Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Eur J Neurol ; 30(11): 3497-3506, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37329328

ABSTRACT

BACKGROUND: Gut microbiota plays a role in the pathophysiology of ischaemic stroke (IS) through the bidirectional gut-brain axis. Nevertheless, little is known about sex-specific microbiota signatures in IS occurrence. METHODS: A total of 89 IS patients and 12 healthy controls were enrolled. We studied the taxonomic differences of the gut microbiota between men and women with IS by shotgun metagenomic sequencing. To evaluate the causal effect of several bacteria on IS risk, we performed a two-sample Mendelian randomisation (MR) with inverse-variance weighting (IVW) using genome-wide association analysis (GWAS) summary statistics from two cohorts of 5959 subjects with genetic and microbiota data and 1,296,908 subjects with genetic and IS data, respectively. RESULTS: α-Diversity analysis measured using Observed Species (p = 0.017), Chao1 (p = 0.009) and Abundance-based Coverage Estimator (p = 0.012) indexes revealed that IS men have a higher species richness compared with IS women. Moreover, we found sex-differences in IS patients in relation to the phylum Fusobacteria, class Fusobacteriia, order Fusobacteriales and family Fusobacteriaceae (all Bonferroni-corrected p < 0.001). MR confirmed that increased Fusobacteriaceae levels in the gut are causally associated with an increased risk of IS (IVW p = 0.02, ß = 0.32). CONCLUSIONS: Our study is the first to indicate that there are gut microbiome differences between men and women with IS, identifying high levels of Fusobacteriaceae in women as a specific risk factor for IS. Incorporating sex stratification analysis is important in the design, analysis and interpretation of studies on stroke and the gut microbiota.

2.
Stroke ; 53(7): 2320-2330, 2022 07.
Article in English | MEDLINE | ID: mdl-35209739

ABSTRACT

BACKGROUND: Stroke onset in women occurs later in life compared with men. The underlying mechanisms of these differences have not been established. Epigenetic clocks, based on DNA methylation (DNAm) profiles, are the most accurate biological age estimate. Epigenetic age acceleration (EAA) measures indicate whether an individual is biologically younger or older than expected. Our aim was to analyze whether sexual dichotomy at age of stroke onset is conditioned by EAA. METHODS: We used 2 DNAm datasets from whole blood samples of case-control genetic studies of ischemic stroke (IS), a discovery cohort of 374 IS patients (N women=163, N men=211), from GRECOS (Genotyping Recurrence Risk of Stroke) and SEDMAN (Dabigatran Study in the Early Phase of Stroke, New Neuroimaging Markers and Biomarkers) studies and a replication cohort of 981 IS patients (N women=411, N men=570) from BASICMAR register. We compared chronological age, 2 DNAm-based biomarkers of aging and intrinsic and extrinsic epigenetic age acceleration EAA (IEAA and extrinsic EAA, respectively), in IS as well as in individual IS etiologic subtypes. Horvath and Hannum epigenetic clocks were used to assess the aging rate. A proteomic study using the SOMAScan multiplex assay was performed on 26 samples analyzing 1305 proteins. RESULTS: Women present lower Hannum-extrinsic EAA values, whereas men have higher Hannum-extrinsic EAA values (women=-0.64, men=1.24, P=1.34×10-2); the same tendency was observed in the second cohort (women=-0.57, men=0.79, P=0.02). These differences seemed to be specific to cardioembolic and undetermined stroke subtypes. Additionally, 42 blood protein levels were associated with Hannum-extrinsic EAA (P<0.05), belonging to the immune effector process (P=1.54×10-6) and platelet degranulation (P<8.74×10-6) pathways. CONCLUSIONS: This study shows that sex-specific underlying biological mechanisms associated with stroke onset could be due to differences in biological age acceleration between men and women.


Subject(s)
Epigenesis, Genetic , Ischemic Stroke , Acceleration , Aging , Child, Preschool , DNA Methylation , Female , Genetic Markers , Humans , Male , Proteomics
3.
Int J Mol Sci ; 23(12)2022 Jun 20.
Article in English | MEDLINE | ID: mdl-35743317

ABSTRACT

Ischaemic stroke is a complex disease with some degree of heritability. This means that heritability factors, such as genetics, could be risk factors for ischaemic stroke. The era of genome-wide studies has revealed some of these heritable risk factors, although the data generated by these studies may also be useful in other disciplines. Analysis of these data can be used to understand the biological mechanisms associated with stroke risk and stroke outcome, to determine the causality between stroke and other diseases without the need for expensive clinical trials, or to find potential drug targets with higher success rates than other strategies. In this review we will discuss several of the most relevant studies regarding the genetics of ischaemic stroke and the potential use of the data generated.


Subject(s)
Brain Ischemia , Ischemic Stroke , Stroke , Brain Ischemia/genetics , Genome-Wide Association Study , Humans , Risk Factors , Stroke/genetics
4.
Int J Mol Sci ; 23(6)2022 Mar 15.
Article in English | MEDLINE | ID: mdl-35328582

ABSTRACT

Small vessel strokes (SVS) and intracerebral haemorrhages (ICH) are acute outcomes of cerebral small vessel disease (SVD). Genetic studies combining both phenotypes have identified three loci associated with both traits. However, the genetic cis-regulation at the protein level associated with SVD has not been studied before. We performed a proteome-wide association study (PWAS) using FUSION to integrate a genome-wide association study (GWAS) and brain proteomic data to discover the common mechanisms regulating both SVS and ICH. Dorsolateral prefrontal cortex (dPFC) brain proteomes from the ROS/MAP study (N = 376 subjects and 1443 proteins) and the summary statistics for the SVS GWAS from the MEGASTROKE study (N = 237,511) and multi-trait analysis of GWAS (MTAG)-ICH−SVS from Chung et al. (N = 240,269) were selected. We performed PWAS and then a co-localization analysis with COLOC. The significant and nominal results were validated using a replication dPFC proteome (N = 152). The replicated results (q-value < 0.05) were further investigated for the causality relationship using summary data-based Mendelian randomization (SMR). One protein (ICA1L) was significantly associated with SVS (z-score = −4.42 and p-value = 9.6 × 10−6) and non-lobar ICH (z-score = −4.8 and p-value = 1.58 × 10−6) in the discovery PWAS, with a high co-localization posterior probability of 4. In the validation PWAS, ICA1L remained significantly associated with both traits. The SMR results for ICA1L indicated a causal association of protein expression levels in the brain with SVS (p-value = 3.66 × 10−5) and non-lobar ICH (p-value = 1.81 × 10−5). Our results show that the association of ICA1L with SVS and non-lobar ICH is conditioned by the cis-regulation of its protein levels in the brain.


Subject(s)
Proteome , Stroke , Cerebral Hemorrhage/complications , Cerebral Hemorrhage/genetics , Genome-Wide Association Study , Humans , Proteome/genetics , Proteomics , Stroke/etiology
5.
Int J Mol Sci ; 20(3)2019 Feb 02.
Article in English | MEDLINE | ID: mdl-30717414

ABSTRACT

Human apolipoprotein A-I (hApoA-I) overexpression improves high-density lipoprotein (HDL) function and the metabolic complications of obesity. We used a mouse model of diabesity, the db/db mouse, to examine the effects of hApoA-I on the two main functional properties of HDL, i.e., macrophage-specific reverse cholesterol transport (m-RCT) in vivo and the antioxidant potential, as well as the phenotypic features of obesity. HApoA-I transgenic (hA-I) mice were bred with nonobese control (db/+) mice to generate hApoA-I-overexpressing db/+ offspring, which were subsequently bred to obtain hA-I-db/db mice. Overexpression of hApoA-I significantly increased weight gain and the incidence of fatty liver in db/db mice. Weight gain was mainly explained by the increased caloric intake of hA-I-db/db mice (>1.2-fold). Overexpression of hApoA-I also produced a mixed type of dyslipidemia in db/db mice. Despite these deleterious effects, the overexpression of hApoA-I partially restored m-RCT in db/db mice to levels similar to nonobese control mice. Moreover, HDL from hA-I-db/db mice also enhanced the protection against low-density lipoprotein (LDL) oxidation compared with HDL from db/db mice. In conclusion, overexpression of hApoA-I in db/db mice enhanced two main anti-atherogenic HDL properties while exacerbating weight gain and the fatty liver phenotype. These adverse metabolic side-effects were also observed in obese mice subjected to long-term HDL-based therapies in independent studies and might raise concerns regarding the use of hApoA-I-mediated therapy in obese humans.


Subject(s)
Apolipoprotein A-I/genetics , Cholesterol/metabolism , Diabetes Mellitus/genetics , Diabetes Mellitus/metabolism , Gene Expression , Macrophages/metabolism , Animals , Biological Transport , Body Weight , Disease Models, Animal , Fatty Liver/genetics , Fatty Liver/metabolism , Fatty Liver/pathology , Gene Expression Profiling , Humans , Mice
6.
Curr Atheroscler Rep ; 16(7): 424, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24821603

ABSTRACT

Sitosterolemia is a rare autosomal recessively inherited disease caused by mutations affecting ABCG5 or ABCG8, which are located on human chromosome band 2p21. Around 100 cases have been reported in the literature. Sitosterolemic patients typically exhibit a 30-fold to 100-fold increase in plasma concentrations of plant sterols. The clinical manifestations include xanthomas, premature atherosclerosis, hemolytic anemia, and macrothrombocytopenia. It is noteworthy that abnormal hematological parameters may be the only clinical feature of sitosterolemic patients, suggesting that sitosterolemia may be more frequent than previously thought. Severe accumulation of plant sterols in mouse models of sitosterolemia induced complex cardiac lesions, anemia, and macrothrombocytopenia, disrupted adrenal and liver cholesterol homeostasis, and caused infertility and hypertriglyceridemia. It remains unclear whether all disease traits are present in sitosterolemic patients. The drug ezetimibe appears to be effective in reducing plasma plant sterol levels, promotes xanthoma regression, and improves the cardiovascular and hematological signs in sitosterolemic patients.


Subject(s)
Atherosclerosis/prevention & control , Hypercholesterolemia/diagnosis , Intestinal Diseases/diagnosis , Lipid Metabolism, Inborn Errors/diagnosis , Phytosterols/adverse effects , Anemia, Hemolytic/etiology , Animals , Anticholesteremic Agents/therapeutic use , Atherosclerosis/etiology , Azetidines/therapeutic use , Ezetimibe , Humans , Hypercholesterolemia/complications , Hypercholesterolemia/drug therapy , Intestinal Diseases/complications , Intestinal Diseases/drug therapy , Lipid Metabolism, Inborn Errors/complications , Lipid Metabolism, Inborn Errors/drug therapy , Mice , Thrombocytopenia/etiology , Xanthomatosis/etiology
7.
J Thromb Haemost ; 22(4): 936-950, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38103737

ABSTRACT

BACKGROUND: Thrombolytic recombinant tissue plasminogen activator (r-tPA) treatment is the only pharmacologic intervention available in the ischemic stroke acute phase. This treatment is associated with an increased risk of intracerebral hemorrhages, known as hemorrhagic transformations (HTs), which worsen the patient's prognosis. OBJECTIVES: To investigate the association between genetically determined natural hemostatic factors' levels and increased risk of HT after r-tPA treatment. METHODS: Using data from genome-wide association studies on the risk of HT after r-tPA treatment and data on 7 hemostatic factors (factor [F]VII, FVIII, von Willebrand factor [VWF], FXI, fibrinogen, plasminogen activator inhibitor-1, and tissue plasminogen activator), we performed local and global genetic correlation estimation multitrait analyses and colocalization and 2-sample Mendelian randomization analyses between hemostatic factors and HT. RESULTS: Local correlations identified a genomic region on chromosome 16 with shared covariance: fibrinogen-HT, P = 2.45 × 10-11. Multitrait analysis between fibrinogen-HT revealed 3 loci that simultaneously regulate circulating levels of fibrinogen and risk of HT: rs56026866 (PLXND1), P = 8.80 × 10-10; rs1421067 (CHD9), P = 1.81 × 10-14; and rs34780449, near ROBO1 gene, P = 1.64 × 10-8. Multitrait analysis between VWF-HT showed a novel common association regulating VWF and risk of HT after r-tPA at rs10942300 (ZNF366), P = 1.81 × 10-14. Mendelian randomization analysis did not find significant causal associations, although a nominal association was observed for FXI-HT (inverse-variance weighted estimate [SE], 0.07 [-0.29 to 0.00]; odds ratio, 0.87; 95% CI, 0.75-1.00; raw P = .05). CONCLUSION: We identified 4 shared loci between hemostatic factors and HT after r-tPA treatment, suggesting common regulatory mechanisms between fibrinogen and VWF levels and HT. Further research to determine a possible mediating effect of fibrinogen on HT risk is needed.


Subject(s)
Hemostatics , Stroke , Humans , Tissue Plasminogen Activator/adverse effects , Tissue Plasminogen Activator/genetics , von Willebrand Factor/analysis , Genome-Wide Association Study , Nerve Tissue Proteins , Receptors, Immunologic/therapeutic use , Stroke/drug therapy , Stroke/genetics , Fibrinogen/analysis , Hemostatics/adverse effects , Risk Factors
8.
J Lipid Res ; 54(3): 649-661, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23307945

ABSTRACT

During the diagnosis of three unrelated patients with severe hypertriglyceridemia, three APOA5 mutations [p.(Ser232_Leu235)del, p.Leu253Pro, and p.Asp332ValfsX4] were found without evidence of concomitant LPL, APOC2, or GPIHBP1 mutations. The molecular mechanisms by which APOA5 mutations result in severe hypertriglyceridemia remain poorly understood, and the functional impairment/s induced by these specific mutations was not obvious. Therefore, we performed a thorough structural and functional analysis that included follow-up of patients and their closest relatives, measurement of apoA-V serum concentrations, and sequencing of the APOA5 gene in 200 nonhyperlipidemic controls. Further, we cloned, overexpressed, and purified both wild-type and mutant apoA-V variants and characterized their capacity to activate LPL. The interactions of recombinant wild-type and mutated apoA-V variants with liposomes of different composition, heparin, LRP1, sortilin, and SorLA/LR11 were also analyzed. Finally, to explore the possible structural consequences of these mutations, we developed a three-dimensional model of full-length, lipid-free human apoA-V. A complex, wide array of impairments was found in each of the three mutants, suggesting that the specific residues affected are critical structural determinants for apoA-V function in lipoprotein metabolism and, therefore, that these APOA5 mutations are a direct cause of hypertriglyceridemia.


Subject(s)
Apolipoproteins A/chemistry , Apolipoproteins A/metabolism , Hypertriglyceridemia/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Apolipoprotein A-V , Apolipoproteins A/genetics , Blotting, Western , Electrophoresis, Polyacrylamide Gel , Female , Humans , Liposomes/chemistry , Liposomes/metabolism , Male , Middle Aged , Mutagenesis, Site-Directed , Mutation , Young Adult
9.
Atherosclerosis ; 386: 117382, 2023 12.
Article in English | MEDLINE | ID: mdl-38006695

ABSTRACT

BACKGROUND AND AIMS: The lipid profile is consistently associated with coronary artery disease (CAD) and ischemic stroke (IS). However, the lipoprotein subfractions have not been deeply explored in stroke subtypes, especially in IS outcome. METHODS: We performed two-sample Mendelian randomization (MR) analysis using 92 lipid traits measured by nuclear magnetic resonance in 115,000 subjects from the UK Biobank. Data for genetic associations with IS, its subtypes, and long-term outcome (LTO) were obtained from three cohorts of European ancestry: GIGASTROKE (73,652 cases, 1,234,808 controls), GODS (n = 1791) and GISCOME (n = 6165). Results obtained using CARDIoGRAMPlusC4D were used to identify differences with CAD. RESULTS: Genetically determined low concentration of medium high-density lipoprotein (HDL) particles (odds ratio (OR) = 0.92, 95% CI 0.88-0.96; p = 3.6 × 10-4) and its cholesterol content (OR = 0.92, 95% CI 0.88-0.96; p = 1.9 × 10-4) showed causal associations with an increased risk of stroke. Genetic predisposition to high apolipoprotein (apo)B to apoA-I ratio was causally associated with an increased risk of IS (OR = 1.12, 95% CI 1.06-1.18, p = 1.1 × 10-4), and a highly suggestive association was found between non-esterified cholesterol in low-density lipoprotein (LDL) and increased risk of atherothrombotic stroke (LAS) (OR = 1.35, 95% CI 1.10-1.66; p = 4.0 × 10-3). Low cholesterol in small and medium LDL was suggestively associated with poor LTO. CONCLUSIONS: Our results support that low medium HDL concentration was causally associated with an increased stroke risk, while high levels of non-esterified cholesterol in LDL were suggestively associated with an increased risk of LAS and with a better LTO.


Subject(s)
Coronary Artery Disease , Ischemic Stroke , Stroke , Humans , Mendelian Randomization Analysis , Triglycerides/genetics , Cholesterol, LDL/genetics , Stroke/diagnosis , Stroke/epidemiology , Stroke/genetics , Cholesterol , Coronary Artery Disease/genetics , Genome-Wide Association Study , Polymorphism, Single Nucleotide , Risk Factors , Cholesterol, HDL
10.
Mol Pain ; 7: 25, 2011 Apr 12.
Article in English | MEDLINE | ID: mdl-21486477

ABSTRACT

BACKGROUND: The local administration of µ-opioid receptor (MOR) agonists attenuates neuropathic pain but the precise mechanism implicated in this effect is not completely elucidated. We investigated if nitric oxide synthesized by neuronal (NOS1) or inducible (NOS2) nitric oxide synthases could modulate the local antiallodynic effects of morphine through the peripheral nitric oxide-cGMP-protein kinase G (PKG)-ATP-sensitive K+ (KATP) channels signaling pathway activation and affect the dorsal root ganglia MOR expression during neuropathic pain. RESULTS: In wild type (WT) mice, the subplantar administration of morphine dose-dependently decreased the mechanical and thermal allodynia induced by the chronic constriction of the sciatic nerve (CCI), which effects were significantly diminished after their co-administration with different subanalgesic doses of a selective NOS1 (N-[(4S)-4-amino-5-[(2-aminoethyl)amino]pentyl]-N'-nitroguanidine tris(trifluoroacetate) salt; NANT), NOS2 (L-N(6)-(1-iminoethyl)-lysine; L-NIL), L-guanylate cyclase (1H-[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one; ODQ), PKG ((Rp)-8-(para-chlorophenylthio)guanosine-3',5'-cyclic monophosphorothioate; Rp-8-pCPT-cGMPs) inhibitor or a KATP channel blocker (glibenclamide). The evaluation of the expression of MOR in the dorsal root ganglia from sham-operated and sciatic nerve-injured WT, NOS1 knockout (KO) and NOS2-KO mice at 21 days after surgery demonstrated that, although the basal mRNA and protein levels of MOR were similar between WT and both NOS-KO animals, nerve injury only decreased their expression in WT mice. CONCLUSIONS: These results suggest that the peripheral nitric oxide-cGMP-PKG-KATP signaling pathway activation participates in the local antiallodynic effects of morphine after sciatic nerve injury and that nitric oxide, synthesized by NOS1 and NOS2, is implicated in the dorsal root ganglia down-regulation of MOR during neuropathic pain.


Subject(s)
Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , Hyperalgesia/drug therapy , Morphine/therapeutic use , Neuralgia/drug therapy , Neuralgia/metabolism , Receptors, Opioid, mu/metabolism , Animals , Hyperalgesia/etiology , Mice , Mice, Knockout , Morphine/antagonists & inhibitors , Morphine/pharmacology , Naloxone/analogs & derivatives , Naloxone/pharmacology , Nitric Oxide/metabolism , Peptide Fragments/pharmacology , Quaternary Ammonium Compounds/pharmacology , Receptors, Opioid, mu/antagonists & inhibitors , Receptors, Opioid, mu/genetics , Sciatic Nerve/injuries , Signal Transduction/drug effects , Somatostatin/pharmacology
11.
Biomedicines ; 8(9)2020 Sep 15.
Article in English | MEDLINE | ID: mdl-32942679

ABSTRACT

Familial hypercholesterolemia (FH) is associated with mutations in the low-density lipoprotein (LDL) receptor (LDLR), apolipoprotein B (APOB), and proprotein convertase subtilisin/kexin 9 (PCSK9) genes. A pathological variant has not been identified in 30-70% of clinically diagnosed FH patients, and a burden of LDL cholesterol (LDL-c)-raising alleles has been hypothesized as a potential cause of hypercholesterolemia in these patients. Our aim was to study the distribution of weighted LDL-c-raising single-nucleotide polymorphism (SNP) scores (weighted gene scores or wGS) in a population recruited in a clinical setting in Catalonia. The study included 670 consecutive patients with a clinical diagnosis of FH and a prior genetic study involving 250 mutation-positive (FH/M+) and 420 mutation-negative (FH/M-) patients. Three wGSs based on LDL-c-raising variants were calculated to evaluate their distribution among FH patients and compared with 503 European samples from the 1000 Genomes Project. The FH/M- patients had significantly higher wGSs than the FH/M+ and control populations, with sensitivities ranging from 42% to 47%. A wGS based only on the SNPs significantly associated with FH (wGS8) showed a higher area under the receiver operating characteristic curve, and higher diagnostic specificity and sensitivity, with 46.4% of the subjects in the top quartile. wGS8 would allow for the assignment of a genetic cause to 66.4% of the patients if those with polygenic FH are added to the 37.3% of patients with monogenic FH. Our data indicate that a score based on 8 SNPs and the75th percentile cutoff point may identify patients with polygenic FH in Catalonia, although with limited diagnostic sensitivity and specificity.

12.
Atherosclerosis ; 283: 52-60, 2019 04.
Article in English | MEDLINE | ID: mdl-30782561

ABSTRACT

BACKGROUND AND AIMS: Primary hypobetalipoproteinemia is generally considered a heterogenic group of monogenic, inherited lipoprotein disorders characterized by low concentrations of LDL cholesterol and apolipoprotein B in plasma. Lipoprotein disorders include abetalipoproteinemia, familial hypobetalipoproteinemia, chylomicron retention disease, and familial combined hypolipidemia. Our aim was to review and analyze the results of the molecular analysis of hypolipidemic patients studied in our laboratory over the last 15 years. METHODS: The study included 44 patients with clinical and biochemical data. Genomic studies were performed and genetic variants were characterized by bioinformatics analysis. A weighted LDL cholesterol gene score was calculated to evaluate common variants associated with impaired lipid concentrations and their distribution among patients. RESULTS: Twenty-three patients were genetically confirmed as affected by primary hypobetalipoproteinemia. In this group of patients, the most prevalent mutated genes were APOB (in 17 patients, with eight novel mutations identified), SAR1B (in 3 patients, with one novel mutation identified), ANGPTL3 (in 2 patients), and MTTP (in 1 patient). The other 21 patients could not be genetically diagnosed with hypobetalipoproteinemia despite presenting suggestive clinical and biochemical features. In these patients, two APOB genetic variants associated with lower LDL cholesterol were more frequent than in controls. Moreover, the LDL cholesterol gene score, calculated with 11 SNPs, was significantly lower in mutation-negative patients. CONCLUSIONS: Around half of the patients could be genetically diagnosed. The results suggest that, in at least some of the patients without an identified mutation, primary hypobetalipoproteinemia may have a polygenic origin.


Subject(s)
Angiopoietin-like Proteins/genetics , Apolipoproteins B/genetics , Carrier Proteins/genetics , DNA/genetics , Hypobetalipoproteinemia, Familial, Apolipoprotein B/genetics , Monomeric GTP-Binding Proteins/genetics , Mutation , Adolescent , Adult , Aged , Angiopoietin-Like Protein 3 , Angiopoietin-like Proteins/metabolism , Apolipoproteins B/metabolism , Carrier Proteins/metabolism , Child , Child, Preschool , DNA Mutational Analysis , Female , Genetic Markers , Heterozygote , Homozygote , Humans , Hypobetalipoproteinemia, Familial, Apolipoprotein B/diagnosis , Hypobetalipoproteinemia, Familial, Apolipoprotein B/metabolism , Infant , Male , Microsomes , Middle Aged , Monomeric GTP-Binding Proteins/metabolism , Phenotype , Young Adult
13.
Clin Chim Acta ; 487: 270-274, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30308187

ABSTRACT

Autosomal dominant hypercholesterolemia, being referred to as familial hypercholesterolemia (FH), is mainly due to defective LDL receptor (LDLR) function, but is also associated with variants in genes encoding APOB (LDLR ligand) and PCSK9, the catabolic regulator of LDLR. The signal-transducing adaptor family member 1 (STAP1) gene has been recently linked to FH. We describe the case of a 56-year-old male patient found to have hypercholesterolemia at age 34, but who did not continue follow-up nor received treatment with lipid-lowering drugs. At age 55 he suffered a myocardial infarction. A systematic NGS analysis did not show point mutations in the LDLR, APOB, LDLRAP1, or PCSK9 genes, nor large rearrangements of the LDLR gene, but revealed the heterozygous missense variant rs199787258 of STAP1 (c.526C > T; p.Pro176Ser). This variant was also found in heterozygosis in the two siblings of the index case, who also had hypercholesterolemia, but did not cosegregate in his progeny. A bioinformatics analysis and available structural information predicts p.Pro176Ser as the most damaging of all STAP1 missense variants associated with familial hypercholesterolemia. Our findings confirm and extend the linkage between STAP1 variants and FH, and point to an important role of this adaptor protein within a signaling pathway that affects cholesterol homeostasis.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Hyperlipoproteinemia Type II/genetics , Humans , Male , Middle Aged , Mutation
14.
J Clin Lipidol ; 12(6): 1452-1462, 2018.
Article in English | MEDLINE | ID: mdl-30293936

ABSTRACT

BACKGROUND: Autosomal dominant hypercholesterolemia (ADH) is associated with mutations in the low-density lipoprotein (LDL) receptor (LDLR), apolipoprotein B (APOB), and proprotein convertase subtilisin/kexin 9 (PCSK9) genes, and it is estimated to be greatly underdiagnosed. The most cost-effective strategy for increasing ADH diagnosis is a cascade screening from mutation-positive probands. OBJECTIVE: The objective of this study was to evaluate the results from 2008 to 2016 of ADH genetic analysis performed in our clinical laboratory, serving most lipid units of Catalonia, a Spanish region with approximately 7.5 million inhabitants. METHODS: After the application of the Dutch Lipid Clinic Network (DLCN) clinical diagnostic score for ADH, this information and blood or saliva from 23 different lipid clinic units were investigated in our laboratory. DNA was screened for mutations in LDLR, APOB, and PCSK9, using the DNA-array LIPOchip, the next-generation sequencing SEQPRO LIPO RS platform, and multiplex ligation-dependent probe amplification (MLPA). The Simon Broome Register Group (SBRG) criteria was calculated and analyzed for comparative purposes. RESULTS: A total of 967 unrelated samples were analyzed. From this, 158 pathogenic variants were detected in 356 patients. The main components of the DLCN criteria associated with the presence of mutation were plasma LDL cholesterol (LDLc), age, and the presence of tendinous xanthomata. The contribution of family history to the diagnosis was lower than in other studies. DLCN and SBRG were similarly useful for predicting the presence of mutation. CONCLUSION: In a real clinical practice, multicenter setting in Catalonia, the percentage of positive genetic diagnosis in patients potentially affected by ADH was 38.6%. The DLCN showed a relatively low capacity to predict mutation detection but a higher one for ruling out mutation.


Subject(s)
Hyperlipoproteinemia Type II/genetics , Hyperlipoproteinemia Type II/metabolism , Adult , Female , Humans , Hyperlipoproteinemia Type II/diagnosis , Hyperlipoproteinemia Type II/epidemiology , Male , Middle Aged , Mutation , Pedigree , Spain/epidemiology
15.
Arterioscler Thromb Vasc Biol ; 25(9): e128-32, 2005 Sep.
Article in English | MEDLINE | ID: mdl-15994442

ABSTRACT

BACKGROUND: Overexpression of human apolipoprotein (apo) A-II in transgenic mice induces high-density lipoprotein (HDL) deficiency, and increased atherosclerosis susceptibility only when fed an atherogenic diet. This may, in part, be caused by impairment in reverse cholesterol transport (RCT). METHODS AND RESULTS: [3H]cholesterol-labeled macrophages were injected intraperitoneally into mice maintained on a chow diet or an atherogenic diet. Plasma [3H]cholesterol did not differ from human apoA-II transgenic and control mice at 24 or 48 hours after the label injection. On the chow diet, human apoA-II transgenic mice presented increased [3H]cholesterol in liver (1.3-fold) and feces (6-fold) compared with control mice (P<0.05). The magnitude of macrophage-specific RCT did not differ between transgenic and control mice fed the atherogenic diet. CONCLUSIONS: Human apoA-II maintains effective RCT from macrophages to feces in vivo despite an HDL deficiency. These findings suggest that the increased atherosclerotic lesions observed in apoA-II transgenic mice fed an atherogenic diet are not caused by impairment in macrophage-specific RCT.


Subject(s)
Apolipoprotein A-II/genetics , Atherosclerosis/immunology , Atherosclerosis/metabolism , Cholesterol/pharmacokinetics , Macrophages/metabolism , Animals , Apolipoprotein A-II/metabolism , Cells, Cultured , Diet, Atherogenic , Feces , Gene Expression , Humans , Lipoproteins, HDL/deficiency , Lipoproteins, HDL/metabolism , Liver/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Tritium
16.
Clin Chim Acta ; 455: 134-48, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-26868089

ABSTRACT

The adequate absorption of lipids is essential for all mammalian species due to their inability to synthesize some essential fatty acids and fat-soluble vitamins. Chylomicrons (CMs) are large, triglyceride-rich lipoproteins that are produced in intestinal enterocytes in response to fat ingestion, which function to transport the ingested lipids to different tissues. In addition to the contribution of CMs to postprandial lipemia, their remnants, the degradation products following lipolysis by lipoprotein lipase, are linked to cardiovascular disease. In this review, we will focus on the structure-function and metabolism of CMs. Second, we will analyze the impact of gene defects reported to affect CM metabolism and, also, the role of CMs in other pathologies, such as atherothrombotic cardiovascular disease and diabetes mellitus. Third, we will provide an overview of the laboratory tests currently used to study CM disorders, and, finally, we will highlight current treatments in diseases affecting CMs.


Subject(s)
Chylomicrons/chemistry , Apolipoproteins/chemistry , Chylomicrons/metabolism , Genetic Therapy , Humans , Hyperlipidemias/metabolism , Hyperlipidemias/therapy
17.
Eur J Intern Med ; 16(7): 507-9, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16275546

ABSTRACT

BACKGROUND: Given the clinical impression that patients with type 1 multiple endocrine neoplasia (MEN-1) frequently display abnormal glucose and lipoprotein concentrations, we compared affected subjects followed in our outpatient clinic with their non-affected relatives. METHODS: The clinical histories of 35 members (19 affected) from six families with MEN-1 were reviewed. Total cholesterol, triglycerides, insulin, glucose, and insulin resistance (homeostasis model) were compared in affected and non-affected subjects. RESULTS: The affected subjects displayed higher total cholesterol and triglycerides and more insulin resistance. CONCLUSION: Patients with MEN-1 seem to be more insulin-resistant and to have a higher cardiovascular risk than their non-affected relatives. Three non-exclusive, biologically plausible hypotheses are proposed.

18.
Atherosclerosis ; 174(2): 275-85, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15136057

ABSTRACT

Phenytoin (PHT) increases high density lipoprotein cholesterol (HDL-C) and reduces coronary artery disease mortality in humans. We report the results of PHT treatment on atherosclerosis susceptibility and lipid profile in four different types of mouse: control C57BL/6 mice and cholesteryl ester transfer protein transgenic mice as models of fatty streak, and LDL receptor-deficient mice and apolipoprotein E-deficient mice as models of mature atherosclerosis. Each mouse type was fed an appropriate diet to induce atherosclerosis and prevent liver toxicity. PHT treatment demonstrated a protective effect in all models. Reduction in aortic atherosclerotic area by PHT treatment was more evident in early atherosclerosis (2.3-fold) than in mature atherosclerosis (decreases of 40 and 23%, respectively, but only in mice in the upper 50% percentile of plasma PHT concentration). Atherosclerosis prevention was not concomitant with a consistent increase in HDL-C or any other protective change in the lipid profile. Different analyses of potential antiatherogenic HDL functions did not provide additional information. Microarray liver gene expression analyses identified a potential atheroprotective mechanism characterized by decreased expression of syndecan-4, RhoA2, double LIM protein-1, zeta-chain-associated protein kinase-70 and interleukin 6 receptor-alpha. However, to demonstrate that these changes are part of a PHT-antiatherogenic effect, they will need to be found also in arteries, maintained at protein level and proved to be causal rather than reactive.


Subject(s)
Arteriosclerosis/drug therapy , Cholesterol, HDL/drug effects , Phenytoin/pharmacology , Analysis of Variance , Animals , Apolipoproteins E/deficiency , Arteriosclerosis/genetics , Cholesterol, HDL/genetics , Diet, Atherogenic , Disease Models, Animal , Down-Regulation , Female , Gene Expression Regulation , Lipid Metabolism , Lipid Peroxidation/drug effects , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phenytoin/metabolism , Probability , RNA/analysis , Random Allocation , Species Specificity , Statistics, Nonparametric , Up-Regulation
19.
Clin Chim Acta ; 429: 61-8, 2014 Feb 15.
Article in English | MEDLINE | ID: mdl-24291057

ABSTRACT

BACKGROUND: Familial chylomicronemia (type I hyperlipidemia) is a rare autosomal recessive disease due mainly to rare variants in the lipoprotein lipase (LPL) gene sequence. Molecular diagnosis of LPL deficiency is now a requirement for the first gene therapy treatment approved in the European Union. Altered coding sequence variants in APOC2, APOA5 or GPIHBP-1 can also cause familial chylomicronemia. Herein, we report the results of our molecular diagnostic activity in this topic, carried out in the setting of a Spanish clinical practice hospital laboratory, which was also extended to some patients who were more likely to have type V hyperlipidemia. METHODS: Samples from twenty-nine unrelated probands with severe hypertriglyceridemia were referred for molecular diagnosis. Samples were first screened for LPL sequence variants by DNA sequencing and, in the absence of alterations, subsequent analysis of APOC2, APOA5, and GPIHBP1 genes was undertaken. Analysis of LPL function in vitro was further studied in two previously uncharacterized LPL sequence variants. RESULTS: Fourteen different, loss-of-function variants were found in the LPL gene: 4 were novel or uncharacterized allelic variants, and of these, 2 were directly shown to affect function. Twenty of 29 probands presented at least one LPL gene allele variant: 8 were homozygous, 9 compound heterozygous and 3 heterozygous. In 13 probands, the finding of two loss-of-function variants supported the diagnosis of LPL deficiency. None of the probands presented sequence variants in the APOC2 gene, whereas 3 presented rare variants within the APOA5 gene. Four of the five patients heterozygous for a common variant in the GPIHBP-1 gene also carried APOA5 sequence variants. CONCLUSIONS: Loss-of-function LPL variants leading to familial chylomicronemia were found in 13 patients, accounting for a significant proportion of the LPL-deficient patients predicted to live in Spain.


Subject(s)
Clinical Laboratory Techniques , Hyperlipidemias/diagnosis , Hyperlipidemias/enzymology , Lipoprotein Lipase/deficiency , Adolescent , Adult , Animals , Apolipoprotein A-V , Apolipoprotein C-II/genetics , Apolipoproteins A/genetics , COS Cells , Child , Child, Preschool , Chlorocebus aethiops , Female , Genetic Variation , Humans , Hyperlipidemias/genetics , Infant , Infant, Newborn , Lipoprotein Lipase/genetics , Male , Middle Aged , Mutagenesis , Receptors, Lipoprotein/genetics , Young Adult
20.
Mol Nutr Food Res ; 57(10): 1814-24, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23754667

ABSTRACT

SCOPE: High plasma homocysteine concentrations have been associated with increased risk of cardiovascular disease both in humans and experimental animal models, whereas plasma HDL-cholesterol concentration is inversely correlated with such disorders. This work aimed to study the impact of methionine-induced hyperhomocysteinemia (HHcy) on two major antiatherogenic functions of HDL, namely their capacity to prevent LDL oxidation and induce in vivo macrophage-specific reverse cholesterol transport. METHODS AND RESULTS: Methionine-induced HHcy in mice resulted in an approximately 20% decreased concentration of HDL-cholesterol and HDL main protein component, apolipoprotein A-I. The HDL potential to resist oxidation as well as to prevent LDL oxidative modification was impaired in hyperhomocysteinemic mice. Activities of paraoxonase-1 and platelet activation factor acetylhydrolase, two of the main HDL-associated enzymes with antioxidant activity, were reduced. The ability of HDL to efflux cholesterol from macrophages was decreased in hyperhomocysteinemic mice; however, the in vivo macrophage-specific reverse cholesterol transport measured as the output of labeled cholesterol into feces did not significantly differ between groups. CONCLUSION: Our data indicate that the HDL from methionine-induced hyperhomocysteinemic mice was more prone to oxidation and displayed lower capacity to protect LDL against oxidative modification than that of control mice, highlighting a mechanism by which a diet-induced HHcy may facilitate progression of atherosclerosis.


Subject(s)
Antioxidants/metabolism , Cholesterol, HDL/antagonists & inhibitors , Hyperhomocysteinemia/chemically induced , Macrophages/metabolism , Methionine/adverse effects , Animals , Apolipoprotein A-I/blood , Aryldialkylphosphatase/blood , Atherosclerosis/pathology , Biological Transport , Cell Line , Cholesterol, HDL/blood , Disease Models, Animal , Female , Homocysteine/blood , Hyperhomocysteinemia/complications , Hyperhomocysteinemia/pathology , Lipid Metabolism/drug effects , Liver/drug effects , Liver/metabolism , Male , Methionine/administration & dosage , Mice , Mice, Inbred C57BL , Mice, Transgenic
SELECTION OF CITATIONS
SEARCH DETAIL