Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.266
Filter
1.
Rev Assoc Med Bras (1992) ; 70(8): e20240263, 2024.
Article in English | MEDLINE | ID: mdl-39166676

ABSTRACT

OBJECTIVE: The aim of the study was to examine the expression profile of genes (APOE, FTO, and LPL) associated with metabolic syndrome (MetS) in subjects with concomitant atrial fibrillation (AF). METHODS: A total of 690 subjects were categorized into control, AF without MetS, and AF with MetS. RESULTS: The expression profiles of the APOE, FTO, and LPL genes were decreased in AF subjects and AF subjects with MetS as compared to the controls. In AF without the MetS group, an inverse relationship was found between the expression of the LPL gene with body mass index (BMI) and a positive relationship with creatine kinase-MB, whereas expression of the FTO gene was inversely associated with fasting blood glucose and positively with cardiac troponin I in AF suffering from MetS. Expression of the LPL gene was directly linked with systolic blood pressure (SBP) and high-density lipoprotein-cholesterol (HDL-C), whereas an inverse correlation with heart rate and expression of the FTO gene in AF with MetS were shown. The expression of the LPL gene was inversely related to BMI in subjects with AF. The expression of the LPL gene was positively correlated with SBP and HDL-C and negatively correlated with heart rate, while the expression of the FTO gene was an important predictor of AF with MetS. CONCLUSION: The decreased expression of APOE, FTO, and LPL genes in AF with and without MetS indicates their potential contributing role in the pathogenesis of AF.


Subject(s)
Alpha-Ketoglutarate-Dependent Dioxygenase FTO , Apolipoproteins E , Atrial Fibrillation , Body Mass Index , Lipoprotein Lipase , Metabolic Syndrome , Humans , Alpha-Ketoglutarate-Dependent Dioxygenase FTO/genetics , Atrial Fibrillation/genetics , Male , Female , Case-Control Studies , Apolipoproteins E/genetics , Metabolic Syndrome/genetics , Middle Aged , Lipoprotein Lipase/genetics , Aged , Cholesterol, HDL/blood , Blood Pressure/genetics , Blood Glucose/analysis
3.
Lifestyle Genom ; 17(1): 82-92, 2024.
Article in English | MEDLINE | ID: mdl-38952113

ABSTRACT

INTRODUCTION: This study aims to investigate if a mixture of functional lipids (FLs), containing conjugated linoleic acid (CLA), tocopherols (TPs), and phytosterols (PSs), prevents some lipid alterations induced by high-fat (HF) diets, without adverse effects. METHODS: Male CF1 mice (n = 6/group) were fed (4 weeks) with control (C), HF, or HF + FL diets. RESULTS: FL prevented the overweight induced by the HF diet and reduced the adipose tissue (AT) weight, associated with lower energy efficiency. After the intervention period, the serum triacylglycerol (TAG) levels in both HF diets underwent a decrease associated with an enhanced LPL activity (mainly in muscle). The beneficial effect of the FL mixture on body weight gain and AT weight might be attributed to the decreased lipogenesis, denoted by the lower mRNA levels of SREBP1-c and ACC in AT, as well as by an exacerbated lipid catabolism, reflected by increased mRNA levels of PPARα, ATGL, HSL, and UCP2 in AT. Liver TAG levels were reduced in the HF + FL group due to an elevated lipid oxidation associated with a higher CPT-1 activity and mRNA levels of PPARα and CPT-1a. Moreover, genes linked to fatty acid biosynthesis (SREBP1-c and ACC) showed decreased mRNA levels in both HF diets, this finding being more pronounced in the HF + FL group. CONCLUSION: The administration of an FL mixture (CLA + TP + PS) prevented some lipid alterations induced by a HF diet, avoiding frequent deleterious effects of CLA in mice through the modulation of gene expression related to the regulation of lipid metabolism.


Subject(s)
Diet, High-Fat , Linoleic Acids, Conjugated , Lipid Metabolism , Liver , PPAR alpha , Sterol Regulatory Element Binding Protein 1 , Triglycerides , Animals , Diet, High-Fat/adverse effects , Mice , Male , Triglycerides/metabolism , Liver/metabolism , Liver/drug effects , Lipid Metabolism/drug effects , PPAR alpha/metabolism , PPAR alpha/genetics , Sterol Regulatory Element Binding Protein 1/metabolism , Sterol Regulatory Element Binding Protein 1/genetics , Linoleic Acids, Conjugated/pharmacology , Lipogenesis/drug effects , Carnitine O-Palmitoyltransferase/metabolism , Carnitine O-Palmitoyltransferase/genetics , Uncoupling Protein 2/metabolism , Uncoupling Protein 2/genetics , Phytosterols/pharmacology , Adipose Tissue/metabolism , Adipose Tissue/drug effects , Weight Gain/drug effects , Lipoprotein Lipase/metabolism , Lipoprotein Lipase/genetics
4.
BMC Vet Res ; 20(1): 286, 2024 Jul 03.
Article in English | MEDLINE | ID: mdl-38961471

ABSTRACT

BACKGROUND: The milk's nutritional value is determined by its constituents, including fat, protein, carbohydrates, and minerals. The mammary gland's ability to produce milk is controlled by a complex network of genes. Thereby, the fat, protein, and lactose synthesis must be boost in milk to increase milk production efficiency. This can be accomplished by fusing genetic advancements with proper management practices. Therefore, this study aimed to investigate the association between the Lipoprotein lipase (LPL), kappa casein CSN3, and Glucose transporter 1 (GLUT1) genes expression levels and such milk components as fat, protein, and lactose in different dairy breeds during different stages of lactation. METHODS: To achieve such a purpose, 94 milk samples were collected (72 samples from 36 multiparous black-white and red-white Holstein-Friesian (HF) cows and 22 milk samples from 11 Egyptian buffaloes) during the early and peak lactation stages. The milk samples were utilized for milk analysis and genes expressions analyses using non- invasive approach in obtaining milk fat globules (MFGs) as a source of Ribonucleic acid (RNA). RESULTS: LPL and CSN3 genes expressions levels were found to be significantly higher in Egyptian buffalo than Holstein-Friesian (HF) cows as well as fat and protein percentages. On the other hand, GLUT1 gene expression level was shown to be significantly higher during peak lactation than early lactation. Moreover, lactose % showed a significant difference in peak lactation phase compared to early lactation phase. Also, fat and protein percentages were significantly higher in early lactation period than peak lactation period but lactose% showed the opposite pattern of Egyptian buffalo. CONCLUSION: Total RNA can be successfully obtained from MFGs. The results suggest that these genes play a role in glucose absorption and lactose synthesis in bovine mammary epithelial cells during lactation. Also, these results provide light on the differential expression of these genes among distinct Holstein-Friesian cow breeds and Egyptian buffalo subspecies throughout various lactation phases.


Subject(s)
Caseins , Glycolipids , Glycoproteins , Lactation , Lipid Droplets , Mammary Glands, Animal , Milk , RNA, Messenger , Animals , Cattle/genetics , Lactation/genetics , Female , Lipid Droplets/metabolism , Milk/chemistry , Milk/metabolism , Glycolipids/metabolism , Caseins/genetics , Caseins/metabolism , Glycoproteins/genetics , Glycoproteins/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Mammary Glands, Animal/metabolism , Lipoprotein Lipase/genetics , Lipoprotein Lipase/metabolism , Glucose Transporter Type 1/genetics , Glucose Transporter Type 1/metabolism , Buffaloes/genetics , Buffaloes/metabolism , Lactose/metabolism , Lactose/analysis , Milk Proteins/analysis , Milk Proteins/metabolism , Milk Proteins/genetics , Gene Expression Regulation
5.
J Transl Med ; 22(1): 615, 2024 Jul 03.
Article in English | MEDLINE | ID: mdl-38961500

ABSTRACT

OBJECTIVE: To explore the correlation between asthma risk and genetic variants affecting the expression or function of lipid-lowering drug targets. METHODS: We conducted Mendelian randomization (MR) analyses using variants in several genes associated with lipid-lowering medication targets: HMGCR (statin target), PCSK9 (alirocumab target), NPC1L1 (ezetimibe target), APOB (mipomersen target), ANGPTL3 (evinacumab target), PPARA (fenofibrate target), and APOC3 (volanesorsen target), as well as LDLR and LPL. Our objective was to investigate the relationship between lipid-lowering drugs and asthma through MR. Finally, we assessed the efficacy and stability of the MR analysis using the MR Egger and inverse variance weighted (IVW) methods. RESULTS: The elevated triglyceride (TG) levels associated with the APOC3, and LPL targets were found to increase asthma risk. Conversely, higher LDL-C levels driven by LDLR were found to decrease asthma risk. Additionally, LDL-C levels (driven by APOB, NPC1L1 and HMGCR targets) and TG levels (driven by the LPL target) were associated with improved lung function (FEV1/FVC). LDL-C levels driven by PCSK9 were associated with decreased lung function (FEV1/FVC). CONCLUSION: In conclusion, our findings suggest a likely causal relationship between asthma and lipid-lowering drugs. Moreover, there is compelling evidence indicating that lipid-lowering therapies could play a crucial role in the future management of asthma.


Subject(s)
Asthma , Hypolipidemic Agents , Mendelian Randomization Analysis , Humans , Asthma/genetics , Asthma/drug therapy , Hypolipidemic Agents/therapeutic use , Hypolipidemic Agents/pharmacology , Proprotein Convertase 9/genetics , Genetic Association Studies , Lung/drug effects , Lung/pathology , Lipoprotein Lipase/genetics , Triglycerides/blood , Receptors, LDL/genetics , Hydroxymethylglutaryl CoA Reductases/genetics , Angiopoietin-Like Protein 3 , Angiopoietin-like Proteins/genetics , Apolipoprotein C-III/genetics , Apolipoproteins B/genetics , Respiratory Function Tests , Cholesterol, LDL/blood , Membrane Transport Proteins , PPAR alpha
6.
Subcell Biochem ; 104: 139-179, 2024.
Article in English | MEDLINE | ID: mdl-38963487

ABSTRACT

Lipoprotein lipase (LPL) is a critical enzyme in humans that provides fuel to peripheral tissues. LPL hydrolyzes triglycerides from the cores of lipoproteins that are circulating in plasma and interacts with receptors to mediate lipoprotein uptake, thus directing lipid distribution via catalytic and non-catalytic functions. Functional losses in LPL or any of its myriad of regulators alter lipid homeostasis and potentially affect the risk of developing cardiovascular disease-either increasing or decreasing the risk depending on the mutated protein. The extensive LPL regulatory network tunes LPL activity to allocate fatty acids according to the energetic needs of the organism and thus is nutritionally responsive and tissue dependent. Multiple pharmaceuticals in development manipulate or mimic these regulators, demonstrating their translational importance. Another facet of LPL biology is that the oligomeric state of the enzyme is also central to its regulation. Recent structural studies have solidified the idea that LPL is regulated not only by interactions with other binding partners but also by self-associations. Here, we review the complexities of the protein-protein and protein-lipid interactions that govern LPL structure and function.


Subject(s)
Lipoprotein Lipase , Lipoprotein Lipase/metabolism , Lipoprotein Lipase/chemistry , Lipoprotein Lipase/genetics , Humans , Animals , Protein Binding , Triglycerides/metabolism , Lipid Metabolism
7.
JCI Insight ; 9(13)2024 Jul 08.
Article in English | MEDLINE | ID: mdl-38973609

ABSTRACT

Lipoprotein lipase (LPL) hydrolyzes circulating triglycerides (TGs), releasing fatty acids (FA) and promoting lipid storage in white adipose tissue (WAT). However, the mechanisms regulating adipose LPL and its relationship with the development of hypertriglyceridemia are largely unknown. WAT from obese humans exhibited high PAR2 expression, which was inversely correlated with the LPL gene. Decreased LPL expression was also inversely correlated with elevated plasma TG levels, suggesting that adipose PAR2 might regulate hypertriglyceridemia by downregulating LPL. In mice, aging and high palmitic acid diet (PD) increased PAR2 expression in WAT, which was associated with a high level of macrophage migration inhibitory factor (MIF). MIF downregulated LPL expression and activity in adipocytes by binding with CXCR2/4 receptors and inhibiting Akt phosphorylation. In a MIF overexpression model, high-circulating MIF levels suppressed adipose LPL, and this suppression was associated with increased plasma TGs but not FA. Following PD feeding, adipose LPL expression and activity were significantly reduced, and this reduction was reversed in Par2-/- mice. Recombinant MIF infusion restored high plasma MIF levels in Par2-/- mice, and the levels decreased LPL and attenuated adipocyte lipid storage, leading to hypertriglyceridemia. These data collectively suggest that downregulation of adipose LPL by PAR2/MIF may contribute to the development of hypertriglyceridemia.


Subject(s)
Down-Regulation , Hypertriglyceridemia , Lipoprotein Lipase , Receptor, PAR-2 , Animals , Lipoprotein Lipase/metabolism , Lipoprotein Lipase/genetics , Hypertriglyceridemia/metabolism , Hypertriglyceridemia/genetics , Mice , Humans , Receptor, PAR-2/metabolism , Receptor, PAR-2/genetics , Male , Mice, Knockout , Triglycerides/metabolism , Triglycerides/blood , Adipose Tissue, White/metabolism , Macrophage Migration-Inhibitory Factors/metabolism , Macrophage Migration-Inhibitory Factors/genetics , Adipocytes/metabolism , Obesity/metabolism , Obesity/genetics , Palmitic Acid/metabolism , Female , Mice, Inbred C57BL , Middle Aged
8.
J Lipid Res ; 65(7): 100578, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38880127

ABSTRACT

Apolipoprotein AV (APOA5) deficiency causes hypertriglyceridemia in mice and humans. For years, the cause remained a mystery, but the mechanisms have now come into focus. Here, we review progress in defining APOA5's function in plasma triglyceride metabolism. Biochemical studies revealed that APOA5 binds to the angiopoietin-like protein 3/8 complex (ANGPTL3/8) and suppresses its ability to inhibit the activity of lipoprotein lipase (LPL). Thus, APOA5 deficiency is accompanied by increased ANGPTL3/8 activity and lower levels of LPL activity. APOA5 deficiency also reduces amounts of LPL in capillaries of oxidative tissues (e.g., heart, brown adipose tissue). Cell culture experiments revealed the likely explanation: ANGPTL3/8 detaches LPL from its binding sites on the surface of cells, and that effect is blocked by APOA5. Both the low intracapillary LPL levels and the high plasma triglyceride levels in Apoa5-/- mice are normalized by recombinant APOA5. Carboxyl-terminal sequences in APOA5 are crucial for its function; a mutant APOA5 lacking 40-carboxyl-terminal residues cannot bind to ANGPTL3/8 and lacks the ability to change intracapillary LPL levels or plasma triglyceride levels in Apoa5-/- mice. Also, an antibody against the last 26 amino acids of APOA5 reduces intracapillary LPL levels and increases plasma triglyceride levels in wild-type mice. An inhibitory ANGPTL3/8-specific antibody functions as an APOA5-mimetic reagent, increasing intracapillary LPL levels and lowering plasma triglyceride levels in both Apoa5-/- and wild-type mice. That antibody is a potentially attractive strategy for treating elevated plasma lipid levels in human patients.


Subject(s)
Apolipoprotein A-V , Hypertriglyceridemia , Lipoprotein Lipase , Animals , Lipoprotein Lipase/metabolism , Lipoprotein Lipase/genetics , Humans , Hypertriglyceridemia/metabolism , Hypertriglyceridemia/genetics , Apolipoprotein A-V/genetics , Apolipoprotein A-V/metabolism , Capillaries/metabolism , Mice , Triglycerides/metabolism , Triglycerides/blood
9.
Expert Rev Endocrinol Metab ; 19(4): 299-306, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38866702

ABSTRACT

INTRODUCTION: Familial chylomicronemia syndrome (FCS) is a rare autosomal recessive condition. Effective treatment is important as patients are at risk for severe and potentially fatal acute pancreatitis. We review recent developments in pharmacologic treatment for FCS, namely biological inhibitors of apolipoprotein (apo) C-III and angiopoietin-like protein 3 (ANGPTL3). AREAS COVERED: FCS follows a biallelic inheritance pattern in which an individual inherits two pathogenic loss-of-function alleles of one of the five causal genes - LPL (in 60-80% of patients), GPIHBP1, APOA5, APOC2, and LMF1 - leading to the absence of lipolytic activity. Patients present from childhood with severely elevated triglyceride (TG) levels >10 mmol/L. Most patients with severe hypertriglyceridemia do not have FCS. A strict low-fat diet is the current first-line treatment, and existing lipid-lowering therapies are minimally effective in FCS. Apo C-III inhibitors are emerging TG-lowering therapies shown to be efficacious and safe in clinical trials. ANGPTL3 inhibitors, another class of emerging TG-lowering therapies, have been found to require at least partial lipoprotein lipase activity to lower plasma TG in clinical trials. ANGPTL3 inhibitors reduce plasma TG in patients with multifactorial chylomicronemia but not in patients with FCS who completely lack lipoprotein lipase activity. EXPERT OPINION: Apo C-III inhibitors currently in development are promising treatments for FCS.


Subject(s)
Angiopoietin-Like Protein 3 , Hyperlipoproteinemia Type I , Humans , Hyperlipoproteinemia Type I/genetics , Hyperlipoproteinemia Type I/drug therapy , Hyperlipoproteinemia Type I/therapy , Apolipoprotein C-III/genetics , Apolipoprotein C-III/antagonists & inhibitors , Hypolipidemic Agents/therapeutic use , Lipoprotein Lipase/genetics , Angiopoietin-like Proteins/antagonists & inhibitors , Angiopoietin-like Proteins/genetics , Diet, Fat-Restricted , Receptors, Lipoprotein
10.
Cell Biochem Funct ; 42(5): e4069, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38940455

ABSTRACT

Stem cells demonstrate differentiation and regulatory functions. In this discussion, we will explore the impacts of cell culture density on stem cell proliferation, adipogenesis, and regulatory abilities. This study aimed to investigate the impact of the initial culture density of human periodontal ligament stem cells (hPDLSCs) on the adipogenic differentiation of autologous cells. Our findings indicate that the proliferation rate of hPDLSCs increased with increasing initial cell density (0.5-8 × 104 cells/cm2). After adipogenic differentiation induced by different initial cell densities of hPDLSC, we found that the mean adipose concentration and the expression levels of lipoprotein lipase (LPL), CCAAT/enhancer binding protein α (CEBPα), and peroxisome proliferator-activated receptor γ (PPAR-γ) genes all increased with increasing cell density. To investigate the regulatory role of hPDLSCs in the adipogenic differentiation of other cells, we used secreted exocrine vesicles derived from hPDLSCs cultivated at different initial cell densities of 50 µg/mL to induce the adipogenic differentiation of human bone marrow stromal cells. We also found that the mean adipose concentration and expression of LPL, CEBPα, and PPARγ genes increased with increasing cell density, with an optimal culture density of 8 × 104 cells/cm2. This study provides a foundation for the application of adipogenic differentiation in stem cells.


Subject(s)
Adipogenesis , Cell Differentiation , Periodontal Ligament , Stem Cells , Humans , Periodontal Ligament/cytology , Periodontal Ligament/metabolism , Stem Cells/cytology , Stem Cells/metabolism , PPAR gamma/metabolism , PPAR gamma/genetics , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/cytology , Cells, Cultured , Lipoprotein Lipase/metabolism , Lipoprotein Lipase/genetics , Cell Proliferation , Cell Count , CCAAT-Enhancer-Binding Protein-alpha/metabolism , CCAAT-Enhancer-Binding Protein-alpha/genetics
11.
Front Endocrinol (Lausanne) ; 15: 1387419, 2024.
Article in English | MEDLINE | ID: mdl-38911039

ABSTRACT

Introduction: Hypertriglyceridemia (HTG) is a complex disorder caused by genetic and environmental factors that frequently results from loss-of-function variants in the gene encoding lipoprotein lipase (LPL). Heterozygous patients have a range of symptoms, while homozygous LPL deficiency presents with severe symptoms including acute pancreatitis, xanthomas, and lipemia retinalis. Methods: We described the clinical characteristics of three Slovenian patients (an 8-year-old female, an 18-year-old man, and a 57-year-old female) and one Pakistani patient (a 59-year-old male) with LPL deficiency. We performed next-generation sequencing (NGS) targeting all coding exons and intron-exon boundaries of the LPL gene, and Sanger sequencing for variant confirmation. In addition, we performed a systematic literature review of all cases with three identified variants and described their clinical characteristics. Results: Two Slovenian patients with a heterozygous pathogenic variant NM_000237.3:c.984G>T (p.Met328Ile) were diagnosed within the first three years of life and had triglyceride (TG) values of 16 and 20 mmol/L. An asymptomatic Pakistani patient with TG values of 36.8 mmol/L until the age of 44 years, was identified as heterozygous for a pathogenic variant NM_000237.3:c.724G>A (p.Asp242Asn). His TG levels dropped to 12.7 mmol/L on dietary modifications and by using fibrates. A Slovenian patient who first suffered from pancreatitis at the age of 18 years with a TG value of 34 mmol/L was found to be homozygous for NM_000237.3:c.337T>C (p.Trp113Arg). Conclusions: Patients with LPL deficiency had high TG levels at diagnosis. Homozygous patients had worse outcomes. Good diet and medication compliance can reduce severity.


Subject(s)
Lipoprotein Lipase , Humans , Male , Female , Slovenia/epidemiology , Adolescent , Middle Aged , Lipoprotein Lipase/genetics , Lipoprotein Lipase/deficiency , Child , Pakistan/epidemiology , Hyperlipoproteinemia Type I/genetics , Mutation
12.
Arterioscler Thromb Vasc Biol ; 44(8): 1873-1883, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38899472

ABSTRACT

BACKGROUND: Pathogenic variants in PLIN1-encoding PLIN1 (perilipin-1) are responsible for an autosomal dominant form of familial partial lipodystrophy (FPL) associated with severe insulin resistance, hepatic steatosis, and important hypertriglyceridemia. This study aims to decipher the mechanisms of hypertriglyceridemia associated with PLIN1-related FPL. METHODS: We performed an in vivo lipoprotein kinetic study in 6 affected patients compared with 13 healthy controls and 8 patients with type 2 diabetes. Glucose and lipid parameters, including plasma LPL (lipoprotein lipase) mass, were measured. LPL mRNA and protein expression were evaluated in abdominal subcutaneous adipose tissue from patients with 5 PLIN1-mutated FPL and 3 controls. RESULTS: Patients with PLIN1-mutated FPL presented with decreased fat mass, insulin resistance, and diabetes (glycated hemoglobin A1c, 6.68±0.70% versus 7.48±1.63% in patients with type 2 diabetes; mean±SD; P=0.27). Their plasma triglycerides were higher (5.96±3.08 mmol/L) than in controls (0.76±0.27 mmol/L; P<0.0001) and patients with type 2 diabetes (2.94±1.46 mmol/L, P=0.006). Compared with controls, patients with PLIN1-related FPL had a significant reduction of the indirect fractional catabolic rate of VLDL (very-low-density lipoprotein)-apoB100 toward IDL (intermediate-density lipoprotein)/LDL (low-density lipoprotein; 1.79±1.38 versus 5.34±2.45 pool/d; P=0.003) and the indirect fractional catabolic rate of IDL-apoB100 toward LDL (2.14±1.44 versus 7.51±4.07 pool/d; P=0.005). VLDL-apoB100 production was not different between patients with PLIN1-related FPL and controls. Compared with patients with type 2 diabetes, patients with PLIN1-related FPL also showed a significant reduction of the catabolism of both VLDL-apoB100 (P=0.031) and IDL-apoB100 (P=0.031). Plasma LPL mass was significantly lower in patients with PLIN1-related FPL than in controls (21.03±10.08 versus 55.76±13.10 ng/mL; P<0.0001), although the LPL protein expression in adipose tissue was similar. VLDL-apoB100 and IDL-apoB100 indirect fractional catabolic rates were negatively correlated with plasma triglycerides and positively correlated with LPL mass. CONCLUSIONS: We show that hypertriglyceridemia associated with PLIN1-related FPL results from a marked decrease in the catabolism of triglyceride-rich lipoproteins (VLDL and IDL). This could be due to a pronounced reduction in LPL availability, related to the decreased adipose tissue mass.


Subject(s)
Diabetes Mellitus, Type 2 , Hypertriglyceridemia , Insulin Resistance , Lipodystrophy, Familial Partial , Lipoprotein Lipase , Lipoproteins , Perilipin-1 , Triglycerides , Humans , Male , Perilipin-1/genetics , Perilipin-1/metabolism , Perilipin-1/blood , Triglycerides/blood , Hypertriglyceridemia/blood , Hypertriglyceridemia/genetics , Female , Adult , Middle Aged , Case-Control Studies , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/complications , Lipoproteins/blood , Lipoprotein Lipase/blood , Lipoprotein Lipase/metabolism , Lipoprotein Lipase/genetics , Lipodystrophy, Familial Partial/genetics , Lipodystrophy, Familial Partial/blood , Lipodystrophy, Familial Partial/metabolism , Mutation , Blood Glucose/metabolism , Lipoproteins, VLDL/blood , Lipoproteins, VLDL/metabolism , Biomarkers/blood , Phenotype , Genetic Predisposition to Disease , Lipolysis , RNA, Messenger/metabolism , RNA, Messenger/genetics
14.
Nat Commun ; 15(1): 4410, 2024 May 23.
Article in English | MEDLINE | ID: mdl-38782979

ABSTRACT

Pancreatic ß cells secrete insulin in response to glucose elevation to maintain glucose homeostasis. A complex network of inter-organ communication operates to modulate insulin secretion and regulate glucose levels after a meal. Lipids obtained from diet or generated intracellularly are known to amplify glucose-stimulated insulin secretion, however, the underlying mechanisms are not completely understood. Here, we show that a Drosophila secretory lipase, Vaha (CG8093), is synthesized in the midgut and moves to the brain where it concentrates in the insulin-producing cells in a process requiring Lipid Transfer Particle, a lipoprotein originating in the fat body. In response to dietary fat, Vaha stimulates insulin-like peptide release (ILP), and Vaha deficiency results in reduced circulatory ILP and diabetic features including hyperglycemia and hyperlipidemia. Our findings suggest Vaha functions as a diacylglycerol lipase physiologically, by being a molecular link between dietary fat and lipid amplified insulin secretion in a gut-brain axis.


Subject(s)
Brain , Drosophila Proteins , Drosophila melanogaster , Insulin Secretion , Insulin , Animals , Drosophila Proteins/metabolism , Drosophila Proteins/genetics , Brain/metabolism , Insulin/metabolism , Insulin-Secreting Cells/metabolism , Brain-Gut Axis/physiology , Lipase/metabolism , Lipase/genetics , Dietary Fats/metabolism , Glucose/metabolism , Fat Body/metabolism , Lipoprotein Lipase/metabolism , Lipoprotein Lipase/genetics , Male
16.
Diabetes Obes Metab ; 26(7): 2969-2978, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38685616

ABSTRACT

AIM: The response rate to pioglitazone and the predictive factors for its effects on improving liver biochemistry in patients with steatotic liver disease (SLD) remain elusive, so we aimed to investigate these issues. METHODS: A 3-year prospective cohort study of 126 Taiwanese patients with SLD treated with pioglitazone (15-30 mg/day) was conducted. Phospholipase domain-containing protein 3 I148M rs738409, methylenetetrahydrofolate reductase rs1801133, aldehyde dehydrogenase 2 (ALDH2) rs671 and lipoprotein lipase rs10099160 single nucleotide polymorphisms were assessed in the patients. RESULTS: Of 126 patients, 78 (61.9%) were men, and the mean and median ages were 54.3 and 56.5 years, respectively. Pioglitazone responders were defined as those with decreased alanine aminotransferase (ALT) levels at 6 months post-treatment, and 105 (83.3%) patients were responders. Compared with non-responders, responders were more frequently women and had higher baseline ALT levels. The proportion of patients with the ALDH2 rs671 GG genotype was lower among responders (38.6% vs. 66.6%, p = .028). Female sex [odds ratio (OR): 4.514, p = .023] and baseline ALT level (OR: 1.015, p = .046; cut-off level: ≥82 U/L) were associated with pioglitazone response. Among responders, the liver biochemistry and homeostasis model assessment of insulin resistance improved from 6 to 24 months post-treatment. The total cholesterol levels decreased within 6 months, while increases in high-density lipoprotein cholesterol levels and decreases in triglyceride levels and fibrosis-4 scores were noted only at 24 months post-treatment. The 2-year cumulative incidences of cardiovascular events, cancers and hepatic events were similar between responders and non-responders. CONCLUSIONS: Regarding liver biochemistry, over 80% of Taiwanese patients with SLD had a pioglitazone response, which was positively associated with female sex and baseline ALT levels. Insulin resistance improved as early as 6 months post-treatment, while liver fibrosis improvement was not observed until 24 months post-treatment. The link between the pioglitazone response and the ALDH2 genotype warrants further investigation.


Subject(s)
Aldehyde Dehydrogenase, Mitochondrial , Hypoglycemic Agents , Pioglitazone , Polymorphism, Single Nucleotide , Humans , Pioglitazone/therapeutic use , Male , Female , Middle Aged , Prospective Studies , Hypoglycemic Agents/therapeutic use , Treatment Outcome , Aldehyde Dehydrogenase, Mitochondrial/genetics , Taiwan/epidemiology , Alanine Transaminase/blood , Thiazolidinediones/therapeutic use , Fatty Liver/drug therapy , Fatty Liver/genetics , Aged , Lipoprotein Lipase/genetics , Liver/drug effects , Liver/pathology , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/complications , Genotype , Adult
17.
Pharmacogenet Genomics ; 34(4): 91-104, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38682317

ABSTRACT

OBJECTIVES: This study explored the association of deleterious variants in pharmacodynamics (PD) genes with statin response and adverse effects in patients with familial hypercholesterolemia (FH) and analyzed their potential effects on protein structure and stability. METHODS: Clinical and laboratory data were obtained from 144 adult FH patients treated with statins. A panel of 32 PD genes was analyzed by exon-targeted gene sequencing. Deleterious variants were identified using prediction algorithms and their structural effects were analyzed by molecular modeling studies. RESULTS: A total of 102 variants were predicted as deleterious (83 missense, 8 stop-gain, 4 frameshift, 1 indel, 6 splicing). The variants ABCA1 rs769705621 (indel), LPA rs41267807 (p.Tyr2023Cys) and KIF6 rs20455 (p.Trp719Arg) were associated with reduced low-density lipoprotein cholesterol (LDLc) response to statins, and the LPL rs1801177 (p.Asp36Asn) with increased LDLc response (P < 0.05). LPA rs3124784 (p.Arg2016Cys) was predicted to increase statin response (P = 0.022), and ABCA1 rs769705621 to increase the risk of statin-related adverse events (SRAE) (P = 0.027). LPA p.Arg2016Cys and LPL p.Asn36Asp maintained interactions with solvent, LPA p.Tyr2023Cys reduced intramolecular interaction with Gln1987, and KIF6 p.Trp719Arg did not affect intramolecular interactions. DDMut analysis showed that LPA p.Arg2016Cys and p.Tyr2023Cys and LPL p.Asp36Asn caused energetically favorable changes, and KIF6 p.Trp719Arg resulted in unfavorable energetic changes, affecting protein stability. CONCLUSION: Deleterious variants in ABCA1, LPA, LPL and KIF6 are associated with variability in LDLc response to statins, and ABCA1 rs769705621 is associated with SRAE risk in FH patients. Molecular modeling studies suggest that LPA p.Tyr2023Cys and KIF6 p.Trp719Arg disturb protein conformational structure and stability.


Subject(s)
ATP Binding Cassette Transporter 1 , Hydroxymethylglutaryl-CoA Reductase Inhibitors , Hyperlipoproteinemia Type II , Kinesins , Lipoprotein Lipase , Humans , Kinesins/genetics , Male , Female , Middle Aged , Hydroxymethylglutaryl-CoA Reductase Inhibitors/adverse effects , Hyperlipoproteinemia Type II/genetics , Hyperlipoproteinemia Type II/drug therapy , ATP Binding Cassette Transporter 1/genetics , Lipoprotein Lipase/genetics , Adult , Protein Stability , Cholesterol, LDL/blood , Polymorphism, Single Nucleotide
18.
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
19.
Lipids Health Dis ; 23(1): 92, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38561841

ABSTRACT

BACKGROUND: Lipoprotein lipase (LPL) plays a crucial role in triglyceride hydrolysis. Rare biallelic variants in the LPL gene leading to complete or near-complete loss of function cause autosomal recessive familial chylomicronemia syndrome. However, rare biallelic LPL variants resulting in significant but partial loss of function are rarely documented. This study reports a novel occurrence of such rare biallelic LPL variants in a Chinese patient with hypertriglyceridemia-induced acute pancreatitis (HTG-AP) during pregnancy and provides an in-depth functional characterization. METHODS: The complete coding sequences and adjacent intronic regions of the LPL, APOC2, APOA5, LMF1, and GPIHBP1 genes were analyzed by Sanger sequencing. The aim was to identify rare variants, including nonsense, frameshift, missense, small in-frame deletions or insertions, and canonical splice site mutations. The functional impact of identified LPL missense variants on protein expression, secretion, and activity was assessed in HEK293T cells through single and co-transfection experiments, with and without heparin treatment. RESULTS: Two rare LPL missense variants were identified in the patient: the previously reported c.809G > A (p.Arg270His) and a novel c.331G > C (p.Val111Leu). Genetic testing confirmed these variants were inherited biallelically. Functional analysis showed that the p.Arg270His variant resulted in a near-complete loss of LPL function due to effects on protein synthesis/stability, secretion, and enzymatic activity. In contrast, the p.Val111Leu variant retained approximately 32.3% of wild-type activity, without impacting protein synthesis, stability, or secretion. Co-transfection experiments indicated a combined activity level of 20.7%, suggesting no dominant negative interaction between the variants. The patient's post-heparin plasma LPL activity was about 35% of control levels. CONCLUSIONS: This study presents a novel case of partial but significant loss-of-function biallelic LPL variants in a patient with HTG-AP during pregnancy. Our findings enhance the understanding of the nuanced relationship between LPL genotypes and clinical phenotypes, highlighting the importance of residual LPL function in disease manifestation and severity. Additionally, our study underscores the challenges in classifying partial loss-of-function variants in classical Mendelian disease genes according to the American College of Medical Genetics and Genomics (ACMG)'s variant classification guidelines.


Subject(s)
Hyperlipidemias , Hypertriglyceridemia , Pancreatitis , Humans , Lipoprotein Lipase/genetics , Acute Disease , HEK293 Cells , Pancreatitis/genetics , Heparin
20.
Int J Hematol ; 119(6): 755-761, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38507115

ABSTRACT

BACKGROUND: Medications used to treat acute lymphoblastic leukemia (ALL), such as L-asparaginase, can cause blood lipid disturbances. These can also be associated with polymorphisms of the lipoprotein lipase (LpL) and apolipoprotein E (APOE) genes. PROCEDURE: We aimed to investigate the association between lipid profile, certain LpL and APOE gene polymorphisms (rs268, rs328, rs1801177 and rs7412, rs429358 respectively) as well as the risk subgroup in 30 pediatric patients being treated for ALL, compared with 30 pediatric ALL survivors and 30 healthy controls. RESULTS: The only APOE gene polymorphism with significant allelic and genotypic heterogeneity was rs429358. Further analysis of this polymorphism showed that genotype (CC, CT, or TT) was significantly associated with (1) changes in the lipid profile at the end of consolidation (total cholesterol, LDL, apo-B100, and lipoprotein a) and during re-induction (total cholesterol and apo-B100), and (2) classification in the high risk-ALL subgroup (for CC genotype/C allele presence). CONCLUSIONS: Lipid abnormalities in children being treated for ALL may be associated with the APOE genotype, which is also possibly associated with risk stratification. Further research is needed to confirm the potential prognostic value of these findings.


Subject(s)
Apolipoproteins E , Lipids , Lipoprotein Lipase , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Humans , Apolipoproteins E/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/blood , Child , Male , Female , Lipoprotein Lipase/genetics , Child, Preschool , Lipids/blood , Adolescent , Polymorphism, Single Nucleotide , Genotype , Alleles , Asparaginase/administration & dosage , Asparaginase/therapeutic use , Asparaginase/adverse effects , Polymorphism, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL