBASE were systematically searched for publications from January 1967 to July 2020 using standardised criteria. Original, observational studies investigating the association between NAFLD and either carotid intima-media thickness (CIMT) and/or coronary artery calcification (CAC) were included. Key outcomes included differences in mean CIMT, the presence of increased CIMT, the presence of CAC and the development/progression of CAC. Pooled ORs and pooled standard differences in means were calculated using random-effects models. Between-study heterogeneity was quantified using the Q statistic and I². Subgroup analyses stratified by region of study (Asian vs Western) were also conducted. Results 64 studies involving a total of 172 385 participants (67 404 with NAFLD) were included. 44 studies assessed the effect of NAFLD on CIMT, with the presence of NAFLD associated with increased CIMT (OR 2.00, 95% CI 1.56 to 2.56). 22 studies assessed the effects of NAFLD on CAC score, with the presence of NAFLD associated with the presence of any coronary calcification (OR 1.21, 95% CI 1.12 to 1.32), and the development/progression of CAC (OR 1.26, 95% CI 1.04 to 1.52). When stratified by region, these associations remained consistent across both Asian and Western populations (p>0.05). The majority (n=39) of studies were classified as ‘high quality’, with the remaining 25 of ‘moderate quality’. Conclusions There is a significant positive association between various measures of subclinical atherosclerosis and NAFLD, seen across both Western and Asian populations. These results re-emphasise the importance of early risk evaluation and prophylactic intervention measures to preclude progression to clinical cardiovascular disease in patients with NAFLD.
Collapse
Affiliation(s)
| | - Jonathan Jiunn Liang Yap
- Department of Cardiology, National Heart Center Singapore, Singapore.,Duke-NUS Medical School, Singapore
| | | | - Mark Cheah
- Department of Gastroenterology & Hepatology, Singapore General Hospital, Singapore
| | - George Boon Bee Goh
- Duke-NUS Medical School, Singapore.,Department of Gastroenterology & Hepatology, Singapore General Hospital, Singapore
| | - Khung Keong Yeo
- Department of Cardiology, National Heart Center Singapore, Singapore .,Duke-NUS Medical School, Singapore
| |
Collapse
|
75
|
Vohra AH, Upadhyay KK, Joshi AS, Vyas HS, Thadani J, Devkar RV. Melatonin-primed ADMSCs elicit an efficacious therapeutic response in improving high-fat diet induced non-alcoholic fatty liver disease in C57BL/6J mice. EGYPTIAN LIVER JOURNAL 2021. [DOI: 10.1186/s43066-021-00157-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Stem cells are widely used for therapy including treatment of liver damage. Adipose-derived mesenchymal stem cells (ADMSCs) administered to treat fatty liver are known to improve liver function but their use is restricted due to a poor success rate. This study investigates efficacy of melatonin-primed ADMSCs (Mel. MSCs) in experimentally induced non-alcoholic fatty liver disease (NAFLD).
Results
MSCs treated with LPS showed prominent DCFDA fluorescence as compared to the untreated cells. Also, the JC-1 staining had accounted for higher intensity of green monomer and a weak fluorescence of red dimer indicating weaker mitochondrial membrane potential. But melatonin co-treatment could make necessary corrective changes as evidenced by reverse set of results. The overall cell survival was also found to be improved following melatonin treatment as evidenced by the MTT assay. Also, the antioxidant (Nrf2 and Ho-1) and anti-inflammatory genes (Il-4 and Il-10) showed a decrement in their mRNA levels following LPS treatment whereas the pro-inflammatory genes (Tnf-α, Il-6, Tlr-4, and Lbp) showed a reciprocal increment in the said group. Melatonin co-treatment accounted for an improved status of antioxidant and anti-inflammatory genes as evidenced by their mRNA levels. High-fat high-fructose diet (HFFD) fed C57BL/6J mice recorded higher serum AST and ALT levels and fatty manifestation in histology of liver along with lowered mRNA levels of antioxidant (Nrf2, Catalase, and Gss) genes and Hgf. These set of parameters showed a significant improvement in HFFD + Mel.MSC group.
Conclusion
A significant improvement in viability of MSCs was recorded due to lowered intracellular oxidative stress and improves mitochondrial membrane potential. Further, melatonin-primed MSCs accounted for a significant decrement in fatty manifestations in liver and an improved physiological status of NAFLD in HFFD fed C57BL/6J mice. Taken together, it is hypothesized that melatonin priming to MSCs prior to its use can significantly augment the success of stem cell therapy.
Collapse
|
76
|
Cardiovascular computed tomography and HIV: The evolving role of imaging biomarkers in enhanced risk prediction. IMAGING 2021. [DOI: 10.1556/1647.2021.00025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Abstract
The treatment of human immunodeficiency virus (HIV) with antiretroviral (ARV) medications has revolutionised the care for these patients. The dramatic increase in life expectancy has brought new challenges in treating diseases of aging in this cohort. Cardiovascular disease (CVD) is now a leading cause of morbidity and mortality with risk matched HIV-positive patients having double the risk of MI compared to HIV-negative patients. This enhanced risk is secondary to the interplay the virus (and accessory proteins), ARV medications and traditional risk factors. The culmination of these factors can lead to a hybrid metabolic syndrome characterised by heightened ectopic fat. Cardiovascular computed tomography (CT) is ideal for quantifying epicardial adipose tissue volumes, hepatosteatosis and cardiovascular disease burden. The CVD risk attributed to disease burden and plaque morphology is well established in general populations but is less clear in HIV populations. The purpose of this review article is to appraise the latest data on CVD development in HIV-positive patients and how the use of cardiovascular CT may be used to enhance risk prediction in this population. This may have important implications on individualised treatment decisions and risk reduction strategies which will improve the care of these patients.
Collapse
|
77
|
Torre P, Motta BM, Sciorio R, Masarone M, Persico M. Inflammation and Fibrogenesis in MAFLD: Role of the Hepatic Immune System. Front Med (Lausanne) 2021; 8:781567. [PMID: 34957156 PMCID: PMC8695879 DOI: 10.3389/fmed.2021.781567] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 11/05/2021] [Indexed: 12/12/2022] Open
Abstract
Metabolic (dysfunction)-associated fatty liver disease (MAFLD) is the definition recently proposed to better circumscribe the spectrum of conditions long known as non-alcoholic fatty liver disease (NAFLD) that range from simple steatosis without inflammation to more advanced liver diseases. The progression of MAFLD, as well as other chronic liver diseases, toward cirrhosis, is driven by hepatic inflammation and fibrogenesis. The latter, result of a "chronic wound healing reaction," is a dynamic process, and the understanding of its underlying pathophysiological events has increased in recent years. Fibrosis progresses in a microenvironment where it takes part an interplay between fibrogenic cells and many other elements, including some cells of the immune system with an underexplored or still unclear role in liver diseases. Some therapeutic approaches, also acting on the immune system, have been probed over time to evaluate their ability to improve inflammation and fibrosis in NAFLD, but to date no drug has been approved to treat this condition. In this review, we will focus on the contribution of the liver immune system in the progression of NAFLD, and on therapies under study that aim to counter the immune substrate of the disease.
Collapse
Affiliation(s)
- Pietro Torre
- Internal Medicine and Hepatology Unit, Department of Medicine, Surgery and Dentistry, “Scuola Medica Salernitana”, University of Salerno, Salerno, Italy
| | - Benedetta Maria Motta
- Department of Medicine, Surgery and Dentistry, “Scuola Medica Salernitana”, University of Salerno, Baronissi, Italy
| | - Roberta Sciorio
- Internal Medicine and Hepatology Unit, Department of Medicine, Surgery and Dentistry, “Scuola Medica Salernitana”, University of Salerno, Salerno, Italy
| | - Mario Masarone
- Internal Medicine and Hepatology Unit, Department of Medicine, Surgery and Dentistry, “Scuola Medica Salernitana”, University of Salerno, Salerno, Italy
| | - Marcello Persico
- Internal Medicine and Hepatology Unit, Department of Medicine, Surgery and Dentistry, “Scuola Medica Salernitana”, University of Salerno, Salerno, Italy
| |
Collapse
|
78
|
Zarghamravanbakhsh P, Frenkel M, Poretsky L. Metabolic causes and consequences of nonalcoholic fatty liver disease (NAFLD). Metabol Open 2021; 12:100149. [PMID: 34870138 PMCID: PMC8626571 DOI: 10.1016/j.metop.2021.100149] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 11/10/2021] [Accepted: 11/10/2021] [Indexed: 12/11/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a multifactorial metabolic disorder that was first described in 1980. It has been prevalent and on the rise for many years and is associated with other metabolic disorders such as obesity and type 2 diabetes mellitus (T2DM). NAFLD can be best described as a metabolic dysfunction that stems from insulin resistance-induced hepatic lipogenesis. This lipogenesis increases oxidative stress and hepatic inflammation and is often potentiated by genetic and gut microbiome dysfunction. As NAFLD progresses from simple steatosis to non-alcoholic steatohepatitis (NASH) and to cirrhosis and hepatocellular carcinoma (HCC), the odds of complications including cardiovascular disease (CVD), chronic kidney disease (CKD), and overall mortality increase. The aim of this review is to describe the metabolic causes and consequences of NAFLD while examining the risks that each stage of NAFLD poses. In this review, the etiology of "lean" NAFLD, the impact of obesity, T2DM, genetics, and microbiome dysbiosis on NAFLD progression are all explored. This review will also discuss the core issue behind the progression of NAFLD: insulin resistance (IR). Upon describing the causes and consequences of NAFLD, the effectiveness of diet modification, lifestyle changes, and glucagon-like peptide 1 receptor (GLP-1) agonists to retard NAFLD progression and stem the rate of complications is examined.
Collapse
Affiliation(s)
- Paria Zarghamravanbakhsh
- Division of Endocrinology, Department of Medicine, Lenox Hill Hospital, Northwell Health, 110 East 59th St #8B, New York, NY, 10022, USA
| | - Michael Frenkel
- The Gerald J. Friedman Diabetes Institute, Northwell Health, 110 East 59th St #8B, New York, NY, 10022, USA
| | - Leonid Poretsky
- Division of Endocrinology, Department of Medicine, Lenox Hill Hospital, Northwell Health, 110 East 59th St #8B, New York, NY, 10022, USA
- The Gerald J. Friedman Diabetes Institute, Northwell Health, 110 East 59th St #8B, New York, NY, 10022, USA
| |
Collapse
|
79
|
Parameswaran M, Hasan HA, Sadeque J, Jhaveri S, Avanthika C, Arisoyin AE, Dhanani MB, Rath SM. Factors That Predict the Progression of Non-alcoholic Fatty Liver Disease (NAFLD). Cureus 2021; 13:e20776. [PMID: 35111461 PMCID: PMC8794413 DOI: 10.7759/cureus.20776] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/28/2021] [Indexed: 02/07/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) refers to a spectrum of diseases involving the deposition of fat in the hepatocytes of people with little to no alcohol consumption. NAFLD is associated with hypertension, diabetes, obesity, etc. As their prevalence increases, the propensity and severity of NAFLD might increase. As per the recently developed multi-hit hypothesis, factors like oxidative stress, genetic predisposition, lipotoxicity, and insulin resistance have been found to play a key role in the development of NAFLD and its associated complications. This article focuses on NAFLD, its pathophysiology, risk factors, and the various genetic and epigenetic factors involved in its development along with possible treatment modalities. We conducted an all-language literature search on Medline, Cochrane, Embase, and Google Scholar until October 2021. The following search strings and Medical Subject Heading (MeSH) terms were used: “NAFLD,” “NASH,” “Fibrosis,” and “Insulin Resistance.” We explored the literature on NAFLD for its epidemiology, pathophysiology, the role of various genes, and how they influence the disease and associated complications about the disease and its hepatic and extrahepatic complications. With its rapidly increasing prevalence rates across the world and serious complications like NASH and hepatocellular carcinoma, NAFLD is becoming a major public health issue and more research is needed to formulate better screening tools and treatment protocols.
Collapse
Affiliation(s)
| | | | - Jafor Sadeque
- Internal Medicine, Al Mostaqbal Hospital, Jeddah, SAU
| | - Sharan Jhaveri
- Internal Medicine, Smt. Nathiba Hargovandas Lakhmichand Municipal Medical College, Ahmedabad, IND
| | | | | | - Maulik B Dhanani
- Internal Medicine, Southwestern University School of Medicine, Cebu City, PHL
| | - Swaroopa M Rath
- Medicine, Srirama Chandra Bhanja Medical College and Hospital, Cuttack, IND
| |
Collapse
|
80
|
Baldini F, Khalil M, Serale N, Voci A, Portincasa P, Vergani L. Extent and features of liver steatosis in vitro pave the way to endothelial dysfunction without physical cell-to-cell contact. Nutr Metab Cardiovasc Dis 2021; 31:3522-3532. [PMID: 34629256 DOI: 10.1016/j.numecd.2021.08.032] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 07/09/2021] [Accepted: 08/04/2021] [Indexed: 02/06/2023]
Abstract
BACKGROUND AND AIMS Several chronic multifactorial diseases originate from energy unbalance between food intake and body energy expenditure, including non-alcoholic fatty liver disease (NAFLD), diabetes, and cardiovascular disorders. Vascular endothelium plays a central role in body homeostasis, and NAFLD is often associated with endothelial dysfunction (ED), the first step in atherosclerosis. Both sugars and fatty acids (FAs) are fuel sources for energy production, but their excess leads to liver steatosis which may trigger ED through a network of mechanisms which need to be clarified. Here, we investigated the crosstalk pathways between in vitro cultured steatotic hepatocytes (FaO) and endothelial cells (HECV) being mediated by soluble factors. METHODS AND RESULTS We employed the conditioned medium approach to test how different extent and features of hepatic steatosis distinctively affect endothelium leading to ED. The steatogenic media collected from steatotic hepatocytes were characterized by high triglyceride content and led to lipid accumulation and fat-dependent dysfunction in HECV cells. We found a parallelism between (i) extent of hepatocyte steatosis and level of lipid accumulation in HECV cells; (ii) type of hepatocyte steatosis (with macro- or microvesicular LDs) and extent of oxidative stress, lipid peroxidation, nitric oxide release and expression of ED markers in HECV cells. CONCLUSIONS The present findings seem to suggest that, in addition to triglycerides, other soluble mediators should be released by steatotic hepatocytes and may influence lipid accumulation and function of HECV cells. Further studies need to depict the exact profile of soluble factors involved in steatotic hepatocyte-endothelium crosstalk.
Collapse
Affiliation(s)
- Francesca Baldini
- Department of Earth, Environment and Life Sciences (DISTAV), University of Genova, Corso Europa 26, 16132, Italy; Nanoscopy and NIC@IIT, Istituto Italiano di Tecnologia, Genoa, Italy
| | - Mohamad Khalil
- Clinica Medica "A. Murri", Dept. of Biomedical Sciences and Human Oncology, Medical School, University of Bari "Aldo Moro", Italy
| | - Nadia Serale
- Department of Earth, Environment and Life Sciences (DISTAV), University of Genova, Corso Europa 26, 16132, Italy; Clinica Medica "A. Murri", Dept. of Biomedical Sciences and Human Oncology, Medical School, University of Bari "Aldo Moro", Italy
| | - Adriana Voci
- Department of Earth, Environment and Life Sciences (DISTAV), University of Genova, Corso Europa 26, 16132, Italy
| | - Piero Portincasa
- Clinica Medica "A. Murri", Dept. of Biomedical Sciences and Human Oncology, Medical School, University of Bari "Aldo Moro", Italy
| | - Laura Vergani
- Department of Earth, Environment and Life Sciences (DISTAV), University of Genova, Corso Europa 26, 16132, Italy.
| |
Collapse
|
81
|
Hepatic Interactions in Atherosclerotic Heart Disease. Am J Med Sci 2021; 363:104-113. [PMID: 34547286 DOI: 10.1016/j.amjms.2021.07.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 04/19/2021] [Accepted: 07/16/2021] [Indexed: 12/20/2022]
Abstract
Atherosclerotic heart disease remains a major cause of morbidity and mortality worldwide. The past few decades have seen the emergence of chronic inflammation as a mediator of atherosclerosis. Although the heart and vascular system remain the organ systems most affected in the atherosclerotic process, chronic inflammation and ischemia trigger a systemic multi-organ response. The liver is a critical organ for systemic hemostasis and recent developments have established an important role of the liver in response to atherosclerosis and myocardial ischemia. In addition, the rapid emergence of systemic liver diseases has unraveled a pathophysiological link with heart disease with therapeutic implications. In this review, we explore the relationship between the liver and the heart in myocardial ischemia, describe epidemiological associations between various liver pathologies and coronary heart disease, and elucidate practical challenges in the clinical management of patients with concomitant coronary heart disease and hepatic abnormalities.
Collapse
|
82
|
Cerebrovascular alterations in NAFLD: Is it increasing our risk of Alzheimer's disease? Anal Biochem 2021; 636:114387. [PMID: 34537182 DOI: 10.1016/j.ab.2021.114387] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 07/27/2021] [Accepted: 09/15/2021] [Indexed: 02/07/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a multisystem disease, which has been classified as an emerging epidemic not only confined to liver-related morbidity and mortality. It is also becoming apparent that NAFLD is associated with moderate cerebral dysfunction and cognitive decline. A possible link between NAFLD and Alzheimer's disease (AD) has only recently been proposed due to the multiple shared genes and pathological mechanisms contributing to the development of these conditions. Although AD is a progressive neurodegenerative disease, the exact pathophysiological mechanism remains ambiguous and similarly to NAFLD, currently available pharmacological therapies have mostly failed in clinical trials. In addition to the usual suspects (inflammation, oxidative stress, blood-brain barrier alterations and ageing) that could contribute to the NAFLD-induced development and progression of AD, changes in the vasculature, cerebral perfusion and waste clearance could be the missing link between these two diseases. Here, we review the most recent literature linking NAFLD and AD, focusing on cerebrovascular alterations and the brain's clearance system as risk factors involved in the development and progression of AD, with the aim of promoting further research using neuroimaging techniques and new mechanism-based therapeutic interventions.
Collapse
|
83
|
Kawachi Y, Fujishima Y, Nishizawa H, Nakamura T, Akari S, Murase T, Saito T, Miyazaki Y, Nagao H, Fukuda S, Kita S, Katakami N, Doki Y, Maeda N, Shimomura I. Increased plasma XOR activity induced by NAFLD/NASH and its possible involvement in vascular neointimal proliferation. JCI Insight 2021; 6:e144762. [PMID: 34494551 PMCID: PMC8492303 DOI: 10.1172/jci.insight.144762] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 07/21/2021] [Indexed: 12/20/2022] Open
Abstract
Xanthine oxidoreductase (XOR) is an enzyme that catalyzes hypoxanthine to xanthine and xanthine to uric acid, respectively. However, the underlying mechanisms of increased plasma XOR and its pathological roles in systemic diseases, such as atherosclerosis, are not fully understood. In this study, we found that changes in plasma XOR activity after bariatric surgery closely associated with those in liver enzymes, but not with those in BMI. In a mouse model of nonalcoholic fatty liver disease/steatohepatitis (NAFLD/NASH), plasma XOR activity markedly increased. Besides, purine catabolism was accelerated in the plasma per se of NASH mice and human patients with high XOR activity. In our NASH mice, we observed an increased vascular neointima formation consisting of dedifferentiated vascular smooth muscle cells (SMCs), which was significantly attenuated by topiroxostat, a selective XOR inhibitor. In vitro, human liver S9–derived XOR promoted proliferation of SMCs with phenotypic modulation and induced ROS production by catabolizing hypoxanthine released from human endothelial cells. Collectively, the results from human and mouse models suggest that increased plasma XOR activity, mainly explained by excess hepatic leakage, was involved in the pathogenesis of vascular injury, especially in NAFLD/NASH conditions.
Collapse
Affiliation(s)
- Yusuke Kawachi
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Yuya Fujishima
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Hitoshi Nishizawa
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | | | - Seigo Akari
- Sanwa Kagaku Kenkyusho Co., Ltd., Inabe, Mie, Japan
| | | | | | | | - Hirofumi Nagao
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Shiro Fukuda
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Shunbun Kita
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan.,Department of Adipose Management, and
| | - Naoto Katakami
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan.,Department of Metabolism and Atherosclerosis, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | | | - Norikazu Maeda
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan.,Department of Metabolism and Atherosclerosis, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Iichiro Shimomura
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| |
Collapse
|
84
|
Pennisi G, Di Marco V, Buscemi C, Mazzola G, Randazzo C, Spatola F, Craxì A, Buscemi S, Petta S. Interplay between non-alcoholic fatty liver disease and cardiovascular risk in an asymptomatic general population. J Gastroenterol Hepatol 2021; 36:2389-2396. [PMID: 33871081 DOI: 10.1111/jgh.15523] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 11/07/2020] [Accepted: 12/03/2020] [Indexed: 12/27/2022]
Abstract
BACKGROUND AND AIMS Non-alcoholic fatty liver (NAFLD) is a major cause of liver disease worldwide leading also to a higher risk of cardiovascular events. We aimed to evaluate the impact of fatty liver and fibrosis on cardiovascular risk in a general population. METHODS Five hundred and forty-two subjects included in the community-based ABCD (Alimentazione, Benessere Cardiovascolare e Diabete) study were recruited. Steatosis (controlled attenuation parameter > 288 dB/m) and severe fibrosis (low risk, liver stiffness measurement [LSM] < 7.9 KPa with M probe and < 5.7 KPa with XL probe; intermediate risk, LSM 7.9-9.5 KPa with M probe and 5.7-9.2 KPa with XL probe; high risk, LSM ≥ 9.6 KPa with M probe and ≥ 9.3 KPa with XL probe) were assessed with FibroScan. Cardiovascular risk was evaluated by the atherosclerotic cardiovascular disease (ASCVD) risk estimator and defined low if < 5%, borderline if 5-7.4%, intermediate if 7.5-19.9% and high if ≥ 20%. Intima-media thickness (IMT) was measured with ultrasound. RESULTS Prevalence of steatosis and of severe fibrosis in this cohort was 31.7% and 4.8%, respectively. ASCVD score was evaluated in patients with and without steatosis and according to the risk of severe fibrosis. By ordinal regression analysis, both steatosis (odds ratio [OR] 1.62, 95% confidence interval [CI] 1.13-2.33, P = 0.009) and severity of fibrosis (OR 1.67, 95% CI 1.18-2.36, P = 0.003) were independent risk factors for a higher ASCVD risk after adjusting for obesity. Subjects with NAFLD, when compared with those without, did not differ for IMT (0.75 vs 0.72 mm; P = 0.11) and IMT ≥ 1 mm (15.6% vs 12.1%; P = 0.24). Higher prevalence of IMT ≥ 1 mm was found in patients at high or intermediate risk of severe fibrosis (24% and 28.6%, respectively) compared with those at low risk (12.1%) (P = 0.03); this association was maintained after adjusting for confounders (OR 2.70, 95% CI 1.01-2.86, P = 0.04). CONCLUSION In the setting of a general adult population, the presence of NAFLD and severe fibrosis are associated with to a higher cardiovascular risk profile, pointing towards the need for specific preventive measures.
Collapse
Affiliation(s)
- Grazia Pennisi
- Section of Gastroenterology and Hepatology, Department of Health Promotion, Maternal and Childhood, Internal and Specialized Medicine of Excellence (PROMISE), University of Palermo, Palermo, Italy
| | - Vito Di Marco
- Section of Gastroenterology and Hepatology, Department of Health Promotion, Maternal and Childhood, Internal and Specialized Medicine of Excellence (PROMISE), University of Palermo, Palermo, Italy
| | - Carola Buscemi
- Section of Gastroenterology and Hepatology, Department of Health Promotion, Maternal and Childhood, Internal and Specialized Medicine of Excellence (PROMISE), University of Palermo, Palermo, Italy
| | - Giovanni Mazzola
- Infectious Diseases Unit, Department of Health Promotion Sciences and Mother Child Care "Giuseppe D'Alessandro", University of Palermo, Palermo, Italy
| | - Cristiana Randazzo
- Endocrine Diseases, Replacement and Nutrition Unit, Policlinico P. Giaccone Palermo, Palermo, Italy
| | - Federica Spatola
- Section of Gastroenterology and Hepatology, Department of Health Promotion, Maternal and Childhood, Internal and Specialized Medicine of Excellence (PROMISE), University of Palermo, Palermo, Italy
| | - Antonio Craxì
- Section of Gastroenterology and Hepatology, Department of Health Promotion, Maternal and Childhood, Internal and Specialized Medicine of Excellence (PROMISE), University of Palermo, Palermo, Italy
| | - Silvio Buscemi
- Endocrine Diseases, Replacement and Nutrition Unit, Policlinico P. Giaccone Palermo, Palermo, Italy
| | - Salvatore Petta
- Section of Gastroenterology and Hepatology, Department of Health Promotion, Maternal and Childhood, Internal and Specialized Medicine of Excellence (PROMISE), University of Palermo, Palermo, Italy
| |
Collapse
|
85
|
Ichikawa K, Miyoshi T, Osawa K, Miki T, Morimitsu Y, Akagi N, Nakashima M, Ito H. Association between higher pericoronary adipose tissue attenuation measured by coronary computed tomography angiography and nonalcoholic fatty liver disease: A matched case-control study. Medicine (Baltimore) 2021; 100:e27043. [PMID: 34449489 PMCID: PMC8389939 DOI: 10.1097/md.0000000000027043] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 08/11/2021] [Indexed: 01/04/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a risk factor for cardiac mortality. Pericoronary adipose tissue (PCAT) attenuation, expressed by the fat attenuation index on coronary computed tomography angiography, reflects pericoronary inflammation. We aimed to investigate the association between PCAT attenuation and NAFLD.This is a single-center cohort study comprising of patients who underwent coronary computed tomography angiography for suspected stable coronary artery disease between January and December 2020. Patient characteristics and coronary computed tomography angiography findings were analyzed between patients with NAFLD (n = 78) and a propensity score-matched cohort of patients without NAFLD (n = 78). PCAT attenuation was assessed in Hounsfield units (HU) of proximal 40-mm segments of the left anterior descending artery (LAD) and right coronary artery.The mean PCAT attenuation in LAD and right coronary artery were significantly higher in patients with NAFLD than those without NAFLD. When patients were divided into 2 groups using the median LAD-PCAT attenuation of -72.5 HU, the high PCAT attenuation group had more males (82% vs 67%, P = .028) and NAFLD patients (63% vs 37%, P = .001) compared to the low PCAT attenuation group. No differences in age, body mass index, conventional cardiovascular risk factors, or the presence of high-risk plaque were observed between the 2 groups. In the multivariate logistic analysis, NAFLD was independently associated with high PCAT attenuation (odds ratio 2.912, 95% confidence interval 1.386 to 6.118, P = .005).NAFLD is associated with high PCAT attenuation on coronary computed tomography angiography. This finding suggests that pericoronary inflammation is involved in the increased cardiac mortality in NAFLD patients.
Collapse
Affiliation(s)
- Keishi Ichikawa
- Department of Cardiovascular Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Toru Miyoshi
- Department of Cardiovascular Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Kazuhiro Osawa
- Department of General Internal Medicine 3, Kawasaki Medical School General Medical Center, Okayama, Japan
| | - Takashi Miki
- Department of Cardiovascular Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Yusuke Morimitsu
- Department of Medical technology, Okayama University Hospital, Okayama, Japan
| | - Noriaki Akagi
- Department of Medical technology, Okayama University Hospital, Okayama, Japan
| | - Mitsutaka Nakashima
- Department of Cardiovascular Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Hiroshi Ito
- Department of Cardiovascular Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| |
Collapse
|
86
|
Ali H, Kazmi M, Choi C, Hashemipour R, Singh I, Pyrsopoulos NT. In-Hospital Outcomes of Patients With Non-Alcoholic Fatty Liver Disease Who Underwent Percutaneous Coronary Intervention: A Nationwide Inpatient Sample Analysis. Cureus 2021; 13:e17338. [PMID: 34430188 PMCID: PMC8378887 DOI: 10.7759/cureus.17338] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/18/2021] [Indexed: 11/05/2022] Open
Abstract
Background Non-alcoholic fatty liver disease (NAFLD) is prevalent in almost 25% of the Western population and is predicted to become one of the leading causes of end-stage liver disease. There is increasing evidence that NAFLD is a risk factor for cardiovascular disease, specifically for coronary artery disease, via disruption of the metabolism of glucose and lipids in the body, leading to a state of systemic inflammation that promotes atherosclerosis. This study aims to explore outcomes in patients who underwent percutaneous coronary intervention (PCI) with or without placement of drug-eluting stents (DES) to determine whether the concurrent diagnosis of NAFLD led to worse in-hospital outcomes. Methods We used the National Inpatient Sample, Healthcare Cost and Utilization Project (HCUP), Agency for Healthcare Research and Quality 2016 to conduct a cross-sectional study that included all adult patients who underwent PCI with or without placement of DES during hospital admission. Patients with NAFLD were identified and compared to patients without NAFLD. Patients were selected by using ICD-10-CM and ICD-10-PCS codes. Outcomes included mortality, length of stay and total hospital charges, and major adverse cardiac events (MACE). Data on patient demographics, inpatient statistics, and comorbidities were obtained and analyzed using cross-tabulation, Pearson χ2 test, and independent samples t-test. Data were adjusted for confounders using logistic and linear regression. Results Among 429,855 patients who underwent PCI with or without placement of DES, 2,560 patients (0.6%) had a diagnosis of NAFLD. There was no significant difference with regard to mortality and MACE. The NAFLD group had a higher proportion of females, a longer average length of hospital stay, and patients presented at a younger average age. Regarding comorbidities, more patients in the NAFLD group had diabetes mellitus type II, obesity, obstructive sleep apnea (OSA), chronic kidney disease (CKD), and peripheral vascular disease (PVD). Conclusion NAFLD is emerging as a risk factor for cardiovascular disease. Increasing evidence suggests that the disease contributes to systemic atherosclerosis and thus coronary artery disease. We found that among patients who underwent PCI in 2016, those with NAFLD had a longer length of stay, were admitted at a younger age, and had significantly more cardiovascular comorbidities than those without NAFLD. Increasing evidence has shown that advanced liver disease due to NAFLD will continue to place a significant burden on the healthcare system and is, therefore, an area that the medical community should continue to focus on, especially, regarding preventative and therapeutic efforts.
Collapse
Affiliation(s)
- Hasan Ali
- Internal Medicine, Rutgers University New Jersey Medical School, Newark, USA
| | - Maryam Kazmi
- Internal Medicine, Rutgers University New Jersey Medical School, Newark, USA
| | - Catherine Choi
- Gastroenterology and Hepatology, Rutgers University New Jersey Medical School, Newark, USA
| | - Reza Hashemipour
- Gastroenterology and Hepatalogy, Rutgers University New Jersey Medical School, Newark, USA
| | - Inderjit Singh
- Cardiology, Rutgers University New Jersey Medical School, Newark, USA
| | - Nikolaos T Pyrsopoulos
- Gastroenterology and Hepatology, Rutgers University New Jersey Medical School, Newark, USA
| |
Collapse
|
87
|
Guzel FB, Ozturk I, Gisi K, Ispiroglu M, Akkus G, Erken E, Altunoren O, Gungor O. The relationship between hepatic fibrosis and arterial stiffness in hemodialysis patients. Semin Dial 2021; 35:222-227. [PMID: 34390271 DOI: 10.1111/sdi.13011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Revised: 07/16/2021] [Accepted: 07/19/2021] [Indexed: 12/01/2022]
Abstract
INTRODUCTION The main cause of death in hemodialysis patients is cardiovascular diseases. Increased arterial stiffness is a predictor of cardiovascular events for hemodialysis patients. Among the nondialysis patient population, arterial stiffness increases in those with hepatic fibrosis and nonalcoholic fatty liver disease. This study aims to examine the relationship between hepatic fibrosis and arterial stiffness in hemodialysis patients for the first time in the literature. MATERIAL AND METHOD The study includes chronic hemodialysis patients over 18 years of age who had been treated for hemodialysis for at least 6 months. Patients with chronic liver disease, chronic viral hepatitis (HBV and HCV), alcohol use, or liver disease accompanied by polycystic kidney disease and active infection were excluded. Hepatic fibrosis scores were measured using the FibroScan device. Single-cuff Mobil-o-Graph was used for measurement of arterial stiffness. RESULTS Fifty-nine patients were enrolled; 54.2% of the patients were male, and the mean age was 53.9 ± 12.9 years. Thirty-nine percent of the patients had diabetes. Average pulse wave velocity (PWV) value of the patients was 8.3 ± 1.6 m/s, and it had positive correlation with age, CAP score, fibrosis score, and body mass index and showed negative correlation to albumin. It was seen that the patients with a PWV value ≥ 10 m/s have significantly higher CAP score compared with the patients with a PWV < 10 m/s. When the factors predicting PWV were examined in the regression analysis, age and systolic blood pressure were found to be determinants. CONCLUSION Increased hepatic fibrosis in hemodialysis patients is associated with increased arterial stiffness, but this relationship is not independent.
Collapse
Affiliation(s)
- Fatma Betul Guzel
- Internal Medicine Department, Kahramanmaraş Necip Fazil City Hospital, Kahramanmaraş, Turkey
| | - Ilyas Ozturk
- Faculty of Medicine, Nephrology Department, Kahramanmaraş Sütçü İmam University, Kahramanmaraş, Turkey
| | - Kadir Gisi
- Faculty of Medicine, Gastroenterology Department, Kahramanmaraş Sütçü İmam University, Kahramanmaraş, Turkey
| | - Murat Ispiroglu
- Faculty of Medicine, Gastroenterology Department, Kahramanmaraş Sütçü İmam University, Kahramanmaraş, Turkey
| | - Gulsum Akkus
- Internal Medicine Department, Ankara Etimesgut Şehit Sait Ertürk State Hospital, Ankara, Turkey
| | - Ertugrul Erken
- Faculty of Medicine, Nephrology Department, Kahramanmaraş Sütçü İmam University, Kahramanmaraş, Turkey
| | - Orcun Altunoren
- Faculty of Medicine, Nephrology Department, Kahramanmaraş Sütçü İmam University, Kahramanmaraş, Turkey
| | - Ozkan Gungor
- Faculty of Medicine, Nephrology Department, Kahramanmaraş Sütçü İmam University, Kahramanmaraş, Turkey
| |
Collapse
|
88
|
Liver fibrosis is associated with carotid atherosclerosis in patients with liver biopsy-proven nonalcoholic fatty liver disease. Sci Rep 2021; 11:15938. [PMID: 34354193 PMCID: PMC8342487 DOI: 10.1038/s41598-021-95581-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 07/26/2021] [Indexed: 12/12/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is related to subclinical atherosclerosis. However, whether the severity of the disease (or which histopathological component) is associated with subclinical atherosclerosis remains controversial. This study aimed to investigate the association between the histopathological severity of NAFLD and carotid intima-media thickness (CIMT) in Japanese patients with liver biopsy-proven NAFLD. Maximum-CIMT (max-CIMT) was measured as an index of carotid atherosclerosis in 195 biopsy-proven NAFLD patients. A significant association was observed between the severity of fibrosis (but not steatosis, inflammation, and ballooning) and max-CIMT. Older age, male gender, hypertension, and advanced fibrosis were independently linked to max-CIMT ≥ 1.2 mm. The prevalence of max-CIMT ≥ 1.2 mm was significantly higher in the advanced fibrosis group than in the non-advanced fibrosis group (75.4% versus 44.0%; p < 0.01). Non-invasive liver fibrosis markers and scoring systems, including fibrosis-4 index, NAFLD fibrosis score, hyaluronic acid, and Wisteria floribunda agglutinin positive Mac-2-binding protein, demonstrated that the diagnostic performance for max-CIMT ≥ 1.2 mm was similar to that of biopsy-based fibrosis staging. In conclusion, advanced fibrosis is significantly and independently associated with high-risk CIMT. Non-invasive fibrosis markers and scoring systems could help estimate the risk of atherosclerosis progression in patients with NAFLD.
Collapse
|
89
|
Møller S, Kimer N, Kronborg T, Grandt J, Hove JD, Barløse M, Gluud LL. Nonalcoholic Fatty Liver Disease and Cardiovascular Disease: Overlapping Mechanisms. Semin Liver Dis 2021; 41:235-247. [PMID: 33992031 DOI: 10.1055/s-0041-1725022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) denotes a condition with excess fat in the liver. The prevalence of NAFLD is increasing, averaging > 25% of the Western population. In 25% of the patients, NAFLD progresses to its more severe form: nonalcoholic steatohepatitis and >25% of these progress to cirrhosis following activation of inflammatory and fibrotic processes. NAFLD is associated with obesity, type 2 diabetes, and the metabolic syndrome and represents a considerable and increasing health burden. In the near future, NAFLD cirrhosis is expected to be the most common cause for liver transplantation. NAFLD patients have an increased risk of developing cardiovascular disease as well as liver-related morbidity. In addition, hepatic steatosis itself appears to represent an independent cardiovascular risk factor. In the present review, we provide an overview of the overlapping mechanisms and prevalence of NAFLD and cardiovascular disease.
Collapse
Affiliation(s)
- Søren Møller
- Department of Clinical Physiology and Nuclear Medicine, Center for Functional and Diagnostic Imaging and Research, Copenhagen University Hospital, Hvidovre, Denmark.,Department of Clinical Medicine, University of Copenhagen, Denmark
| | - Nina Kimer
- Gastro Unit, Medical Division, Copenhagen University Hospital Hvidovre, Denmark.,Bridge Translational Excellence Program, Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Denmark
| | - Thit Kronborg
- Gastro Unit, Medical Division, Copenhagen University Hospital Hvidovre, Denmark
| | - Josephine Grandt
- Gastro Unit, Medical Division, Copenhagen University Hospital Hvidovre, Denmark
| | - Jens Dahlgaard Hove
- Department of Clinical Physiology and Nuclear Medicine, Center for Functional and Diagnostic Imaging and Research, Copenhagen University Hospital, Hvidovre, Denmark.,Department of Cardiology, Copenhagen University Hospital, Hvidovre, Denmark
| | - Mads Barløse
- Department of Clinical Physiology and Nuclear Medicine, Center for Functional and Diagnostic Imaging and Research, Copenhagen University Hospital, Hvidovre, Denmark
| | - Lise Lotte Gluud
- Department of Clinical Medicine, University of Copenhagen, Denmark.,Gastro Unit, Medical Division, Copenhagen University Hospital Hvidovre, Denmark
| |
Collapse
|
90
|
Incorporating fatty liver disease in multidisciplinary care and novel clinical trial designs for patients with metabolic diseases. Lancet Gastroenterol Hepatol 2021; 6:743-753. [PMID: 34265276 DOI: 10.1016/s2468-1253(21)00132-1] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 03/24/2021] [Accepted: 03/26/2021] [Indexed: 02/08/2023]
Abstract
With the global epidemics of obesity and associated conditions, including type 2 diabetes, metabolic dysfunction-associated fatty liver disease, chronic kidney disease, hypertension, stroke, cardiovascular disease, osteoporosis, cancer, and cognitive changes, the prevalence of multimorbidity is rapidly increasing worldwide. In this Review, a panel of international experts from across the spectrum of metabolic diseases come together to identify the challenges and provide perspectives on building a framework for a virtual primary care-driven, patient-centred, multidisciplinary model to deliver holistic care for patients with metabolic dysfunction-associated fatty liver disease and associated metabolic diseases. We focus on clinical care and innovative trial design for metabolic dysfunction-associated fatty liver disease and associated metabolic diseases. This work represents a call to action to promote collaboration and partnerships between stakeholders for improving the lives of people with, or at risk of, metabolic dysfunction-associated fatty liver disease and associated metabolic diseases.
Collapse
|
91
|
Akinci B, Subauste A, Ajluni N, Esfandiari NH, Meral R, Neidert AH, Eraslan A, Hench R, Rus D, Mckenna B, Hussain HK, Chenevert TL, Tayeh MK, Rupani AR, Innis JW, Mantzoros CS, Conjeevaram HS, Burant CL, Oral EA. Metreleptin therapy for nonalcoholic steatohepatitis: Open-label therapy interventions in two different clinical settings. MED 2021; 2:814-835. [DOI: 10.1016/j.medj.2021.04.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
92
|
Meyersohn NM, Mayrhofer T, Corey KE, Bittner DO, Staziaki PV, Szilveszter B, Hallett T, Lu MT, Puchner SB, Simon TG, Foldyna B, Voora D, Ginsburg GS, Douglas PS, Hoffmann U, Ferencik M. Association of Hepatic Steatosis With Major Adverse Cardiovascular Events, Independent of Coronary Artery Disease. Clin Gastroenterol Hepatol 2021; 19:1480-1488.e14. [PMID: 32707340 PMCID: PMC7855524 DOI: 10.1016/j.cgh.2020.07.030] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 07/12/2020] [Accepted: 07/14/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND & AIMS Hepatic steatosis has been associated with increased risk of major adverse cardiovascular events (MACE) but it is not clear whether steatosis is independently associated with risk of MACE. We investigated whether steatosis is associated with risk of MACE independently of the presence and extent of baseline coronary artery disease, assessed by comprehensive contrast-enhanced computed tomography angiography (CTA). METHODS We conducted a nested cohort study of 3756 subjects (mean age, 60.6 years; 48.4% men) who underwent coronary CTA at 193 sites in North America, from July 2010 through September 2013, as part of the PROMISE study, which included noninvasive cardiovascular analyses of symptomatic outpatients without coronary artery disease. Independent core laboratory readers measured hepatic and splenic attenuation, using non-contrast computed tomography images to identify steatosis, and evaluated coronary plaques and stenosis in coronary CTA images. We collected data on participants' cardiovascular risk factors, presence of metabolic syndrome, and body mass index. The primary endpoint was an adjudicated composite of MACE (death, myocardial infarction, or unstable angina) during a median follow-up time of 25 months. RESULTS Among the 959 subjects who had steatosis (25.5% of the cohort), 42 had MACE (4.4%), whereas among the 2797 subjects without steatosis, 73 had MACE (2.6%) (hazard ratio [HR] for MACE in subjects with steatosis, 1.69; 95% CI, 1.16-2.48; P = .006 for MACE in subjects with vs without steatosis). This association remained after adjustment for atherosclerotic cardiovascular disease risk scores, significant stenosis, and metabolic syndrome (adjusted HR, 1.72; 95% CI, 1.16-2.54; P = .007) or obesity (adjusted HR, 1.75; 95% CI, 1.19-2.59; P = .005). Steatosis remained independently associated with MACE after adjustment for all CTA measures of plaques and stenosis. CONCLUSIONS Hepatic steatosis is associated with MACE independently of other cardiovascular risk factors or extent of coronary artery disease. Strategies to reduce steatosis might reduce risk of MACE. ClinicalTrials.gov no: NCT01174550.
Collapse
Affiliation(s)
- Nandini M. Meyersohn
- Cardiovascular Imaging Research Center, Massachusetts General Hospital, Boston, MA
| | - Thomas Mayrhofer
- Cardiovascular Imaging Research Center, Massachusetts General Hospital, Boston, MA,School of Business Studies, Stralsund University of Applied Sciences, Stralsund, Germany
| | - Kathleen E. Corey
- Division of Gastroenterology, Massachusetts General Hospital, Boston, MA
| | - Daniel O. Bittner
- Cardiovascular Imaging Research Center, Massachusetts General Hospital, Boston, MA,Friedrich-Alexander University Erlangen-Nürnberg, Department of Cardiology, University Hospital Erlangen, Germany
| | - Pedro V. Staziaki
- Cardiovascular Imaging Research Center, Massachusetts General Hospital, Boston, MA
| | - Balint Szilveszter
- Cardiovascular Imaging Research Center, Massachusetts General Hospital, Boston, MA
| | - Travis Hallett
- Cardiovascular Imaging Research Center, Massachusetts General Hospital, Boston, MA
| | - Michael T. Lu
- Cardiovascular Imaging Research Center, Massachusetts General Hospital, Boston, MA
| | - Stefan B. Puchner
- Cardiovascular Imaging Research Center, Massachusetts General Hospital, Boston, MA,Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Tracey G. Simon
- Division of Gastroenterology, Massachusetts General Hospital, Boston, MA
| | - Borek Foldyna
- Cardiovascular Imaging Research Center, Massachusetts General Hospital, Boston, MA
| | - Deepak Voora
- Duke Center for Applied Genomics & Precision Medicine, Duke University School of Medicine, Durham, NC
| | - Geoffrey S. Ginsburg
- Duke Center for Applied Genomics & Precision Medicine, Duke University School of Medicine, Durham, NC
| | - Pamela S. Douglas
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC
| | - Udo Hoffmann
- Cardiovascular Imaging Research Center, Massachusetts General Hospital, Boston, MA
| | - Maros Ferencik
- Cardiovascular Imaging Research Center, Massachusetts General Hospital, Boston, MA,Knight Cardiovascular Institute, Oregon Health and Science University, Portland, OR
| |
Collapse
|
93
|
Muzurović E, Mikhailidis DP, Mantzoros C. Non-alcoholic fatty liver disease, insulin resistance, metabolic syndrome and their association with vascular risk. Metabolism 2021; 119:154770. [PMID: 33864798 DOI: 10.1016/j.metabol.2021.154770] [Citation(s) in RCA: 135] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 03/25/2021] [Accepted: 03/27/2021] [Indexed: 02/07/2023]
Abstract
The prevalence of non-alcoholic fatty liver disease (NAFLD), one of the most common liver diseases, is rising. About 25% of adults worldwide are probably affected by NAFLD. Insulin resistance (IR) and fat accumulation in the liver are strongly related. The association between NAFLD, metabolic syndrome (MetS) and IR is established, but an independent impact of NAFLD on vascular risk and progression of cardiovascular (CV) disease (CVD) still needs to be confirmed. This narrative review considers the evidence regarding the link between NAFLD, IR and CVD risk. There is strong evidence for a "concomitantly rising incidence" of NAFLD, IR, MetS and CVD but there is no definitive evidence regarding whether NAFLD is, or is not, an independent and significant risk factor the development of CVD. There are also considerations that type 2 diabetes mellitus (T2DM) may be a common link between NAFLD/non-alcoholic steatohepatitis (NASH) and CVD. NAFLD may be associated with widespread abnormal peri-organ or intra-organ fat (APIFat) deposition (e.g. epicardial adipose tissue) which may further contribute to CV risk. It is clear that NAFLD patients have a greater CV risk (independent or not) which needs to be addressed in clinical practice.
Collapse
Affiliation(s)
- Emir Muzurović
- Department of Internal Medicine, Endocrinology Section, Clinical Centre of Montenegro, Ljubljanska bb, 81000 Podgorica, Montenegro; Faculty of Medicine, University of Montenegro, Kruševac bb, 81000 Podgorica, Montenegro.
| | - Dimitri P Mikhailidis
- Department of Clinical Biochemistry, Royal Free Hospital Campus, University College London Medical School, University College London (UCL), Pond Street, London NW3 2QG, UK; Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, United Arab Emirates
| | - Christos Mantzoros
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA; Section of Endocrinology, Boston VA Healthcare System, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
94
|
Abstract
TGR5 (G protein-coupled bile acid receptor 1, GPBAR-1) is a G protein-coupled receptor with seven transmembrane domains and is widely distributed in various organs and tissues. As an important bile acid receptor, TGR5 can be activated by primary and secondary bile acids. Increased expression of TGR5 is a risk factor for polycystic liver disease and hepatobiliary cancer. However, there is evidence that the anti-inflammatory effect of the TGR5 receptor and its regulatory effect on hydrophobic bile acid confer protective effects against most liver diseases. Recent studies have shown that TGR5 receptor activation can alleviate the development of diabetic liver fibrosis, regulate the differentiation of natural killer T cells into NKT10 cells, increase the secretion of anti-inflammatory factors, inhibit the invasion of hepatitis B virus, promote white adipose tissue browning, improve arterial vascular dynamics, maintain tight junctions between bile duct cells, and protect against apoptosis. In portal hypertension, TGR5 receptor activation can inhibit the contraction of hepatic stellate cells and improve intrahepatic microcirculation. In addition, the discovery of the regulatory relationship between the TGR5 receptor and miRNA-26a provides a new direction for further studies of the molecular mechanism underlying the effects of TGR5. In this review, we describe recent findings linking TGR5 to various liver diseases, with a focus on the mechanisms underlying its effects and potential therapeutic implications.
Collapse
Affiliation(s)
- Ke Ma
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Dan Tang
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Chang Yu
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Lijin Zhao
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| |
Collapse
|
95
|
Iruzubieta P, Arias-Loste MT, Fortea JI, Cuadrado A, Rivas-Rivas C, Rodriguez-Duque JC, García-Ibarbia C, Hernández JL, Crespo J. National digestive disease specialists survey on cardiovascular risk management in non-alcoholic fatty liver disease in spanish hospitals. Liver Int 2021; 41:1243-1253. [PMID: 33527637 DOI: 10.1111/liv.14807] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 01/27/2021] [Accepted: 01/28/2021] [Indexed: 12/14/2022]
Abstract
BACKGROUND & AIMS Cardiovascular disease (CVD) is the main cause of mortality among non-alcoholic fatty liver disease (NAFLD) patients. The aim was to explore the level of knowledge and clinical management of cardiovascular risk (CVR) in NAFLD patients by Digestive Disease specialists. METHODS An anonymous web-based survey was designed with 44 close-ended questions, divided into five sections, that were based on current guidelines on CVD prevention. Between November 2019 and January 2020, Digestive Disease specialists from Spanish hospitals were invited to participate in this survey via email and Twitter. Student's t, chi-square and Fishers' exact tests, and logistic regression were used for data analysis. RESULTS 208 clinicians completed the survey. Most respondents (83.2%) believe that NAFLD is an independent risk factor for CVD, especially in the presence of NASH and fibrosis. Personal history of CVDs and cardiovascular risk-related comorbidities are collected by more than 75% of respondents. However, less than 17% perform an elementary physical examination to address the CVR, except weight which is evaluated by 69.8%. Over 54% of respondents do not perform or request any supplementary tests for CVR assessment, and only 10.2% use specific calculators. Furthermore, 54.3% spend less than 5 minutes giving lifestyle advice, and more than 52% do not start drug treatment after a recent diagnosis of any cardiovascular comorbidity. Only 25.6% have a multidisciplinary Unit for metabolic comorbidities in their hospitals, although 89% of the respondents would support the implementation of this Unit. CONCLUSIONS Cardiovascular risk management in daily clinical practice by Digestive Disease specialists in Spain remains suboptimal.
Collapse
Affiliation(s)
- Paula Iruzubieta
- Gastroenterology and Hepatology Department, Marqués de Valdecilla University Hospital. Clinical and Translational Digestive Research Group, IDIVAL, Santander, Spain
| | - María Teresa Arias-Loste
- Gastroenterology and Hepatology Department, Marqués de Valdecilla University Hospital. Clinical and Translational Digestive Research Group, IDIVAL, Santander, Spain
| | - José Ignacio Fortea
- Gastroenterology and Hepatology Department, Marqués de Valdecilla University Hospital. Clinical and Translational Digestive Research Group, IDIVAL, Santander, Spain
| | - Antonio Cuadrado
- Gastroenterology and Hepatology Department, Marqués de Valdecilla University Hospital. Clinical and Translational Digestive Research Group, IDIVAL, Santander, Spain
| | - Coral Rivas-Rivas
- Gastroenterology and Hepatology Department, Marqués de Valdecilla University Hospital. Clinical and Translational Digestive Research Group, IDIVAL, Santander, Spain
| | - Juan Carlos Rodriguez-Duque
- Gastroenterology and Hepatology Department, Marqués de Valdecilla University Hospital. Clinical and Translational Digestive Research Group, IDIVAL, Santander, Spain
| | - Carmen García-Ibarbia
- Lipid and Vascular Risk Unit, Department of Internal Medicine, Marqués de Valdecilla University Hospital, IDIVAL, University of Cantabria, Santander, Spain
| | - José Luis Hernández
- Lipid and Vascular Risk Unit, Department of Internal Medicine, Marqués de Valdecilla University Hospital, IDIVAL, University of Cantabria, Santander, Spain
| | - Javier Crespo
- Gastroenterology and Hepatology Department, Marqués de Valdecilla University Hospital. Clinical and Translational Digestive Research Group, IDIVAL, Santander, Spain
| |
Collapse
|
96
|
Tarantino G, Citro V, Balsano C, Capone D. Age and Interleukin-15 Levels Are Independently Associated With Intima-Media Thickness in Obesity-Related NAFLD Patients. Front Med (Lausanne) 2021; 8:634962. [PMID: 34095164 PMCID: PMC8175965 DOI: 10.3389/fmed.2021.634962] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Accepted: 04/06/2021] [Indexed: 01/06/2023] Open
Abstract
Common carotid intima-media thickness (IMT) represents a functional and structural marker of early, precocious, and subclinical atherosclerosis, independently from the carotid plaque. Macrophage cells, which have been detected in adipose tissue and atherosclerotic plaques, are regulated by interleukin-15 (IL-15). At the light of the conflicting results concerning the role of IL-15 in atherosclerosis, the aim of the study was to retrospectively evaluate in a population of 80 obese patients, with median age of 46 years (IQR 34–53 years), with a low rate of comorbidities but with non-alcoholic fatty liver disease (NAFLD) or hepatic steatosis (HS), the relationship between IMT and serum concentrations of IL-15. Anthropometric measures, metabolic profile, and serum inflammatory markers, as well as the levels of IL-15, MCP-1, b FGF, and GM-CSF, were analyzed by a bead-based assay. IMT, HS, subcutaneous, and visceral adipose tissues were detected by ultrasonography. The IL-15 levels of the obese patients were increased with respect to those of 44 young healthy subjects, i.e., 2.77 (1.21–4.8) vs. 1.55 (1–2.4) pg/mL (P = 0.002). In the univariate analysis, IL-15 levels were associated to IMT and to those of MCP-1, b FGF, and GM-CSF, without any relation to other inflammatory markers such as CRP and ferritin, except fibrinogen. In the multivariate analysis, after adjusting the HS severity for the extent of visceral adiposity, a dramatic change in prediction of IMT by HS was shown (β from 0.29 to 0.10, P from 0.008 to 0.37). When the visceral adipose tissue was combined with IL-15, on the one hand, and the well-known coronary artery disease (CAD) risk factors—i.e., age, gender, smoking status, HDL-cholesterol concentrations, triglycerides levels, and HOMA—on the other, only age and IL-15 remained the predictors of IMT (β = 0.60, P = 0.0001 and β = 0.25, P = 0.024, respectively). There was no association of IL-15 with various anthropometric parameters nor with body fat distribution and severity of HS, also after adjusting for age. Age is resulted to be the main factor in the prediction of IMT and thus of early atherosclerosis. The prediction of IMT by IL-15 coupled with the lack of prediction by the well-known CAD risks is in agreement with recent data, which emphasizes the main role of the immune system in the onset/worsening of atherosclerosis, even though the role of visceral adiposity should be further deepened. Age and IL-15 levels were both predictors of early atherosclerosis in this population of obese patients with NAFLD, suggesting a possible role of this cytokine in the atherosclerosis process.
Collapse
Affiliation(s)
- Giovanni Tarantino
- Department of Clinical Medicine and Surgery, Federico II University Medical School of Naples, Naples, Italy
| | - Vincenzo Citro
- Department of General Medicine, "Umberto I" Hospital, Nocera Inferiore, Italy
| | - Clara Balsano
- Department of Clinical Medicine, Life, Health and Environmental Sciences-MESVA, University of L'Aquila, L'Aquila, Italy
| | | |
Collapse
|
97
|
Nonalcoholic fatty liver and left ventricular remodelling: now the prospective evidence. J Hypertens 2021; 39:864-866. [PMID: 33824256 DOI: 10.1097/hjh.0000000000002750] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
98
|
Andreadou I, Daiber A, Baxter GF, Brizzi MF, Di Lisa F, Kaludercic N, Lazou A, Varga ZV, Zuurbier CJ, Schulz R, Ferdinandy P. Influence of cardiometabolic comorbidities on myocardial function, infarction, and cardioprotection: Role of cardiac redox signaling. Free Radic Biol Med 2021; 166:33-52. [PMID: 33588049 DOI: 10.1016/j.freeradbiomed.2021.02.012] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 02/03/2021] [Accepted: 02/06/2021] [Indexed: 02/06/2023]
Abstract
The morbidity and mortality from cardiovascular diseases (CVD) remain high. Metabolic diseases such as obesity, hyperlipidemia, diabetes mellitus (DM), non-alcoholic fatty liver disease (NAFLD) and non-alcoholic steatohepatitis (NASH) as well as hypertension are the most common comorbidities in patients with CVD. These comorbidities result in increased myocardial oxidative stress, mainly from increased activity of nicotinamide adenine dinucleotide phosphate oxidases, uncoupled endothelial nitric oxide synthase, mitochondria as well as downregulation of antioxidant defense systems. Oxidative and nitrosative stress play an important role in ischemia/reperfusion injury and may account for increased susceptibility of the myocardium to infarction and myocardial dysfunction in the presence of the comorbidities. Thus, while early reperfusion represents the most favorable therapeutic strategy to prevent ischemia/reperfusion injury, redox therapeutic strategies may provide additive benefits, especially in patients with heart failure. While oxidative and nitrosative stress are harmful, controlled release of reactive oxygen species is however important for cardioprotective signaling. In this review we summarize the current data on the effect of hypertension and major cardiometabolic comorbidities such as obesity, hyperlipidemia, DM, NAFLD/NASH on cardiac redox homeostasis as well as on ischemia/reperfusion injury and cardioprotection. We also review and discuss the therapeutic interventions that may restore the redox imbalance in the diseased myocardium in the presence of these comorbidities.
Collapse
Affiliation(s)
- Ioanna Andreadou
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece.
| | - Andreas Daiber
- Department of Cardiology 1, Molecular Cardiology, University Medical Center, Langenbeckstr. 1, 55131, Mainz, Germany; Partner Site Rhine-Main, German Center for Cardiovascular Research (DZHK), Langenbeckstr, Germany.
| | - Gary F Baxter
- Division of Pharmacology, School of Pharmacy and Pharmaceutical Sciences, Cardiff University, United Kingdom
| | | | - Fabio Di Lisa
- Department of Biomedical Sciences, University of Padova, Italy; Neuroscience Institute, National Research Council of Italy (CNR), Padova, Italy
| | - Nina Kaludercic
- Neuroscience Institute, National Research Council of Italy (CNR), Padova, Italy
| | - Antigone Lazou
- Laboratory of Animal Physiology, School of Biology, Aristotle University of Thessaloniki, Thessaloniki, 54124, Greece
| | - Zoltán V Varga
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary; HCEMM-SU Cardiometabolic Immunology Research Group, Budapest, Hungary
| | - Coert J Zuurbier
- Laboratory of Experimental Intensive Care Anesthesiology, Department Anesthesiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Rainer Schulz
- Institute of Physiology, Justus Liebig University Giessen, Giessen, Germany.
| | - Péter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary; Pharmahungary Group, Szeged, Hungary
| |
Collapse
|
99
|
Oni E, Ogunmoroti O, Allen N, A-Mallah MH, Blankstein R, Martin SS, Zeb I, Cushman M, Joshi PH, Budoff MJ, Blaha MJ, Blumenthal RS, Veledar E, Nasir K. Life's Simple 7 and Nonalcoholic Fatty Liver Disease: The Multiethnic Study of Atherosclerosis. Am J Med 2021; 134:519-525. [PMID: 33285128 DOI: 10.1016/j.amjmed.2020.09.023] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 09/11/2020] [Accepted: 09/14/2020] [Indexed: 10/22/2022]
Abstract
BACKGROUND The American Heart Association (AHA) has defined Life's Simple 7 (LS7) as a measure of overall cardiovascular health . Nonalcoholic fatty liver disease (NAFLD) has been involved as a risk factor for cardiovascular disease. We evaluated the association between LS7 and NAFLD. METHODS We evaluated participants form the Multi-Ethnic Study of Atherosclerosis (MESA) cohort. Cardiovascular health score was calculated from the Life's Simple 7 metrics. A score of 0-8 was considered inadequate, 9-10 average, and 11-14 optimal. NAFLD was defined using noncontrast cardiac computed tomography (CT) and a liver/spleen attenuation ratio (L/S) < 1. Multivariable regression were performed to evaluate the association. RESULTS Our cross-sectional analysis of 3901 participants showed 19% (n = 747) had optimal cardiovascular health, 33% (n = 1270) had average, and 48% (n = 1884) had inadequate. White participants were most likely to have an optimal score (51%, n = 378), whereas African American participants had the lowest proportion with optimal scores (16%, n = 120; P < 0.001). The overall prevalence of NAFLD was 18% with a distribution of 7%, 14%, and 25% in the optimal, average, and inadequate score categories, respectively (P < 0.001). Adjusted for risk factors, average and optimal health categories had lower odds of NAFLD compared to those with inadequate scores: odds ratio for average, 0.44 (95% confidence interval 0.36-0.54); optimal, odds ratio 0.19 (95% confidence interval 0.14-0.26). This association was similar across gender, race and age groups. CONCLUSION A more favorable cardiovascular health score was associated with a lower prevalence of NAFLD. This study may suggest a potential of Life's Simple 7 in the prevention of liver disease.
Collapse
Affiliation(s)
- Ebenezer Oni
- Division of Cardiology, Heart and Vascular Institute, Albert Einstein Medical Center, Philadelphia, Penn.
| | - Oluseye Ogunmoroti
- The Johns Hopkins Ciccarone Center for the Prevention of Cardiovascular Disease, Baltimore, Md
| | - Norrina Allen
- Department of Preventive Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Ill
| | - Mouaz H A-Mallah
- Preventive Cardiology, Houston Methodist DeBakey Cardiology Associates, Houston Tex
| | - Ron Blankstein
- Departments of Medicine (Cardiovascular Division) and Radiology, Brigham and Women's Hospital, Boston, Mass
| | - Seth S Martin
- The Johns Hopkins Ciccarone Center for the Prevention of Cardiovascular Disease, Baltimore, Md
| | - Irfan Zeb
- Division of Cardiology, Heart and Vascular Institute at West Virginia University Hospital, Morgantown, WVa
| | - Mary Cushman
- Departments of Medicine and Pathology and Laboratory Medicine, Larner College of Medicine at the University of Vermont, Burlington, Vt
| | - Parag H Joshi
- Department of Internal Medicine, Division of Cardiology, UT Southwestern Medical Center, Dallas, Tex
| | - Matthew J Budoff
- Division of Cardiology, Harbor-UCLA Medical Center, Torrance, Calif
| | - Michael J Blaha
- The Johns Hopkins Ciccarone Center for the Prevention of Cardiovascular Disease, Baltimore, Md
| | - Roger S Blumenthal
- The Johns Hopkins Ciccarone Center for the Prevention of Cardiovascular Disease, Baltimore, Md
| | - Emir Veledar
- Corporate Clinical Administration, Baptist Health South Florida, Miami, Fla
| | - Khurram Nasir
- Preventive Cardiology, Houston Methodist DeBakey Cardiology Associates, Houston Tex
| |
Collapse
|
100
|
Cognitive Dysfunction in Non-Alcoholic Fatty Liver Disease-Current Knowledge, Mechanisms and Perspectives. J Clin Med 2021; 10:jcm10040673. [PMID: 33572481 PMCID: PMC7916374 DOI: 10.3390/jcm10040673] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 01/29/2021] [Accepted: 01/31/2021] [Indexed: 12/11/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) has emerged as the hepatic component of the metabolic syndrome and now seemingly affects one-fourth of the world population. Features associated with NAFLD and the metabolic syndrome have frequently been linked to cognitive dysfunction, i.e. systemic inflammation, vascular dysfunction, and sleep apnoea. However, emerging evidence suggests that NAFLD may be a cause of cognitive dysfunction independent of these factors. NAFLD in addition exhibits dysbiosis of the gut microbiota and impaired urea cycle function, favouring systemic ammonia accumulation and further promotes systemic inflammation. Such disruption of the gut–liver–brain axis is essential in the pathogenesis of hepatic encephalopathy, the neuropsychiatric syndrome associated with progressive liver disease. Considering the growing burden of NAFLD, the morbidity from cognitive impairment is expected to have huge societal and economic impact. The present paper provides a review of the available evidence for cognitive dysfunction in NAFLD and outlines its possible mechanisms. Moreover, the clinical challenges of characterizing and diagnosing cognitive dysfunction in NAFLD are discussed.
Collapse
|