Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Diagnostics (Basel) ; 11(1)2021 Jan 09.
Article in English | MEDLINE | ID: mdl-33435415

ABSTRACT

Liver fibrosis predicts liver-related and cardiovascular outcomes in chronic liver disease patients. We compared the diagnostic performance of various liver fibrosis biomarkers for identifying histological significant/advanced fibrosis. Additionally, the correlations of such liver fibrosis biomarkers with cardiovascular risk (CVR) scores were evaluated. 173 patients with viral hepatitis (157 HCV and 16 HBV) and 107 with a non-alcoholic fatty liver disease (NAFLD) were consecutively enrolled. Various liver fibrosis biomarkers: aspartate aminotransferase (AST) to alanine aminotransferase (ALT) ratio (ARR), AST to Platelet Ratio Index (APRI), Fibrosis-4 (FiB-4), Forns index, NAFLD fibrosis score (NFS), BARD (body mass index (BMI), AAR, Diabetes) score, and Hepamet fibrosis score (HFS), were used to identify significant/advanced fibrosis. CVR was assessed by using the SCORE, the Progetto CUORE, or the Framingham risk scoring systems. Liver fibrosis biomarkers performed better in predicting advanced rather than significant liver fibrosis in all patients, regardless of chronic liver disease aetiology. Forns index and HFS performed best in predicting advanced fibrosis in patients with viral chronic liver disease and NAFLD. Lower cut-offs of these liver fibrosis biomarkers had high negative predictive values for advanced fibrosis overall, as well as in patients with NAFLD or viral chronic liver disease. FIB-4, Forns index, NFS, and HFS were positively correlated with SCORE and Framingham risk scores. In conclusion, liver fibrosis biomarkers accurately exclude advanced fibrosis and positively correlate with CVR scores in patients with chronic liver disease.

3.
Acta Diabetol ; 56(4): 385-396, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30519965

ABSTRACT

Evidence suggests a close relationship between nonalcoholic fatty liver disease (NAFLD) and type two diabetes (T2D). On the grounds of prevalence of disease, both conditions account for a significant financial cost for health care systems and individuals. Aim of this review article is to explore the epidemiological basis and the putative molecular mechanisms underlying the association of NAFLD with T2D. Epidemiological studies have shown that NAFLD is associated to the development of incident T2D and either reversal or improvement of NAFLD will result into decreased risk of developing incident T2D. On the other side of the coin data have shown that T2D will worsen the course of NAFLD doubling the risk of disease progression (i.e. evolution from simple steatosis to advanced fibrosis, cirrhosis, hepatocellular carcinoma, liver transplant and death). Conversely, NAFLD will contribute to metabolic decompensation of T2D. The pathogenesis of T2D in NAFLD patients may be mediated by several hepatokines impairing metabolic control. Among these, Fetuin-B, which causes glucose intolerance and is increased in patients with T2D and NAFLD with fibrosis is one of the most promising. T2D may affect the progression of NAFLD by acting at different levels of the pathogenic cascade involving gut microbiota and expanded, inflamed, dysfunctional adipose tissue. In conclusion, T2D and NAFLD are mutually, closely and bi-directionally associated. An improved understanding of molecular pathogenesis underlying this bi-directional association may allow us to be able to prevent the development of T2D by halting the progression of NAFLD.


Subject(s)
Diabetes Mellitus/epidemiology , Diabetes Mellitus/genetics , Non-alcoholic Fatty Liver Disease/epidemiology , Non-alcoholic Fatty Liver Disease/genetics , Biomarkers/metabolism , Diabetes Complications/diagnosis , Diabetes Complications/epidemiology , Diabetes Complications/genetics , Diabetes Complications/therapy , Diabetes Mellitus/diagnosis , Diabetes Mellitus/therapy , Diabetes Mellitus, Type 2/epidemiology , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/prevention & control , Disease Progression , Glucose Intolerance/complications , Glucose Intolerance/epidemiology , Glucose Intolerance/genetics , Glucose Intolerance/therapy , Humans , Molecular Diagnostic Techniques , Molecular Targeted Therapy/methods , Molecular Targeted Therapy/trends , Non-alcoholic Fatty Liver Disease/complications , Non-alcoholic Fatty Liver Disease/therapy , Prevalence , Risk Factors
4.
Diseases ; 6(1)2018 Feb 16.
Article in English | MEDLINE | ID: mdl-29462898

ABSTRACT

BACKGROUND AND AIMS: Nonalcoholic fatty liver disease (NAFLD) is strongly associated with both atherosclerotic cardiovascular disease (CVD) and Fetuin-A. However, the association of Fetuin-A with atherosclerosis is more controversial. We hypothesized that the pathogenic interplay of NAFLD, Fetuin-A and atherosclerosis varies based on arterial site. Accordingly, we aimed to assess NAFLD prevalence, Fetuin-A values and their relationship with symptomatic atherosclerosis occurring in different localizations: coronary artery disease (CAD) vs. peripheral arterial disease (PAD). METHODS: One hundred and forty-nine consecutive patients with symptomatic atherosclerotic CVD were recruited: 45 with CAD diagnosed by coronary angiography and 104 with PAD detected by doppler-ultrasound and/or computed tomography angiography and/or angiography. NAFLD was diagnosed based on both ultrasonography and exclusion of competing etiologies. Serum Fetuin-A was measured with ELISA. RESULTS: NAFLD was detected in 54% of the overall group, with higher rates in PAD (59%) than CAD (42%) patients. Median Fetuin-A values were 256 (111-662) µg/mL, higher in patients with CAD (378 (124-662) µg/mL) than those with PAD (236 (111-461) µg/mL). The main findings were: (1) CAD patients had higher Fetuin-A values and less frequently NAFLD than PAD patients; (2) NAFLD was positively associated with Fetuin-A values; however, this association was limited to CAD patients only; (3) Fetuin-A values were positively associated with both CAD and NAFLD. CONCLUSION: The pathogenic interplay of NAFLD, Fetuin-A and atherosclerosis probably varies according to the arterial site.

5.
Metabolism ; 72: 57-65, 2017 07.
Article in English | MEDLINE | ID: mdl-28641784

ABSTRACT

BACKGROUND AND AIMS: Fatty liver is a common feature of different types of liver diseases. The sensitivity and specificity of ultrasonography for diagnosing fatty liver are variable. A semi-quantitative ultrasound score, i.e., the ultrasonographic fatty liver indicator (US-FLI), is closely associated with metabolic/histological variables in patients with nonalcoholic fatty liver disease (NAFLD). The main aims of this study were to assess the diagnostic performance of US-FLI in detecting varying degrees of histological steatosis, and to examine the association of US-FLI with metabolic/histological parameters in 352 biopsied patients with various chronic liver diseases (173 with hepatitis C [HCV], 23 with hepatitis B [HBV], 123 with NAFLD and 33 with other etiologies). RESULTS: US-FLI accurately detected mild steatosis (minimum amount 10% on histology) with a cut-off value ≥2 (sensitivity 90.1%, specificity 90%), moderate steatosis (≥30%) with a cut-off value ≥3 (sensitivity 86.4%, specificity 92.5%) and severe steatosis (>66%) with a cut-off value ≥5 (sensitivity 88.5%, specificity 87%). US-FLI was correlated with steatosis percentage in each liver disease group as well as with lobular inflammation, ballooning, portal fibrosis, grading and staging in patients with NAFLD or HCV. US-FLI was also correlated with waist circumference, body mass index and insulin resistance both in the whole sample and in each liver disease group. CONCLUSIONS: US-FLI accurately identifies histological severity and is correlated with metabolic parameters in patients with various steatogenic liver diseases. US-FLI is an easy and versatile tool for the screening of steatosis and the metabolic health of these patients.


Subject(s)
Fatty Liver/diagnostic imaging , Liver Diseases/metabolism , Metabolomics/methods , Ultrasonography/statistics & numerical data , Adult , Fatty Liver/virology , Female , Hepatitis B/complications , Hepatitis C/complications , Humans , Insulin Resistance , Liver Diseases/virology , Male , Middle Aged , Non-alcoholic Fatty Liver Disease/diagnostic imaging , Non-alcoholic Fatty Liver Disease/metabolism , Retrospective Studies , Severity of Illness Index , Waist Circumference
6.
Adv Ther ; 34(6): 1291-1326, 2017 06.
Article in English | MEDLINE | ID: mdl-28526997

ABSTRACT

Nonalcoholic fatty liver disease (NAFLD) spans steatosis through nonalcoholic steatohepatis, cirrhosis, and hepatocellular carcinoma (HCC) associated with striking systemic features and excess cardiovascular and liver-related mortality. The pathogenesis of NAFLD is complex and multifactorial. Endocrine derangements are closely linked with dysmetabolic traits. For example, in animal and human studies, female sex is protected from dysmetabolism thanks to young individuals' ability to partition fatty acids towards ketone body production rather than very low density lipoprotein (VLDL)-triacylglycerol, and to sex-specific browning of white adipose tissue. Ovarian senescence facilitates both the development of massive hepatic steatosis and the fibrotic progression of liver disease in an experimental overfed zebrafish model. Consistently, estrogen deficiency, by potentiating hepatic inflammatory changes, hastens the progression of disease in a dietary model of nonalcoholic steatohepatitis (NASH) developing in ovariectomized mice fed a high-fat diet. In humans, NAFLD more often affects men; and premenopausal women are equally protected from developing NAFLD as they are from cardiovascular disease. It would be expected that early menarche, definitely associated with estrogen activation, would produce protection against the risk of NAFLD. Nevertheless, it has been suggested that early menarche may confer an increased risk of NAFLD in adulthood, excess adiposity being the primary culprit of this association. Fertile age may be associated with more severe hepatocyte injury and inflammation, but also with a decreased risk of liver fibrosis compared to men and postmenopausal status. Later in life, ovarian senescence is strongly associated with severe steatosis and fibrosing NASH, which may occur in postmenopausal women. Estrogen deficiency is deemed to be responsible for these findings via the development of postmenopausal metabolic syndrome. Estrogen supplementation may at least theoretically protect from NAFLD development and progression, as suggested by some studies exploring the effect of hormonal replacement therapy on postmenopausal women, but the variable impact of different sex hormones in NAFLD (i.e., the pro-inflammatory effect of progesterone) should be carefully considered.


Subject(s)
Cardiovascular Diseases/epidemiology , Estrogens/metabolism , Non-alcoholic Fatty Liver Disease/epidemiology , Non-alcoholic Fatty Liver Disease/physiopathology , Animals , Disease Progression , Female , Humans , Liver/pathology , Male , Mice , Risk Factors , Sex Distribution
7.
World J Gastroenterol ; 23(8): 1458-1468, 2017 Feb 28.
Article in English | MEDLINE | ID: mdl-28293093

ABSTRACT

AIM: To characterize natural history of cryptogenic cirrhosis (CC) and compare its clinical features and outcomes to those of hepatitis C virus (HCV)-related cirrhosis. METHODS: A prospective cohort of 102 consecutive patients at their first diagnosis of CC were enrolled in this study. The clinical data and outcomes were compared to an age- and Child-Pugh class-matched cohort of 110 patients with HCV-related cirrhosis. Diagnosis of cirrhosis was based on compatible clinical and laboratory parameters, ultrasound/endoscopic parameters and, whenever possible, on histological grounds and transient elastography. All cases of cirrhosis without a definite etiology were enrolled in the CC group. The parameters assessed were: (1) severity of liver disease at the time of first diagnosis; (2) liver decompensation during follow-up; (3) hepatocellular carcinoma (HCC); (4) orthotopic liver transplantation; and (5) death. The independent associated factors were evaluated by multiple logistic regression analysis, and survival and its determinants by the Kaplan-Meier model, log-rank test and Cox regression. RESULTS: At the first observation, median age was 66 and 65 years and male gender was 36% and 58% for CC and HCV cirrhosis, respectively. CC showed Child-Pugh class A/B/C of 47%/31%/22%, respectively. Compared to HCV cirrhosis, CC exhibited a significantly higher prevalence of metabolic syndrome (12% vs 54%, respectively), overweight/obesity, high BMI, impaired glucose tolerance, high blood pressure, dyslipidemia, hyperuricemia, cardiovascular diseases, extrahepatic cancer, and gallstones. Over a median period of 42 mo of follow-up, liver decompensation, HCC development and death for CC and HCV-related cirrhosis were 60.8%, and 54.4%, 16.7% and 17.2%, 39.2% and 30%, respectively. The median survival was 60 mo for CC. Independent predictors of death were age and Child-Pugh class at diagnosis. CC showed an approximately twofold higher incidence of HCC in Child-Pugh class A. CONCLUSION: Undiagnosed nonalcoholic fatty liver disease has an etiologic role in CC that is associated with a poor prognosis, early HCC development, high risk of cardiovascular disease and extrahepatic cancer.


Subject(s)
Hepatitis C/diagnosis , Liver Cirrhosis/congenital , Liver Cirrhosis/diagnosis , Adult , Aged , Aged, 80 and over , Carcinoma, Hepatocellular/diagnosis , Female , Follow-Up Studies , Hepatitis C/mortality , Humans , Kaplan-Meier Estimate , Liver Cirrhosis/mortality , Liver Cirrhosis/virology , Liver Failure/complications , Liver Neoplasms/diagnosis , Male , Middle Aged , Non-alcoholic Fatty Liver Disease/complications , Proportional Hazards Models , Prospective Studies , Regression Analysis , Treatment Outcome
8.
Int J Mol Sci ; 17(3): 355, 2016 Mar 09.
Article in English | MEDLINE | ID: mdl-27005620

ABSTRACT

The pathogenesis of type 2 diabetes (T2D) involves chronic hyperinsulinemia due to systemic and hepatic insulin resistance (IR), which if uncorrected, will lead to progressive pancreatic beta cell failure in predisposed individuals. Non-alcoholic fatty liver disease (NAFLD) encompasses a spectrum of fatty (simple steatosis and steatohepatitis) and non-fatty liver changes (NASH-cirrhosis with or without hepatocellular carcinoma (HCC)) that are commonly observed among individuals with multiple metabolic derangements, notably including visceral obesity, IR and T2D. Hepatitis C virus (HCV) infection is also often associated with both hepatic steatosis and features of a specific HCV-associated dysmetabolic syndrome. In recent years, the key role of the steatotic liver in the development of IR and T2D has been increasingly recognized. Thus, in this comprehensive review we summarize the rapidly expanding body of evidence that links T2D with NAFLD and HCV infection. For each of these two liver diseases with systemic manifestations, we discuss the epidemiological burden, the pathophysiologic mechanisms and the clinical implications. To date, substantial evidence suggests that NAFLD and HCV play a key role in T2D development and that the interaction of T2D with liver disease may result in a "vicious circle", eventually leading to an increased risk of all-cause mortality and liver-related and cardiovascular complications. Preliminary evidence also suggests that improvement of NAFLD is associated with a decreased incidence of T2D. Similarly, the prevention of T2D following HCV eradication in the era of direct-acting antiviral agents is a biologically plausible result. However, additional studies are required for further clarification of mechanisms involved.


Subject(s)
Diabetes Mellitus, Type 2/epidemiology , Hepatitis C/epidemiology , Liver/metabolism , Non-alcoholic Fatty Liver Disease/epidemiology , Diabetes Mellitus, Type 2/etiology , Hepatitis C/etiology , Humans , Liver/pathology , Liver/physiopathology , Non-alcoholic Fatty Liver Disease/etiology
9.
Adv Ther ; 33(3): 291-319, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26921205

ABSTRACT

Nonalcoholic fatty liver disease (NAFLD) describes steatosis, nonalcoholic steatohepatitis with or without fibrosis, and hepatocellular carcinoma, namely the entire alcohol-like spectrum of liver disease though observed in the nonalcoholic, dysmetabolic, individual free of competing causes of liver disease. NAFLD, which is a major public health issue, exhibits intrahepatic triglyceride storage giving rise to lipotoxicity. Nuclear receptors (NRs) are transcriptional factors which, activated by ligands, are master regulators of metabolism and also have intricate connections with circadian control accounting for cyclical patterns in the metabolic fate of nutrients. Several transcription factors, such as peroxisome proliferator-activated receptors, liver X receptors, farnesoid X receptors, and their molecular cascades, finely regulate energetic fluxes and metabolic pathways. Dysregulation of such pathways is heavily implicated in those metabolic derangements characterizing insulin resistance and metabolic syndrome and in the histogenesis of progressive NAFLD forms. We review the role of selected NRs in NAFLD pathogenesis. Secondly, we analyze the role of NRs in the natural history of human NAFLD. Next, we discuss the results observed in humans following administration of drug agonists or antagonists of the NRs pathogenically involved in NAFLD. Finally, general principles of treatment and lines of research in human NAFLD are briefly examined.


Subject(s)
Non-alcoholic Fatty Liver Disease/drug therapy , Non-alcoholic Fatty Liver Disease/physiopathology , Receptors, Cytoplasmic and Nuclear/agonists , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Humans , Liver X Receptors , Orphan Nuclear Receptors/metabolism , Peroxisome Proliferator-Activated Receptors/metabolism
10.
J Gastroenterol Hepatol ; 31(5): 936-44, 2016 May.
Article in English | MEDLINE | ID: mdl-26667191

ABSTRACT

BACKGROUND AND AIM: The magnitude of the risk of incident type 2 diabetes (T2D) and metabolic syndrome (MetS) among patients with nonalcoholic fatty liver disease (NAFLD) is poorly known. We gauged the risk of developing T2D and MetS in patients with NAFLD diagnosed by either serum liver enzymes (aminotransferases or gamma-glutamyltransferase [GGT]) or ultrasonography. METHODS: Pertinent prospective studies were identified through extensive electronic database research, and studies fulfilling enrolment criteria were included in the meta-analysis. RESULTS: Overall, in a pooled population of 117020 patients (from 20 studies), who were followed-up for a median period of 5 years (range: 3-14.7 years), NAFLD was associated with an increased risk of incident T2D with a pooled relative risk of 1.97 (95% confidence interval [CI], 1.80-2.15) for alanine aminotransferase, 1.58 (95% CI, 1.43-1.74) for aspartate aminotransferase, 1.86 (95% CI, 1.71-2.03) for GGT (last vs first quartile or quintile), and 1.86 (95% CI, 1.76-1.95) for ultrasonography, respectively. Overall, in a pooled population of 81411 patients (from eight studies) who were followed-up for a median period of 4.5 years (range: 3-11 years), NAFLD was associated with an increased risk of incident MetS with a pooled relative risk of 1.80 (95% CI, 1.72-1.89) for alanine aminotransferase (last vs first quartile or quintile), 1.98 (95% CI, 1.89-2.07) for GGT, and 3.22 (95% CI, 3.05-3.41) for ultrasonography, respectively. CONCLUSIONS: Nonalcoholic fatty liver disease, as diagnosed by either liver enzymes or ultrasonography, significantly increases the risk of incident T2D and MetS over a median 5-year follow-up.


Subject(s)
Diabetes Mellitus, Type 2/epidemiology , Metabolic Syndrome/epidemiology , Non-alcoholic Fatty Liver Disease/epidemiology , Alanine Transaminase/blood , Aspartate Aminotransferases/blood , Biomarkers/blood , Chi-Square Distribution , Clinical Enzyme Tests , Diabetes Mellitus, Type 2/diagnosis , Humans , Incidence , Metabolic Syndrome/diagnosis , Non-alcoholic Fatty Liver Disease/blood , Non-alcoholic Fatty Liver Disease/diagnostic imaging , Prognosis , Risk Assessment , Risk Factors , Time Factors , Ultrasonography , gamma-Glutamyltransferase/blood
11.
World J Gastroenterol ; 20(39): 14185-204, 2014 Oct 21.
Article in English | MEDLINE | ID: mdl-25339806

ABSTRACT

Nonalcoholic fatty liver disease (NAFLD) is common in the elderly, in whom it carries a more substantial burden of hepatic (nonalcoholic steatohepatitis, cirrhosis and hepatocellular carcinoma) and extra-hepatic manifestations and complications (cardiovascular disease, extrahepatic neoplasms) than in younger age groups. Therefore, proper identification and management of this condition is a major task for clinical geriatricians and geriatric hepatologists. In this paper, the epidemiology and pathophysiology of this condition are reviewed, and a full discussion of the link between NAFLD and the aspects that are peculiar to elderly individuals is provided; these aspects include frailty, multimorbidity, polypharmacy and dementia. The proper treatment strategy will have to consider the peculiarities of geriatric patients, so a multidisciplinary approach is mandatory. Non-pharmacological treatment (diet and physical exercise) has to be tailored individually considering the physical limitations of most elderly people and the need for an adequate caloric supply. Similarly, the choice of drug treatment must carefully balance the benefits and risks in terms of adverse events and pharmacological interactions in the common context of both multiple health conditions and polypharmacy. In conclusion, further epidemiological and pathophysiological insight is warranted. More accurate understanding of the molecular mechanisms of geriatric NAFLD will help in identifying the most appropriate diagnostic and therapeutic approach for individual elderly patients.


Subject(s)
Aging , Non-alcoholic Fatty Liver Disease/epidemiology , Non-alcoholic Fatty Liver Disease/therapy , Adult , Age Factors , Aged , Aged, 80 and over , Aging/metabolism , Cardiovascular Diseases/epidemiology , Comorbidity , Dementia/epidemiology , Frail Elderly , Geriatric Assessment , Host-Pathogen Interactions , Humans , Intestines/microbiology , Life Style , Middle Aged , Non-alcoholic Fatty Liver Disease/diagnosis , Non-alcoholic Fatty Liver Disease/physiopathology , Polypharmacy , Prognosis , Risk Assessment , Risk Factors , Sarcopenia/epidemiology
12.
World J Gastroenterol ; 20(23): 7089-103, 2014 Jun 21.
Article in English | MEDLINE | ID: mdl-24966582

ABSTRACT

Hepatitis C virus (HCV) is a successful pathogen on the grounds that it exploits its host's metabolism to build up viral particles; moreover it favours its own survival by inducing chronic disease and the development of specific anatomic changes in the infected organ. Steatosis, therefore, is associated with HCV infection by necessity rather than by chance alone. Approximately 6% of HCV patients have steatohepatitis. Interestingly, HCV steatosis occurs in the setting of multiple metabolic abnormalities (hyperuricemia, reversible hypocholesterolemia, insulin resistance, arterial hypertension and expansion of visceral adipose tissue) collectively referred to as "hepatitis C-associated dysmetabolic syndrome" (HCADS). General, nonalcoholic fatty liver disease (NAFLD)-like, mechanisms of steatogenesis (including increased availability of lipogenic substrates and de novo lipogenesis; decreased oxidation of fatty substrates and export of fatty substrates) are shared by all HCV genotypes. However, genotype 3 seemingly amplifies such steatogenic molecular mechanisms reported to occur in NAFLD via more profound changes in microsomal triglyceride transfer protein; peroxisome proliferator-activated receptor alpha; sterol regulatory element-binding proteins and phosphatase and tensin homologue. HCV steatosis has a remarkable clinical impact in as much as it is an acknowledged risk factor for accelerated fibrogenesis; for impaired treatment response to interferon and ribavirin; and development of hepatocellular carcinoma. Recent data, moreover, suggest that HCV-steatosis contributes to premature atherogenesis via both direct and indirect mechanisms. In conclusion, HCV steatosis fulfills all expected requirements necessary to perpetuate the HCV life cycle. A better understanding of the physiology of HCADS will likely result in a more successful handling of disease with improved antiviral success rates.


Subject(s)
Fatty Liver/virology , Hepacivirus/pathogenicity , Hepatitis C/virology , Liver/virology , Animals , Antiviral Agents/therapeutic use , Atherosclerosis/metabolism , Atherosclerosis/virology , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/virology , Fatty Liver/diagnosis , Fatty Liver/drug therapy , Fatty Liver/metabolism , Hepacivirus/drug effects , Hepacivirus/growth & development , Hepatitis C/complications , Hepatitis C/diagnosis , Hepatitis C/drug therapy , Hepatitis C/metabolism , Host-Pathogen Interactions , Humans , Lipid Metabolism , Liver/drug effects , Liver/metabolism , Liver/pathology , Liver Cirrhosis/metabolism , Liver Cirrhosis/virology , Liver Neoplasms/metabolism , Liver Neoplasms/virology , Oxidative Stress , Prognosis , Risk Factors
13.
Clin Interv Aging ; 9: 425-32, 2014.
Article in English | MEDLINE | ID: mdl-24669190

ABSTRACT

BACKGROUND: The modifications of cholesterol metabolism associated with aging are ill-defined. The objective of this study was to define age-associated alterations of the different metabolic pathways controlling cholesterol homeostasis by analyzing circulating sterols. METHODS: We analyzed serum samples collected from 201 adult (75 male, 126 female) subjects within the epidemiological MICOL study (Multicentrica Italiana Colelitiasi). The age range was 38-79 years; 103 had evidence of gallstones. The concentrations of the different sterols, recognized as markers of the main pathways of cholesterol homeostasis, were analyzed by gas chromatography-mass spectrometry, including lathosterol (synthesis), campesterol and sitosterol (absorption), and 7α-hydroxy-4-cholesten-3-one (degradation to bile acids). RESULTS: A significant direct correlation was detected between age and cholesterol levels (r =0.34, P<0.01). The lathosterol/cholesterol ratio was lower in older age quartiles (P<0.05 by analysis of variance), with an inverse correlation between the lathosterol/cholesterol ratio and age (r=-0.32, P<0.01). Such correlation was particularly evident in females. The campesterol/cholesterol and sitosterol/cholesterol ratios were inversely correlated with aging in control, but not in gallstone patients. The levels of 7α-hydroxy-4-cholesten-3-one were not correlated with age. CONCLUSION: These data show a reduction of cholesterol synthesis with aging which is associated with increased circulating cholesterol levels. The finding might be related to a reduced metabolic need for cholesterol in advancing age, leading to a downregulation of the main mechanisms of cholesterol intake in the liver. A different age-related behavior was observed in gallstone-free versus gallstone patients regarding cholesterol absorption. The possible implications in terms of the pharmacological management of hypercholesterolemia in the elderly remain to be defined.


Subject(s)
Aging/metabolism , Cholesterol/metabolism , Homeostasis , Adult , Age Factors , Aged , Aging/physiology , Cholestenones/blood , Cholesterol/analogs & derivatives , Cholesterol/blood , Cholesterol/physiology , Cross-Sectional Studies , Female , Gallstones/blood , Gallstones/metabolism , Gallstones/physiopathology , Gas Chromatography-Mass Spectrometry , Homeostasis/physiology , Humans , Italy/epidemiology , Male , Middle Aged , Phytosterols/blood , Sitosterols/blood
14.
JPEN J Parenter Enteral Nutr ; 38(2): 220-6, 2014 Feb.
Article in English | MEDLINE | ID: mdl-23426743

ABSTRACT

BACKGROUND: Artificial nutrition is frequently associated with hepatobiliary complications, probably due to the inherent derangement of the gastrointestinal tract physiology. Alterations of hepatic lipid metabolism are likely to be involved. The aim of the present study was to investigate the effect of artificial nutrition on bile acid production, a key event in cholesterol homeostasis, in humans. PATIENTS AND METHODS: Eleven patients receiving artificial nutrition, either parenteral nutrition (PN; n = 6) or enteral nutrition (EN; n = 5) with no previous history of liver disease, underwent analysis of cholesterol 7α-hydroxylation rates in vivo, a measure of bile acid formation, by isotope release analysis after intravenous injection of [7α-(3)H]cholesterol. The results were compared with those obtained in a population of 16 age-matched control subjects. RESULTS: Hydroxylation rates were lower in patients with artificial nutrition (PN: 94 ± 13 mg/d; EN: 230 ± 39 mg/d, mean ± SEM) when compared with controls (385 ± 47 mg/d) (P < .01, 1-way analysis of variance). In a patient receiving EN, hydroxylation rates increased 3.5-fold after treatment with the cholecystokinin analogue ceruletide (20 µg bid for 2 weeks intramuscularly). Serum lathosterol-to-cholesterol ratio, a marker of cholesterol synthesis, was also significantly reduced in artificial nutrition, whereas serum levels of fibroblast growth factor 19 (FGF19) were increased. CONCLUSION: In vivo 7α-hydroxylation is suppressed in artificial nutrition, particularly in PN. The finding associates with reduced cholesterol production, possibly as a metabolic consequence. The data suggest a regulatory role of gastrointestinal hormones and FGF19 on bile acid production and might suggest a pathophysiological basis for some common complications of artificial nutrition, such as gallstone disease and cholestasis.


Subject(s)
Bile Acids and Salts/blood , Cholesterol/blood , Enteral Nutrition , Parenteral Nutrition , Administration, Intravenous , Adult , Aged , Body Mass Index , Body Weight , Cholecystokinin/therapeutic use , Enteral Nutrition/adverse effects , Female , Fibroblast Growth Factors/blood , Gastrointestinal Hormones/metabolism , Gastrointestinal Tract/metabolism , Homeostasis , Humans , Hydroxylation , Linear Models , Lipid Metabolism , Liver/metabolism , Liver Diseases/therapy , Male , Middle Aged , Parenteral Nutrition/adverse effects
15.
Metab Syndr Relat Disord ; 11(4): 289-95, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23600632

ABSTRACT

BACKGROUND: Nonalcoholic fatty liver disease (NAFLD) patients are prone to coronary artery disease (CAD). Fetuin-A inhibits arterial calcification, induces insulin resistance, and is increased in NAFLD. Data on fetuin-A levels in CAD are conflicting. We tried to ascertain whether NAFLD and CAD are associated and if fetuin-A predicts CAD and/or NAFLD. METHODS: CAD was diagnosed by ≥50% stenosis in coronary arteries and NAFLD by ultrasound imaging in the absence of any other liver disease. Seventy patients who underwent elective coronarography at our hospital were recruited in this cross-sectional study. Twenty-four patients had no CAD (9 with and 15 without NAFLD) and 46 had CAD (20 with and 26 without NAFLD). Standard anthropometric indices and metabolic parameters were recorded. Fetuin-A was determined by enzyme-linked immunosorbent assay (ELISA). Visceral fat thickness and visceral/subcutaneous fat ratio were assessed by ultrasonography. RESULTS: NAFLD was not associated with CAD, probably owing to the limited series. Fetuin-A was significantly lower, whereas visceral fat thickness and visceral/subcutaneous fat ratio were higher in patients with CAD versus those without CAD. Younger age and higher body mass index (BMI), waist circumference, triglycerides, fasting glucose, homeostasis model assessment, spleen area, subcutaneous fat thickness, and prevalence of metabolic derangements were associated with NAFLD. At multivariate analysis, elevated fetuin-A levels were an independent negative predictor of CAD [odds ratio (OR)=0.995, P=0.049]. Fetuin-A was an independent predictor of NAFLD (OR=1.005, P=0.036) in the model including BMI. CONCLUSIONS: This prospective cross-sectional study demonstrates high fetuin-A levels to be independently associated with NAFLD and a lower risk of coronarographically diagnosed CAD.


Subject(s)
Coronary Artery Disease/blood , Coronary Artery Disease/complications , Fatty Liver/blood , Fatty Liver/complications , alpha-2-HS-Glycoprotein/metabolism , Aged , Aged, 80 and over , Biomarkers/blood , Coronary Angiography , Coronary Artery Disease/diagnostic imaging , Cross-Sectional Studies , Fatty Liver/diagnostic imaging , Female , Humans , Intra-Abdominal Fat/pathology , Male , Middle Aged , Multivariate Analysis , Non-alcoholic Fatty Liver Disease , Prospective Studies , Risk Factors , Subcutaneous Fat/pathology
16.
Curr Pharm Des ; 19(29): 5280-96, 2013.
Article in English | MEDLINE | ID: mdl-23394096

ABSTRACT

BACKGROUND: Nonalcoholic fatty liver disease (NAFLD), the most common chronic liver disorder worldwide, comprises a spectrum of conditions ranging from simple steatosis to nonalcoholic steatohepatitis (NASH) and cirrhosis. NASH is associated with an increased risk of hepatocellular carcinoma (HCC) and cardiometabolic disease. Insulin resistance (IR) is the underlying pathogenic mechanism for NAFLD, the presence of which in turn, is a strong predictor for the development of metabolic disorders. Hence, therapy of NAFLD with insulin-sensitizing drugs (ISDs) should ideally improve the key hepatic histological changes (steatosis, inflammation and fibrosis), but should also reduce cardiometabolic and cancer risk. OBJECTIVES: In this review, the rationale for the use of ISDs and the evidence for their efficacy are detailed. In particular, the mechanism of action, potential for use, limitations and untoward effects of metformin and thiazolidinediones are systematically reviewed. Further, we discuss novel ISDs that may have potential clinical utility in NAFLD. RESULTS AND CONCLUSION: Despite the theoretical prediction that ISDs might have beneficial effects on disease outcomes, evidence that ISDs are able to alter the natural history of NAFLD are presently not available. The exploration of novel strategies exploiting "nonconventional" ISDs is encouraged.


Subject(s)
Fatty Liver/prevention & control , Hypoglycemic Agents/therapeutic use , Metformin/therapeutic use , Carcinoma, Hepatocellular/etiology , Child , Fatty Liver/complications , Humans , Liver Neoplasms/etiology , Non-alcoholic Fatty Liver Disease
17.
Dig Liver Dis ; 45(3): 226-32, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23022423

ABSTRACT

BACKGROUND: Genetic alterations in the ATP-binding cassette subfamily B member 4 (ABCB4) and ATP-binding cassette subfamily B member 11 (ABCB11) have been associated to the onset of intrahepatic cholestasis of pregnancy (ICP) in predisposed women. AIMS: To identify new and/or frequent ABCB4 and ABCB11 genes variants in a cohort of Italian patients with ICP and to evaluate the possible pathogenetic role for the novel mutations identified. METHODS: DNA of 33 unrelated Italian women with obstetric cholestasis were screened for mutations in the entire coding sequence of ABCB4 and ABCB11 genes. Polymerase chain reaction and automated sequencing was performed on the 27 coding exons of both genes. RESULTS: Genotyping revealed 11 mutations, 5 of whom were novel variants: 2 localized on ABCB4 (p.I587DfsX603, p.I738LfsX744) and 3 on ABCB11 (p.V284D, p.Q558H, p.P731S). The most severe phenotypes were associated with the variants p.I587DfsX603, p.I738LfsX744 and p.V284D. Moreover, the already described mutation p.N510S found in ABCB4 seems to be strictly involved in the onset of ICP in that particular patient. CONCLUSIONS: Our data support the hypothesis of a significant involvement of ABCB4 mutations in the onset of ICP, but also confirm an important role for ABCB11 mutations in increasing the susceptibility to cholestasis of pregnancy.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B/genetics , ATP-Binding Cassette Transporters/genetics , Cholestasis, Intrahepatic/genetics , Pregnancy Complications/genetics , ATP Binding Cassette Transporter, Subfamily B, Member 11 , Adult , Case-Control Studies , Cholestasis, Intrahepatic/ethnology , Cohort Studies , Female , Genetic Association Studies , Genetic Predisposition to Disease , Humans , Italy , Mutation , Pregnancy , Pregnancy Complications/ethnology , Severity of Illness Index , White People
18.
Neuromolecular Med ; 12(3): 248-59, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20033627

ABSTRACT

Patients affected by recurrent seizures frequently present increased homocysteine plasma levels in consequence of treatment with antiepileptic drugs. Homocysteine is proconvulsant and can affect the response to antiepileptic drugs. In addition, high homocysteine plasma levels represent a risk factor for cardiovascular and neurodegenerative diseases. To better define the role of increased homocysteine in epilepsy, we analyzed the effects of homocysteine pretreatment in the pilocarpine model of status epilepticus (SE), which is used to mimic temporal lobe epilepsy (TLE) in rodents. Precisely, we investigated whether a moderate hyperhomocysteinemia, unable to cause seizures, could sensitize rats to pilocarpine and cooperate in inducing brain lesions. We found that a subthreshold dose of pilocarpine (200 mg/kg) is sufficient to induce SE in the majority (approximately 90%) of rats pretreated with homocysteine for 2 weeks, whereas only 40% of saline-treated controls developed SE following the same pilocarpine dose. Furthermore, homocysteine pretreatment led to a significant increase in neuronal cell loss evaluated by counting toluidine blue-stained or Fluoro-Jade-positive cells in hippocampal and parahippocampal regions. Pilocarpine augmented amyloid beta expression in both animal groups. However, pretreatment with homocysteine favored the intraneuronal fibrillar conformation of amyloid beta, thus promoting neurodegeneration. These findings indicate that increased homocysteine levels enhance seizure activity and neurodegeneration in pilocarpine-treated rats and suggest that similar detrimental effects may occur in patients affected by TLE.


Subject(s)
Epilepsy, Temporal Lobe , Homocysteine , Nerve Degeneration/chemically induced , Pilocarpine/pharmacology , Status Epilepticus , Amyloid beta-Peptides/metabolism , Animals , Behavior, Animal/drug effects , Disease Models, Animal , Epilepsy, Temporal Lobe/blood , Epilepsy, Temporal Lobe/pathology , Fluoresceins , Hippocampus/cytology , Hippocampus/drug effects , Hippocampus/pathology , Homocysteine/blood , Homocysteine/pharmacology , Humans , Male , Organic Chemicals/metabolism , Rats , Rats, Sprague-Dawley , Status Epilepticus/blood , Status Epilepticus/chemically induced , Status Epilepticus/pathology
19.
Epilepsia ; 50 Suppl 1: 53-8, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19125849

ABSTRACT

PURPOSE: Cytochrome P450 cholesterol side-chain cleavage enzyme (P450scc) catalyzes the initial step in the biosynthesis of neurosteroids within the brain. We sought to determine which cells express P450cc and whether neurosteroids play a role in the regulation of epileptogenesis following pilocarpine-induced status epilepticus (SE). METHODS: Rats experienced uninterrupted SE or SE terminated with diazepam at 60, 120, and 180 min. P450scc induction in CA3 hippocampus was determined by double immunolabeling with P450scc antiserum and monoclonal antibodies against GFAP (astrocytes), RIP (oligodendrocytes), or heme oxygenase-1 (microglia). RESULTS: SE was associated with P450scc induction in many astrocytes and a small number of microglia and oligodendrocytes in the hippocampal CA3 strata radiatum and lacunosum-moleculare. The extent of P450scc induction increased with increasing SE duration. Paradoxically, increased P450scc induction in rats experiencing SE for 180 min or more was associated with the delayed onset of spontaneous recurrent seizures. Treatment with the 5 alpha-reductase inhibitor finasteride (100 mg/kg/day for 25 days), which inhibits the synthesis of gamma-aminobutyric acid (GABA)(A) receptor modulating neurosteroids such as allopregnanolone, was associated with a significant reduction in time to the onset of spontaneous seizures in rats exposed to 180-min but not 90-min SE. DISCUSSION: P450scc is induced by SE in a diverse population of hippocampal glia. Induction of P450scc is associated with the delayed onset of spontaneous seizures. Conversely, inhibition of neurosteroid synthesis accelerated the onset of spontaneous seizures, but only in animals exhibiting significant increases in P450scc. These findings suggest that induction of neurosteroid synthesis in reactive glial cells is associated with delayed onset of spontaneously recurrent seizures.


Subject(s)
Cholesterol Side-Chain Cleavage Enzyme/biosynthesis , Disease Models, Animal , Neurotransmitter Agents/biosynthesis , Pilocarpine/toxicity , Reaction Time/physiology , Status Epilepticus/enzymology , Animals , Cholesterol Side-Chain Cleavage Enzyme/physiology , Enzyme Induction/drug effects , Enzyme Induction/physiology , Male , Neurotransmitter Agents/physiology , Rats , Rats, Sprague-Dawley , Reaction Time/drug effects , Receptors, GABA-A/biosynthesis , Receptors, GABA-A/physiology , Recurrence , Seizures/enzymology , Seizures/metabolism , Seizures/physiopathology , Status Epilepticus/chemically induced , Status Epilepticus/metabolism
20.
J Neuropathol Exp Neurol ; 67(7): 687-701, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18596544

ABSTRACT

In limbic seizures, neuronal excitation is conveyed from the entorhinal cortex directly to CA1 and subicular regions. This phenomenon is associated with a reduced ability of CA3 to respond to entorhinal cortex inputs. Here, we describe a lesion that destroys the perforant path in CA3 after status epilepticus (SE) induced by pilocarpine injection in 8-week-old rats. Using magnetic resonance imaging, immunohistochemical, and ultrastructural analyses, we determined that this lesion develops after 30 minutes of SE and is characterized by microhemorrhages and ischemia. After a longer period of SE, the lesion invariably involves the upper blade of the dentate gyrus. Adult rats treated with subcutaneous diazepam (20 mg kg for 3 days) did not develop the dentate gyrus lesion and had less frequent spontaneous recurrent seizures (p < 0.01). Young (3-week-old) rats rarely (20%) developed the CA3 lesion, and their spontaneous seizures were delayed (p < 0.01). To investigate the role of the damaged CA3 in seizure activity, we reinduced SE in adult and young epileptic rats. Using FosB/DeltaFosB markers, we found induction of FosB/DeltaFosB immunopositivity in CA3 neurons of young but not in adult rats. These experiments indicate that SE can produce a focal lesion in the perforant path that may affect the roles of the hippocampus in epileptic rats.


Subject(s)
Brain Injuries/pathology , Entorhinal Cortex/pathology , Hippocampus/pathology , Status Epilepticus/pathology , Age Factors , Animals , Animals, Newborn , Anticonvulsants/therapeutic use , Brain Injuries/chemically induced , Brain Injuries/complications , Brain Injuries/drug therapy , Diazepam/therapeutic use , Disease Models, Animal , Entorhinal Cortex/drug effects , Entorhinal Cortex/ultrastructure , Gene Expression Regulation/drug effects , Guanine Nucleotide Exchange Factors , Heme Oxygenase-1/metabolism , Hippocampus/drug effects , Hippocampus/ultrastructure , Laminin/metabolism , Magnetic Resonance Imaging/methods , Male , Microscopy, Electron, Transmission/methods , Nerve Tissue Proteins/metabolism , Neural Pathways/pathology , Neural Pathways/ultrastructure , Pilocarpine , Proto-Oncogene Proteins , Proto-Oncogene Proteins c-fos/metabolism , Rats , Rats, Sprague-Dawley , Rho Guanine Nucleotide Exchange Factors , Status Epilepticus/chemically induced , Status Epilepticus/complications , Status Epilepticus/drug therapy , Status Epilepticus/prevention & control
SELECTION OF CITATIONS
SEARCH DETAIL