Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 816
Filter
1.
Bull Exp Biol Med ; 177(3): 395-400, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39134815

ABSTRACT

We compared 2 models of metabolic syndrome in rats: high-fat diet (58% calories) with single streptozotocin injection at a dose of 25 mg/kg and replacement of water with 20% fructose solution. The model with fructose solution did not cause the main signs of metabolic syndrome over 24 weeks: concentrations of glucose, triglycerides, cholesterol, weight, and BP did not significantly differ from the control group (standard diet). At the same time, single streptozotocin administration was followed by the development of persistent hyperglycemia, hypertriglyceridemia, hypercholesterolemia, and signs of visceral obesity. High-fat diet combined with injection of streptozotocin in a low dose can be considered a more representative model of metabolic syndrome in humans.


Subject(s)
Blood Glucose , Diet, High-Fat , Metabolic Syndrome , Streptozocin , Triglycerides , Animals , Diet, High-Fat/adverse effects , Rats , Male , Metabolic Syndrome/metabolism , Triglycerides/blood , Triglycerides/metabolism , Blood Glucose/metabolism , Rats, Wistar , Hyperglycemia/metabolism , Hyperglycemia/chemically induced , Cholesterol/blood , Cholesterol/metabolism , Body Weight/drug effects , Fructose/administration & dosage , Hypertriglyceridemia/metabolism , Hypertriglyceridemia/chemically induced , Hypertriglyceridemia/blood , Hypertriglyceridemia/etiology , Hypercholesterolemia/metabolism , Hypercholesterolemia/etiology , Dietary Carbohydrates/administration & dosage , Blood Pressure/drug effects
2.
Int J Mol Sci ; 25(16)2024 Aug 13.
Article in English | MEDLINE | ID: mdl-39201497

ABSTRACT

Hypertriglyceridemia is a risk factor for type 2 diabetes and cardiovascular disease (CVD). Plasma triglycerides (TGs) are a key factor for assessing the risk of diabetes or CVD. However, previous lipidomics studies have demonstrated that not all TG molecules behave the same way. Individual TGs with different fatty acid compositions are regulated differentially under various conditions. In addition, distinct groups of TGs were identified to be associated with increased diabetes risk (TGs with lower carbon number [C#] and double-bond number [DB#]), or with decreased risk (TGs with higher C# and DB#). In this study, we examined the effects of high-fat feeding in rats on plasma lipid profiles with special attention to TG profiles. Wistar rats were maintained on either a low-fat (control) or high-fat diet (HFD) for 2 weeks. Plasma samples were obtained before and 2.5 h after a meal (n = 10 each) and subjected to lipidomics analyses. High-fat feeding significantly impacted circulating lipid profiles, with the most significant effects observed on TG profile. The effects of an HFD on individual TG species depended on DB# in their fatty acid chains; an HFD increased TGs with low DB#, associated with increased diabetes risk, but decreased TGs with high DB#, associated with decreased risk. These changes in TGs with an HFD were associated with decreased indices of hepatic stearoyl-CoA desaturase (SCD) activity, assessed from hepatic fatty acid profiles. Decreased SCD activity would reduce the conversion of saturated to monounsaturated fatty acids, contributing to the increases in saturated TGs or TGs with low DB#. In addition, an HFD selectively depleted ω-3 polyunsaturated fatty acids (PUFAs), contributing to the decreases in TGs with high DB#. Thus, an HFD had profound impacts on circulating TG profiles. Some of these changes were at least partly explained by decreased hepatic SCD activity and depleted ω-3 PUFA.


Subject(s)
Diet, High-Fat , Fatty Acids, Omega-3 , Rats, Wistar , Triglycerides , Animals , Triglycerides/blood , Triglycerides/metabolism , Fatty Acids, Omega-3/metabolism , Fatty Acids, Omega-3/blood , Diet, High-Fat/adverse effects , Rats , Male , Fatty Acids, Nonesterified/blood , Fatty Acids, Nonesterified/metabolism , Stearoyl-CoA Desaturase/metabolism , Hypertriglyceridemia/metabolism , Hypertriglyceridemia/blood , Hypertriglyceridemia/etiology , Lipidomics
3.
J Appl Lab Med ; 9(4): 728-740, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38574000

ABSTRACT

BACKGROUND: Although low high-density lipoprotein cholesterol (HDL-C) levels are a common metabolic abnormality associated with insulin resistance, their role in cardiovascular risk stratification remains controversial. Recently, we developed a simple, high-throughput, cell-free assay system to evaluate the "cholesterol uptake capacity (CUC)" as a novel concept for HDL functionality. In this study, we assessed the CUC in patients with hypertriglyceridemia and diabetes mellitus. METHODS: The CUC was measured using cryopreserved serum samples from 285 patients who underwent coronary angiography or percutaneous coronary intervention between December 2014 and May 2019 at Kobe University Hospital. RESULTS: The CUC was significantly lower in diabetic patients (n = 125) than in nondiabetic patients (93.0 vs 100.7 arbitrary units (A.U.), P = 0.002). Patients with serum triglyceride (TG) levels >150 mg/dL (n = 94) also had a significantly lower CUC (91.8 vs 100.0 A.U., P = 0.004). Furthermore, the CUC showed a significant inverse correlation with TG, hemoglobin A1c (Hb A1c), homeostasis model assessment of insulin resistance (HOMA-IR), and body mass index (BMI). Finally, the HDL-C/Apolipoprotein A1 (ApoA1) ratio, calculated as a surrogate index of HDL particle size, was significantly positively correlated with the CUC (r2 = 0.49, P < 0.001), but inversely correlated with TG levels (r2 = -0.30, P < 0.001). CONCLUSIONS: The CUC decreased in patients with hypertriglyceridemia and diabetes mellitus, and HDL particle size was a factor defining the CUC and inversely correlated with TG levels, suggesting that impaired CUC in insulin-resistant states was partially due to the shift in HDL towards smaller particles. These findings provide a better understanding of the mechanisms underlying impaired HDL functionality.


Subject(s)
Cholesterol, HDL , Hypertriglyceridemia , Insulin Resistance , Triglycerides , Humans , Hypertriglyceridemia/blood , Hypertriglyceridemia/diagnosis , Hypertriglyceridemia/complications , Hypertriglyceridemia/etiology , Male , Female , Middle Aged , Aged , Cholesterol, HDL/blood , Triglycerides/blood , Diabetes Mellitus/blood , Diabetes Mellitus/diagnosis , Apolipoprotein A-I/blood , Cholesterol/blood , Glycated Hemoglobin/metabolism , Glycated Hemoglobin/analysis
4.
BMC Endocr Disord ; 24(1): 47, 2024 Apr 15.
Article in English | MEDLINE | ID: mdl-38622573

ABSTRACT

BACKGROUND: Familial chylomicronemia syndrome (FCS) is a rare monogenic form of severe hypertriglyceridemia, caused by mutations in genes involved in triglyceride metabolism. Herein, we report the case of a Korean family with familial chylomicronemia syndrome caused by compound heterozygous deletions of glycosylphosphatidylinositol-anchored high-density lipoprotein-binding protein 1 (GPIHBP1). CASE PRESENTATION: A 4-year-old boy was referred for the evaluation of severe hypertriglyceridemia (3734 mg/dL) that was incidentally detected 4 months prior. His elder brother also demonstrated an elevated triglyceride level of 2133 mg/dL at the age of 9. Lipoprotein electrophoresis revealed the presence of chylomicrons, an increase in the proportion of pre-beta lipoproteins, and low serum lipoprotein lipase levels. The patient's parents and first elder brother had stable lipid profiles. For suspected FCS, genetic testing was performed using the next-generation sequencing-based analysis of 31 lipid metabolism-associated genes, which revealed no pathogenic variants. However, copy number variant screening using sequencing depth information suggested large heterozygous deletion encompassing all the coding exons of GPIHBP1. A real-time quantitative polymerase chain reaction was performed to validate the deletion site. The results showed that the siblings had two heterozygous copy number variants consisting of the whole gene and an exon 4 deletion, each inherited from their parents. During the follow-up period of 17 months, the patient did not develop pancreatitis, following dietary intervention. CONCLUSION: These siblings' case of familial chylomicronemia syndrome caused by rare GPIHBP1 deletions highlight the implementation of copy number variants-beyond next-generation sequencing-as an important consideration in diagnosis. Accurate genetic diagnosis is necessary to establish the etiology of severe hypertriglyceridemia, which increases the risk of pancreatitis.


Subject(s)
Hyperlipoproteinemia Type I , Hypertriglyceridemia , Pancreatitis , Receptors, Lipoprotein , Child, Preschool , Humans , Male , Hyperlipoproteinemia Type I/diagnosis , Hyperlipoproteinemia Type I/genetics , Hypertriglyceridemia/etiology , Lipoprotein Lipase/genetics , Lipoprotein Lipase/metabolism , Receptors, Lipoprotein/genetics , Receptors, Lipoprotein/chemistry , Receptors, Lipoprotein/metabolism , Siblings , Triglycerides , Child
5.
Acta Biochim Pol ; 70(4): 823-828, 2023 Dec 03.
Article in English | MEDLINE | ID: mdl-38043131

ABSTRACT

INTRODUCTION: Liver transplantation (LTx) is the only successful treatment for end-stage liver disease. The results of liver transplantation depend not only on graft survival but may be also affected by superimposed cardiovascular morbidities. The aim of this retrospective study was to assess the prevalence of lipid disorders as one of the important cardiovascular risk factors in patients before and after successful LTx. MATERIAL AND METHODS: One hundred eleven patients who underwent liver transplantation because of liver cirrhosis and survived at least 2 years with functioning graft between November 2005 and May 2014 were included in this retrospective analysis. The mean age of the patients at the time of liver transplantation was 49.7±12.2 years. The prevalence of dyslipidemia was assessed before and two years after liver transplantation. This was analyzed in relation to the etiology of liver disease, including alcohol toxicity, viral or autoimmune diseases. RESULTS: The prevalence of hypertriglyceridemia before and after LTx was 13.5% and 40.5%, respectively (P<0.001). Similarly, hypercholesterolemia was noted in 17.1% and 51.4% respectively (P<0.001). The annual incidence of hypertriglyceridemia and hypercholesterolemia during the first two years after LTx was 16.2% and 20.7%, respectively. The prevalence of hypertriglyceridemia (18.5% vs 66.7%, P<0.001) and hypercholesterolemia (29.6% vs 70.0%, P=0.002) was significantly lower in patients with the autoimmune cause of liver cirrhosis in comparison to patients with the alcoholic liver disease. CONCLUSIONS: The prevalence of dyslipidemia is increased after liver transplantation. The prevalence of dyslipidemia may be related to the cause of liver injury before LTx.


Subject(s)
Hypercholesterolemia , Hypertriglyceridemia , Liver Transplantation , Humans , Adult , Middle Aged , Liver Transplantation/adverse effects , Retrospective Studies , Hypercholesterolemia/etiology , Liver Cirrhosis/epidemiology , Liver Cirrhosis/surgery , Hypertriglyceridemia/etiology , Lipids
6.
J Atheroscler Thromb ; 30(12): 1778-1790, 2023 Dec 01.
Article in English | MEDLINE | ID: mdl-37100628

ABSTRACT

AIM: This study aimed to investigate the association between a combination of elevated triglyceride (TG) and reduced high-density lipoprotein cholesterol (HDL-C) levels and target lesion revascularization (TLR) following everolimus-eluting stent (EES) implantation. The adverse impact of clinical, lesion, and procedural characteristics on TLR in patients with elevated TG and reduced HDL-C levels was also assessed. METHODS: We retrospectively collected data on 3,014 lesions from 2,022 consecutive patients, who underwent EES implantation at Koto Memorial Hospital. Atherogenic dyslipidemia (AD) is defined as a combination of non-fasting serum TG ≥ 175 mg/dL and HDL-C <40 mg/dL. RESULTS: AD was observed in 212 lesions in 139 (6.9%) patients. The cumulative incidence of clinically driven TLR was significantly higher in patients with AD than in those without AD (hazard ratio [HR] 2.31, 95% confidence interval [CI] 1.43-3.73, P=0.0006). Subgroup analysis showed that AD increased the risk of TLR with the implantation of small stents (≤ 2.75 mm). Multivariable Cox regression analysis showed that AD was an independent predictor of TLR in the small EES stratum (adjusted HR 3.00, 95% CI 1.53-5.93, P=0.004), whereas the incidence of TLR was similar in the non-small-EES stratum, irrespective of the presence or absence of AD. CONCLUSIONS: Patients with AD had a higher risk of TLR after EES implantation, and this risk was greater for lesions treated with small stents.


Subject(s)
Coronary Artery Disease , Drug-Eluting Stents , Hypertriglyceridemia , Myocardial Infarction , Percutaneous Coronary Intervention , Humans , Everolimus , Sirolimus/therapeutic use , Myocardial Infarction/etiology , Drug-Eluting Stents/adverse effects , Lipoproteins, HDL , Lipoproteins, LDL , Retrospective Studies , Treatment Outcome , Percutaneous Coronary Intervention/adverse effects , Stents/adverse effects , Hypertriglyceridemia/etiology , Hypertriglyceridemia/drug therapy , Risk Factors , Coronary Artery Disease/complications
7.
Clin Investig Arterioscler ; 34(6): 326-329, 2022.
Article in English, Spanish | MEDLINE | ID: mdl-36184299

ABSTRACT

Familial chylomicronemia syndrome (FCS) is a genetic entity with autosomal recessive inheritance. Mutations in genes (such as APOC2, APOAV, LMF-1, GPIHBP-1) that code for proteins that regulate the maturation, transport, or polymerization of lipoprotein lipase-1 are the most common causes, but not the only ones. The objective of this study was to report the first documented case in Ecuador. CLINICAL CASE: A 38-year-old man presented with chronic hepatosplenomegaly, thrombocytopenia, pancreatic atrophy, and severe hypertriglyceridemia refractory to treatment. A molecular analysis was performed by next generation sequencing that determined a deficiency of Lipoprotein Lipase OMIM #238600 in homozygosis. Genetic confirmation is necessary in order to establish the etiology of HTGS for an adequate management of this pathology.


Subject(s)
Hyperlipoproteinemia Type I , Hypertriglyceridemia , Humans , Male , Adult , Hyperlipoproteinemia Type I/diagnosis , Hyperlipoproteinemia Type I/genetics , Hyperlipoproteinemia Type I/metabolism , Lipoprotein Lipase/genetics , Ecuador , Hypertriglyceridemia/etiology
9.
Nutrients ; 14(14)2022 Jul 19.
Article in English | MEDLINE | ID: mdl-35889913

ABSTRACT

Excessive alcohol consumption, as part of an unhealthy lifestyle, can contribute to metabolic abnormalities. This study investigated the sex differences in the relationship between excessive drinking and the risk of metabolic abnormalities. This community-based study included 3387 participants (age range: 30-103 years, mean age ± SD: 57 ± 13.5 years, 38.2% males) from the northeastern region of Taiwan. All participants completed a demographic survey and were subjected to blood tests. The risks of excessive drinking were evaluated using the Alcohol Use Disorder Identification Test (AUDIT). The results showed that males were at higher risks of obesity, hypertension, and hypertriglyceridemia, but at a lower risk of abdominal obesity than females. Males with hazardous drinking were at greater risks of hypertension, hyperglycemia, low serum levels of high-density lipoprotein cholesterol, and hypertriglyceridemia compared to those with no drinking. Females with hazardous drinking were at a greater risk of hypertension than those with no drinking. There was no interaction effect of sex and excessive drinking on the risks of metabolic abnormalities after controlling for demographics and lifestyle-related habits. Future studies are warranted to explore the sex-specific risk factors for metabolic abnormalities and to elucidate the mechanism underlying this association between alcohol consumption and metabolic abnormalities.


Subject(s)
Hypertension , Hypertriglyceridemia , Metabolic Syndrome , Adult , Aged , Aged, 80 and over , Alcohol Drinking/adverse effects , Alcohol Drinking/epidemiology , Female , Humans , Hypertension/epidemiology , Hypertension/etiology , Hypertriglyceridemia/epidemiology , Hypertriglyceridemia/etiology , Male , Metabolic Syndrome/epidemiology , Metabolic Syndrome/etiology , Middle Aged , Obesity , Risk Factors , Sex Characteristics , Taiwan/epidemiology
10.
Mol Nutr Food Res ; 66(7): e2101115, 2022 04.
Article in English | MEDLINE | ID: mdl-35124887

ABSTRACT

SCOPE: The aim of this study is to delineate the contribution of dietary saturated fatty acids (FA) versus liquid fructose to fatty liver and hypertriglyceridemia. METHODS AND RESULTS: Three groups of female rats are maintained for 3 months in standard chow (CT); High-fat diet (46.9% of fat-derived calories, rich in palmitic and stearic FA, HFD); and HFD with 10% w/v fructose in drinking water (HFHFr). Zoometric parameters, plasma biochemistry, and liver Oil-Red O (ORO) staining, lipidomics, and expression of proteins involved in FA metabolism are analyzed. Both diets increase ingested calories without modifying body weight. Only the HFHFr diet increases liver triglycerides (x11.0), with hypertriglyceridemia (x1.7) and reduces FA ß-oxidation (x0.7), and increases liver FA markers of DNL (de novo lipogenesis). Whereas HFD livers show a high content of ceramides, HFHFr samples show unchanged ceramides, and an increase in diacylglycerols. Only the HFHFr diet leads to a marked increase in the expression of enzymes involved in DNL and triglyceride metabolism, such as carbohydrate response element binding protein ß (ChREBPß, x3.2), a transcription factor that regulates DNL, and patatin-like phospholipase domain-containing 3 (PNPLA3, x2.6), a lipase that mobilizes stored triglycerides for VLDL secretion. CONCLUSION: The addition of liquid-fructose to dietary FA is determinant in liver steatosis and hypertriglyceridemia production, through increased DNL and PNPLA3 expression, and reduced FA catabolism.


Subject(s)
Fatty Liver , Hypertriglyceridemia , Animals , Diet, High-Fat/adverse effects , Female , Fructose/adverse effects , Fructose/metabolism , Hypertriglyceridemia/etiology , Lipogenesis/physiology , Liver/metabolism , Rats , Transcription Factors/metabolism , Triglycerides
11.
Nutr Clin Pract ; 37(5): 1172-1179, 2022 Oct.
Article in English | MEDLINE | ID: mdl-34528297

ABSTRACT

BACKGROUND: Levocarnitine deficiency has been observed in patients receiving parenteral nutrition (PN) and can cause or worsen hypertriglyceridemia. The objective was to characterize use of levocarnitine supplementation in PN and evaluate its effect on triglyceride levels in hospitalized adults. METHODS: This retrospective, single-center study included patients with triglyceride levels ≥175 mg/dl while receiving PN who had a subsequent reduction in lipid injectable emulsion dose. A piecewise linear regression was used to evaluate trends in triglyceride levels before and after the intervention, defined as initiation of levocarnitine in PN for the levocarnitine group, or reduction in lipid injectable emulsion alone for the control group. RESULTS: Two hundred sixty-one patients who received PN had an elevated triglyceride level and lipid injectable emulsion dose reduction, of which 97 (37.2%) received levocarnitine in PN. The median (IQR) levocarnitine dose added to PN was 8.0 (5.7-9.9) mg/kg. Triglyceride levels at 30 days post-intervention did not differ between groups (125 vs 176 mg/dl, P = .345). The addition of levocarnitine to PN was associated with a significantly greater rate of reduction in triglyceride levels pre-intervention to post-intervention compared with a reduction in lipid injectable emulsion alone (-11 vs -3 mg/dl per day; 95% CI, -15 to -2; P = .012). CONCLUSION: In hospitalized adults with hypertriglyceridemia who had a lipid injectable emulsion dose reduction, the addition of levocarnitine in PN was not associated with a difference in triglyceride levels at 30 days; however, a greater rate of improvement in pre-intervention to post-intervention triglyceride levels was observed.


Subject(s)
Fat Emulsions, Intravenous , Hypertriglyceridemia , Carnitine/therapeutic use , Dietary Supplements , Fat Emulsions, Intravenous/therapeutic use , Humans , Hypertriglyceridemia/drug therapy , Hypertriglyceridemia/etiology , Parenteral Nutrition/adverse effects , Retrospective Studies , Triglycerides
12.
Hepatology ; 75(5): 1169-1180, 2022 05.
Article in English | MEDLINE | ID: mdl-34580885

ABSTRACT

BACKGROUND AND AIMS: Lipoprotein lipase (LPL) is responsible for the lipolytic processing of triglyceride-rich lipoproteins, the deficiency of which causes severe hypertriglyceridemia. Liver LPL expression is high in suckling rodents but relatively low at adulthood. However, the regulatory mechanism and functional significance of liver LPL expression are incompletely understood. We have established the zinc finger protein ZBTB20 as a critical factor for hepatic lipogenesis. Here, we evaluated the role of ZBTB20 in regulating liver Lpl gene transcription and plasma triglyceride metabolism. APPROACH AND RESULTS: Hepatocyte-specific inactivation of ZBTB20 in mice led to a remarkable increase in LPL expression at the mRNA and protein levels in adult liver, in which LPL protein was mainly localized onto sinusoidal epithelial cells and Kupffer cells. As a result, the LPL activity in postheparin plasma was substantially increased, and postprandial plasma triglyceride clearance was significantly enhanced, whereas plasma triglyceride levels were decreased. The dysregulated liver LPL expression and low plasma triglyceride levels in ZBTB20-deficient mice were normalized by inactivating hepatic LPL expression. ZBTB20 deficiency protected the mice against high-fat diet-induced hyperlipidemia without causing excessive triglyceride accumulation in the liver. Chromatin immunoprecipitation and gel-shift assay studies revealed that ZBTB20 binds to the LPL promoter in the liver. A luciferase reporter assay revealed that ZBTB20 inhibits the transcriptional activity of LPL promoter. The regulation of LPL expression by ZBTB20 is liver-specific under physiological conditions. CONCLUSIONS: Liver ZBTB20 serves as a key regulator of LPL expression and plasma triglyceride metabolism and could be a therapeutic target for hypertriglyceridemia.


Subject(s)
BTB-POZ Domain , Hypertriglyceridemia , Animals , Hepatocytes/metabolism , Hypertriglyceridemia/etiology , Hypertriglyceridemia/metabolism , Lipoprotein Lipase/genetics , Lipoprotein Lipase/metabolism , Liver/metabolism , Mice , Transcription Factors/metabolism , Transcription, Genetic , Triglycerides/metabolism , Zinc Fingers
13.
Lipids Health Dis ; 20(1): 177, 2021 Dec 12.
Article in English | MEDLINE | ID: mdl-34895245

ABSTRACT

BACKGROUND: Hypertriglyceridemia can occur in lymphoproliferative disorders. Infectious mononucleosis is a self-limiting, benign lymphoproliferative disorder. This study aimed to investigate the serum triglyceride concentrations and their change over time in patients with infectious mononucleosis. METHODS: We evaluated an adult patient with severe hypertriglyceridemia (>1000 mg/dL) during infectious mononucleosis and reviewed the records of 360 patients admitted to our hospital because of infectious mononucleosis (median age, 19 years; range, 15-87 years; 51.4% male). We compared the serum triglyceride concentrations with those of a control sample from the general population (n=75). A second triglyceride measurement, obtained during convalescence (median of 30 days after the initial determination), was available for 160 patients. RESULTS: The triglyceride concentrations in the acute phase (median: 156 mg/dL) were significantly higher than those of the controls (median, 76 mg/dL; P<0.001). A total of 194 (53.9%) patients presented with hypertriglyceridemia (>150 mg/dL), which was more common in the patients older than 30 years than in the younger patients (78.6% vs. 50.6%; P<0.001). A significant correlation (P<0.005) was observed between the triglyceride levels and white blood cell counts, total cholesterol levels, and liver damage markers. The triglyceride concentrations decreased during convalescence (P<0.001) and were lower than the initial measurement in 83.7% of the cases. Conversely, the total cholesterol concentrations during the acute phase were lower than those of the controls and increased during convalescence (P<0.001). CONCLUSIONS: Patients with severe infectious mononucleosis frequently show mild, transient hypertriglyceridemia. Further studies are needed to elucidate the mechanisms underlying this finding.


Subject(s)
Herpesvirus 4, Human , Hypertriglyceridemia/etiology , Infectious Mononucleosis/complications , Adolescent , Adult , Aged , Aged, 80 and over , Case-Control Studies , Cholesterol/blood , Female , Humans , Hypertriglyceridemia/virology , Infectious Mononucleosis/blood , Infectious Mononucleosis/metabolism , Male , Middle Aged , Time Factors , Triglycerides/blood , Young Adult
14.
Presse Med ; 50(3): 104085, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34728268

ABSTRACT

Lipodystrophy syndromes are rare complex multisystem disorders caused by generalized or partial lack of adipose tissue. Adipose tissue dysfunction in lipodystrophy is associated with leptin deficiency. Lipodystrophy leads to severe metabolic problems. These abnormalities include, but are not limited to, insulin-resistant diabetes, severe hypertriglyceridemia, and lipid accumulation in ectopic organs such as the liver, and are associated with end-organ complications. Metabolic abnormalities can be present at the time of diagnosis or may develop over time as the disease progresses. In addition to metabolic abnormalities, subtype-specific presentations due to underlying molecular etiology in genetic forms and autoimmunity in acquired forms contribute to severe morbidity in lipodystrophy.


Subject(s)
Leptin/deficiency , Lipodystrophy/complications , Autoimmune Diseases/etiology , Diabetes Mellitus/etiology , Heart Diseases/genetics , Humans , Hypertriglyceridemia/etiology , Insulin Resistance , Kidney Diseases/complications , Lipid Metabolism , Lipodystrophy/genetics , Lipodystrophy/metabolism , Lipodystrophy/mortality , Liver/metabolism , Metabolic Syndrome/etiology , Neuromuscular Diseases/etiology , Non-alcoholic Fatty Liver Disease/etiology , Pancreatitis/etiology , Syndrome
15.
PLoS One ; 16(11): e0260495, 2021.
Article in English | MEDLINE | ID: mdl-34847179

ABSTRACT

OBJECTIVE: To assess the response of serum triglycerides (TG) to continuous insulin infusion (CII) in adults with hypertriglyceridemia-associated acute pancreatitis (HTGP). METHODS: Retrospective analysis of TG response to standardized CII therapy in 77 adults admitted to intensive care with TG >1000 mg/dL and HTGP. RESULTS: Participants had initial TG 3869.0 [2713.5, 5443.5] mg/dL and were 39.3 ± 9.7 years old, 66.2% males, 58.4% Hispanic, BMI 30.2 [27.0, 34.8] kg/m2, 74.0% with diabetes mellitus (DM) and 50.6% with excess alcohol use. TG-goal, defined as ≤1,000 ± 100 mg/dL, was achieved in 95%. Among the 73 TG-goal achievers (responders), 53.4% reached TG-goal in <36 hours after CII initiation (rapid responders). When compared to slow responders taking≥36 hours, rapid responders had lower initial TG (2862.0 [1965.0, 4519.0] vs 4814.5 [3368.8, 6900.0] mg/dL), BMI (29.4 [25.9, 32.8] vs 31.9 [28.2, 38.3] kg/m2), DM prevalence (56.4 vs 94.1%), and reached TG-50% (half of respective initial TG) faster (12.0 [6.0, 17.0] vs 18.5 [13.0, 32.8] hours). Those with DM (n = 57) vs non-DM (n = 20) were obese (31.4 [28.0, 35.6] vs 27.8 [23.6, 30.3] kg/m2), took longer to reach TG-final (41.0 [25.0, 60.5] vs 14.5 [12.5, 25.5] hours) and used more daily insulin (1.7 [1.3, 2.1] vs 1.1 [0.5, 1.9] U/kg/day). Among those with DM, the rapid responders had higher daily use of insulin vs slow responders 1.9 [1.4, 2.3] vs 1.6 [1.1, 1.8] U/kg/day. All results significant. In multivariable analysis, predictors of faster TG response were absence of DM, lower BMI and initial TG. CONCLUSION: CII was effective in reaching TG-goal in 95% of patients with HTGP. Half achieved TG-goal within 36 hours. Presence of DM, higher BMI and initial TG slowed the time to reach TG-goal. These baseline parameters and rate of decline to TG-50% may be real-time indicators to initiate and adjust the CII for quicker response.


Subject(s)
Hypertriglyceridemia , Insulin/administration & dosage , Pancreatitis , Triglycerides/blood , Adult , Female , Humans , Hypertriglyceridemia/blood , Hypertriglyceridemia/drug therapy , Hypertriglyceridemia/etiology , Male , Middle Aged , Pancreatitis/blood , Pancreatitis/complications , Pancreatitis/drug therapy , Retrospective Studies
16.
FASEB J ; 35(12): e22046, 2021 12.
Article in English | MEDLINE | ID: mdl-34800307

ABSTRACT

Hexosaminidase A (HexA), a heterodimer consisting of HEXA and HEXB, converts the ganglioside sphingolipid GM2 to GM3 by removing a terminal N-acetyl-d-galactosamine. HexA enzyme deficiency in humans leads to GM2 accumulation in cells, particularly in neurons, and is associated with neurodegeneration. While HexA and sphingolipid metabolism have been extensively investigated in the context of neuronal lipid metabolism, little is known about the metabolic impact of HexA and ganglioside degradation in other tissues. Here, we focussed on the role of HexA in the liver, which is a major regulator of systemic lipid metabolism. We find that hepatic Hexa expression is induced by lipid availability and increased in the presence of hepatic steatosis, which is associated with increased hepatic GM3 content. To assess the impact of HEXA on hepatic lipid metabolism, we used an adeno-associated virus to overexpress HEXA in the livers of high-fat diet fed mice. HEXA overexpression was associated with increased hepatic GM3 content and increased expression of enzymes involved in the degradation of glycated sphingolipids, ultimately driving sphingomyelin accumulation in the liver. In addition, HEXA overexpression led to substantial proteome remodeling in cell surface lipid rafts, which was associated with increased VLDL processing and secretion, hypertriglyceridemia and ectopic lipid accumulation in peripheral tissues. This study established an important role of HEXA in modulating hepatic sphingolipid and lipoprotein metabolism.


Subject(s)
Fatty Liver/pathology , Hexosaminidase A/metabolism , Hypertriglyceridemia/pathology , Lipids/analysis , Lipoproteins, VLDL/metabolism , Membrane Microdomains/pathology , Sphingolipids/metabolism , Animals , Fatty Liver/etiology , Fatty Liver/metabolism , Hexosaminidase A/genetics , Hypertriglyceridemia/etiology , Hypertriglyceridemia/metabolism , Membrane Microdomains/metabolism , Mice , Mice, Inbred C57BL
17.
Can J Physiol Pharmacol ; 99(11): 1137-1147, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34582252

ABSTRACT

Type-2 diabetes (T2D) is associated with liver toxicity. L-ergothioneine (L-egt) has been reported to reduce toxicity in tissues exposed to injury, while metformin is commonly prescribed to manage T2D. Hence, this study evaluates the hepatoprotective role of L-egt, with or without metformin, in T2D male rats. A total of 36 adult male Sprague-Dawley rats were randomly divided into non-diabetic (n = 12) and diabetic (n = 24) groups. After induction of diabetes, animals were divided into six groups (n = 6) and treated orally either with deionized water, L-egt (35 mg/kg bodyweight (bwt)), metformin (500 mg/kg bwt), or a combination of L-egt and metformin for 7 weeks. Body weight and blood glucose were monitored during the experiment. Thereafter, animals were euthanized and liver tissue was excised for biochemical, ELISA, real-time quantitative PCR, and histopathological analysis. L-egt with or without metformin reduced liver hypertrophy, liver injury, triglycerides, oxidative stress, and inflammation. Also, L-egt normalized mRNA expression of SREBP-1c, fatty acid synthase, nuclear factor kappa B, transforming growth factor ß1, nuclear factor erythroid 2-related factor 2, and sirtuin-1 in diabetic rats. Furthermore, co-administration of L-egt with metformin to diabetic rats reduced blood glucose and insulin resistance. These results provide support to the therapeutic benefits of L-egt in the management of liver complications associated with T2D.


Subject(s)
Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/drug therapy , Ergothioneine/therapeutic use , Hypertriglyceridemia/drug therapy , Liver Diseases/drug therapy , Liver Diseases/etiology , Metformin/therapeutic use , Oxidative Stress/drug effects , Administration, Oral , Animals , Blood Glucose/metabolism , Drug Therapy, Combination , Ergothioneine/administration & dosage , Ergothioneine/pharmacology , Hypertriglyceridemia/etiology , Inflammation , Insulin Resistance , Male , Metformin/administration & dosage , Metformin/pharmacology , Rats, Sprague-Dawley
18.
Nutrients ; 13(9)2021 Aug 28.
Article in English | MEDLINE | ID: mdl-34578886

ABSTRACT

Dietary choices may have differing effects on low-density lipoprotein cholesterol or triglyceride levels. The aim of this study was to investigate daily nutrient intake and dietary patterns of individuals with hyper-low-density lipoprotein cholesterolemia (hLDL) and hypertriglyceridemia (hTG) in a large Korean population-based study using propensity score (PS) matching. This study used data from the Korea National Health and Nutrition Examination Survey. Propensity score values for the predicted probability of patients with hLDL or hTG were estimated using logistic regression analysis, with age, sex, body mass index, alcohol consumption, smoking status, physical activity status, hypertension, and diabetes. After PS matching, intake of carbohydrates (%) was significantly lower (p = 0.021), and intake of fats (%) and saturated fatty acids (%) was significantly higher in the hLDL group than in the non-hLDL group (p = 0.025 and p = 0.013, respectively). The percentage of individuals with a high score for the Korean Healthy Eating Index (KHEI) "whole grains" or "saturated fatty acids" components was higher in the non-hLDL group than in the hLDL group (p < 0.05 for both). Dietary sodium/potassium ratio was significantly higher in the hTG than in the non-hTG (p = 0.049). Our results suggest that individualized dietary information and counseling require consideration of a person's specific lipid levels.


Subject(s)
Cholesterol, LDL/blood , Diet , Feeding Behavior , Hypercholesterolemia/etiology , Hypertriglyceridemia/blood , Nutrients , Triglycerides/blood , Adult , Aged , Counseling , Diet, Healthy , Dietary Carbohydrates/administration & dosage , Dietary Fats/administration & dosage , Dietary Fats/adverse effects , Dietary Fats/blood , Eating , Fatty Acids/administration & dosage , Fatty Acids/adverse effects , Fatty Acids/blood , Female , Humans , Hypercholesterolemia/blood , Hypertriglyceridemia/etiology , Hypertriglyceridemia/prevention & control , Male , Middle Aged , Nutrients/administration & dosage , Nutrients/adverse effects , Nutrition Surveys , Odds Ratio , Republic of Korea , Sodium, Dietary/administration & dosage , Sodium, Dietary/adverse effects , Whole Grains
19.
Mol Med ; 27(1): 101, 2021 09 06.
Article in English | MEDLINE | ID: mdl-34488621

ABSTRACT

BACKGROUND: Diet-induced obesity can result in the development of a diverse spectrum of cardiovascular and metabolic diseases, including type 2 diabetes, dyslipidemia, non-alcoholic liver steatosis and atherosclerotic disease. MicroRNAs have been described to be important regulators of metabolism and disease development. METHODS: In the current study, we investigated the effects of ubiquitous miR-100 overexpression on weight gain and the metabolic phenotype in a newly generated transgenic mouse strain under normal chow and high fat diet and used microarray expression analysis to identify new potential target genes of miR-100. RESULTS: While transgenic overexpression of miR-100 did not significantly affect weight and metabolism under a normal diet, miR-100 overexpressing mice showed a reduced weight gain under a high fat diet compared to wildtype mice, despite an equal calorie intake. This was accompanied by less visceral and subcutaneous fat development and lover serum LDL cholesterol. In addition, transgenic miR-100 mice were more glucose tolerant and insulin sensitive and demonstrated increased energy expenditure under high fat diet feeding. A comprehensive gene expression profiling revealed the differential expression of several genes involved in lipid storage- and metabolism, among them CD36 and Cyp4A14. Our data showed a direct regulation of CD36 by miR-100, leading to a reduced fatty acid uptake in primary hepatocytes overexpressing miR-100 and the downregulation of several downstream mediators of lipid metabolism such as ACC1, FABP4, FAS and PPARγ in the liver. CONCLUSIONS: Our findings demonstrate a protective role of miR-100 in high fat diet induced metabolic syndrome and liver steatosis, partially mediated by the direct repression of CD36 and attenuation of hepatic lipid storage, implicating miR-100 as a possible therapeutic target in liver steatosis.


Subject(s)
Hypertriglyceridemia/etiology , Hypertriglyceridemia/metabolism , Metabolic Syndrome/etiology , Metabolic Syndrome/metabolism , MicroRNAs/genetics , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/metabolism , 3' Untranslated Regions , Animals , Biomarkers , Cells, Cultured , Diet, High-Fat/adverse effects , Disease Models, Animal , Disease Susceptibility , Gene Expression Regulation , Glucose/metabolism , Hepatocytes/metabolism , Insulin/metabolism , Lipid Metabolism , Male , Mice , Mice, Transgenic , Phenotype , RNA Interference , Transcriptome , Weight Gain
20.
Eur Rev Med Pharmacol Sci ; 25(17): 5525-5528, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34533798

ABSTRACT

The Pfizer-BioNTech coronavirus disease 2019 (COVID-19) vaccine is the first novel nucleoside-modified messenger ribonucleic acid (modRNA) vaccine to receive Emergency Use Authorization from the Food and Drug Administration in the United States. It is indicated to be used in patients ≥12 years-of-age as of May 25th, 2021, including populations with high atherosclerotic cardiovascular disease (ASCVD) burden. However, little is known about the potential impact this vaccine may have on serum lipoprotein levels in patients with familial hypercholesteremia (FH), who are predisposed to high ASCVD burden due to elevated low-density lipoprotein cholesterol (LDL-C). We present an interesting case where a patient with heterozygous FH (HeFH) and elevated triglycerides (TG)-controlled for years on medication and apheresis-experienced significantly elevated TG, one day after receiving his second Pfizer-BioNTech COVID-19 vaccine dose. It is not known whether this adverse event may be seen in other FH patients and may be worth assessing in such patients to determine the possibility of a rare adverse reaction from a COVID-19 vaccine.


Subject(s)
COVID-19 Vaccines/adverse effects , Hyperlipoproteinemia Type II/blood , Hypertriglyceridemia/etiology , COVID-19/prevention & control , Cholesterol/blood , Humans , Hypertriglyceridemia/blood , Male , Middle Aged , SARS-CoV-2/immunology , Triglycerides/blood , Vaccination
SELECTION OF CITATIONS
SEARCH DETAIL