Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
J Lipid Res ; 59(9): 1640-1648, 2018 09.
Article in English | MEDLINE | ID: mdl-30021760

ABSTRACT

Abetalipoproteinemia (ABL) and chylomicron retention disease (CMRD) are extremely rare recessive forms of hypobetalipoproteinemia characterized by intestinal lipid malabsorption and severe vitamin E deficiency. Vitamin E is often supplemented in the form of fat-soluble vitamin E acetate, but fat malabsorption considerably limits correction of the deficiency. In this crossover study, we administered two different forms of vitamin E, tocofersolan (a water-soluble derivative of RRR-α-tocopherol) and α-tocopherol acetate, to three patients with ABL and four patients with CMRD. The aims of this study were to evaluate the intestinal absorption characteristics of tocofersolan versus α-tocopherol acetate by measuring the plasma concentrations of α-tocopherol over time after a single oral load and to compare efficacy by evaluating the ability of each formulation to restore vitamin E storage after 4 months of treatment. In patients with ABL, tocofersolan and α-tocopherol acetate bioavailabilities were extremely low (2.8% and 3.1%, respectively). In contrast, bioavailabilities were higher in patients with CMRD (tocofersolan, 24.7%; α-tocopherol acetate, 11.4%). Plasma concentrations of α-tocopherol at 4 months were not significantly different by formulation type in ABL or CMRD. This study provides new insights about vitamin E status in ABL and CMRD and suggests the potential of different formulations as treatment options.


Subject(s)
Abetalipoproteinemia/metabolism , Hypobetalipoproteinemias/metabolism , Malabsorption Syndromes/metabolism , Vitamin E/pharmacokinetics , alpha-Tocopherol/pharmacokinetics , Adult , Biological Availability , Case-Control Studies , Drug Compounding , Drug Storage , Female , Humans , Intestinal Absorption , Male , Middle Aged , Safety , Vitamin E/blood , Vitamin E/metabolism , alpha-Tocopherol/blood , alpha-Tocopherol/metabolism
2.
Atherosclerosis ; 247: 97-104, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26894473

ABSTRACT

Familial hypercholesterolemia (FH) is a co-dominantly inherited disorder of plasma lipoprotein metabolism. The prevalence of heterozygous FH (HeFH) is between 1/500 and 1/200 whereas that of homozygous form (HoFH) is about 1/1,000,000. Diagnosis is based on cutaneous xanthomas and untreated levels of LDL-cholesterol over 500 mg/dl before 10 years of age. Life expectancy, without treatment, does not exceed 20 years of age. The aim of this study is to characterise in details a cohort of 8 HoFH paediatric patients in order to illustrate all the current therapeutic options and to add some clinical and genetic information about this rare disease. We collected demographic, clinical, biological, imaging and genotype details. Furthermore, clinical and biochemical response to different treatment methods was retrospectively evaluated. All patients had genetically proven HoFH. All patients were subject to a lipid-lowering diet and medical treatment (except one), three patients underwent a liver transplant and one an hepatocytes infusion. Medical treatment was well tolerated with a median reduction of 44% and 47% in LDL-Cholesterol and Total Cholesterol respectively. The hepatocytes transplant produced a further, though slight, decrease in cholesterol levels as opposed to medical therapy alone. Transplanted patients normalized their cholesterol levels. Since the very high cardiovascular risk, HoFH requires immediate diagnosis, treatment and monitoring. Nowadays, the use of statins remains the cornerstone of medical therapy and liver transplantation is the possibly curative therapy. Besides, high hopes are pinned in new drugs (antibody targeting PCSK9, Mipomersen and Lomitapide) and stem cells.


Subject(s)
Cholesterol/blood , Homozygote , Hyperlipoproteinemia Type II/genetics , Liver/metabolism , Mutation , Receptors, LDL/genetics , Anticholesteremic Agents/therapeutic use , Biomarkers/blood , Cardiovascular Diseases/genetics , Cardiovascular Diseases/prevention & control , Child , Child, Preschool , Combined Modality Therapy , DNA Mutational Analysis , Diet, Fat-Restricted , Female , Genetic Association Studies , Genetic Markers , Genetic Predisposition to Disease , Hepatocytes/transplantation , Humans , Hyperlipoproteinemia Type II/diagnosis , Hyperlipoproteinemia Type II/therapy , Infant , Infant, Newborn , Liver/surgery , Liver Transplantation , Male , Phenotype , Predictive Value of Tests , Retrospective Studies , Risk Factors , Treatment Outcome
3.
Pediatr Transplant ; 20(2): 241-8, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26750745

ABSTRACT

Cardiovascular diseases induce long-term morbidity and mortality of adult LT recipients. The aim of this retrospective study was to assess CVRF, lipid abnormalities, and atherosclerosis (appraised by c-IMT), more than 10 yr after pediatric LT. Thirty-one children who underwent LT between December 1990 and December 2000 were included. Median age at LT was 14 months (range 4-64), and median follow-up after LT was 11.9 yr (range 9.0-17.3). In our cohort, obesity (9.7%) and treated hypertension (9.7%) were rare. None of the patients was smoker or diabetic. High TC and TG were both observed in 6.5% of the patients. The mean c-IMT for male patients was 1.22 ± 1.55 and 1.58 ± 1.23 mm in female patients. Seven patients (22%) had a mean c-IMT above +2 s.d. Values below the 5th percentile were noted for LDL-cholesterol (58.1%), HDL-cholesterol (25.8%), apolipoprotein B (40%), and apolipoprotein A1 (20%). LDL-cholesterol and apolipoprotein B levels were significantly lower in patients treated by tacrolimus in comparison with CsA (p < 0.05). In conclusion, our results suggest that pediatric LT patients do not present significant CVRF; moreover, instead of hyperlipidemia, hypocholesterolemia (LDL-C) is frequent and immunosuppressive therapy is probably the cause.


Subject(s)
Cardiovascular Diseases/blood , Liver Failure/blood , Liver Transplantation , Adolescent , Apolipoprotein A-I/blood , Apolipoproteins B/blood , Biopsy , Cardiovascular Diseases/complications , Child , Child, Preschool , Cholesterol, HDL/blood , Cholesterol, LDL/blood , Cyclosporine/therapeutic use , Dyslipidemias/complications , Female , Follow-Up Studies , Humans , Immunosuppressive Agents/therapeutic use , Infant , Liver/pathology , Liver Failure/complications , Liver Failure/surgery , Male , Retrospective Studies , Risk Factors , Tacrolimus/therapeutic use , Transplant Recipients , Treatment Outcome
4.
PLoS One ; 9(6): e99721, 2014.
Article in English | MEDLINE | ID: mdl-24886863

ABSTRACT

BACKGROUND: Determination of lipoprotein lipase (LPL) activity is important for hyperchylomicronemia diagnosis, but remains both unreliable and cumbersome with current methods. Consequently by using human VLDL as substrate we developed a new LPL assay which does not require sonication, radioactive or fluorescent particles. METHODS: Post-heparin plasma was added to the VLDL substrate prepared by ultracentrifugation of heat inactivated normolipidemic human serums, diluted in buffer, pH 8.15. Following incubation at 37°c, the NEFA (non esterified fatty acids) produced were assayed hourly for 4 hours. LPL activity was expressed as µmol/l/min after subtraction of hepatic lipase (HL) activity, obtained following LPL inhibition with NaCl 1.5 mmol/l. Molecular analysis of LPL, GPIHBP1, APOA5, APOC2, APOE genes was available for 62 patients. RESULTS: Our method was reproducible (coefficient of variation (CV): intra-assay 5.6%, inter-assay 7.1%), and tightly correlated with the conventional radiolabelled triolein emulsion method (n = 26, r = 0.88). Normal values were established at 34.8 ± 12.8 µmol/l/min (mean ± SD) from 20 control subjects. LPL activities obtained from 71 patients with documented history of major hypertriglyceridemia showed a trimodal distribution. Among the 11 patients with a very low LPL activity (< 10 µmol/l/min), 5 were homozygous or compound heterozygous for LPL or GPIHBP1 deleterious mutations, 3 were compound heterozygous for APOA5 deleterious mutations and the p.S19W APOA5 susceptibility variant, and 2 were free of any mutations in the usual candidate genes. No homozygous gene alteration in LPL, GPIHBP1 and APOC2 genes was found in any of the patients with LPL activity > 10 µmol/l/min. CONCLUSION: This new reproducible method is a valuable tool for routine diagnosis and reliably identifies LPL activity defects.


Subject(s)
Biological Assay/methods , Heparin/chemistry , Lipoprotein Lipase/chemistry , Lipoproteins, VLDL/chemistry , Plasma/chemistry , Triglycerides/chemistry , Adolescent , Adult , Aged , Aged, 80 and over , Apolipoprotein A-V , Apolipoprotein C-II/metabolism , Apolipoproteins A/metabolism , Apolipoproteins E/metabolism , Female , Humans , Hypertriglyceridemia/blood , Hypertriglyceridemia/diagnosis , Hypertriglyceridemia/metabolism , Lipolysis , Male , Middle Aged , Plasma/metabolism , Receptors, Lipoprotein/metabolism , Triglycerides/blood , Young Adult
5.
J Hepatol ; 61(4): 891-902, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24842304

ABSTRACT

BACKGROUND & AIMS: Non-alcoholic steatohepatitis leading to fibrosis occurs in patients with abetalipoproteinemia (ABL) and homozygous or compound heterozygous familial hypobetalipoproteinemia (Ho-FHBL). We wanted to establish if liver alterations were more frequent in one of both diseases and were influenced by comorbidities. METHODS: We report genetic, clinical, histological and biological characteristics of new cases of ABL (n =7) and Ho-FHBL (n = 7), and compare them with all published ABL (51) and Ho-FHBL (22) probands. RESULTS: ABL patients, diagnosed during infancy, presented mainly with diarrhea, neurological and ophthalmological impairments and remained lean, whereas Ho-FHBL were diagnosed later, with milder symptoms often becoming overweight in adulthood. Despite subtle differences in lipid phenotype, liver steatosis was observed in both groups with a high prevalence of severe fibrosis (5/27 for Ho-FHBL vs. 4/58 for ABL (n.s.)). Serum triglycerides concentration was higher in Ho-FHBL whereas total and HDL-cholesterol were similar in both groups. In Ho-FHBL liver alterations were found to be independent from the apoB truncation size and apoB concentrations. CONCLUSIONS: Our findings provide evidence for major liver abnormalities in both diseases. While ABL and Ho-FHBL patients have subtle differences in lipid phenotype, carriers of APOB mutations are more frequently obese. These results raise the question of a complex causal link between apoB metabolism and obesity. They suggest that the genetic defect in VLDL assembly is critical for the occurrence of liver steatosis leading to fibrosis and shows that obesity and insulin resistance might contribute by increasing lipogenesis.


Subject(s)
Abetalipoproteinemia , Apolipoprotein B-100/genetics , Carrier Proteins/genetics , Hypobetalipoproteinemias , Non-alcoholic Fatty Liver Disease , Obesity , Abetalipoproteinemia/blood , Abetalipoproteinemia/diagnosis , Abetalipoproteinemia/epidemiology , Abetalipoproteinemia/genetics , Adolescent , Adult , Cholesterol, HDL/blood , Cohort Studies , Comorbidity , Female , France/epidemiology , Humans , Hypobetalipoproteinemias/blood , Hypobetalipoproteinemias/diagnosis , Hypobetalipoproteinemias/epidemiology , Hypobetalipoproteinemias/genetics , Insulin Resistance , Lipid Metabolism/genetics , Liver/metabolism , Male , Non-alcoholic Fatty Liver Disease/diagnosis , Non-alcoholic Fatty Liver Disease/epidemiology , Non-alcoholic Fatty Liver Disease/etiology , Obesity/epidemiology , Obesity/genetics , Prevalence , Triglycerides/blood
6.
PLoS One ; 9(5): e96482, 2014.
Article in English | MEDLINE | ID: mdl-24788417

ABSTRACT

BACKGROUND: Determination of lipoprotein lipase (LPL) activity is important for hyperchylomicronemia diagnosis, but remains both unreliable and cumbersome with current methods. Consequently by using human VLDL as substrate we developed a new LPL assay which does not require sonication, radioactive or fluorescent particles. METHODS: Post-heparin plasma was added to the VLDL substrate prepared by ultracentrifugation of heat inactivated normolipidemic human serums, diluted in buffer, pH 8.15. Following incubation at 37°c, the NEFA (non esterified fatty acids) produced were assayed hourly for 4 hours. LPL activity was expressed as µmol/l/min after subtraction of hepatic lipase (HL) activity, obtained following LPL inhibition with NaCl 1.5 mmol/l. Molecular analysis of LPL, GPIHBP1, APOA5, APOC2, APOE genes was available for 62 patients. RESULTS: Our method was reproducible (coefficient of variation (CV): intra-assay 5.6%, inter-assay 7.1%), and tightly correlated with the conventional radiolabelled triolein emulsion method (n = 26, r = 0.88). Normal values were established at 34.8 ± 12.8 µmol/l/min (mean ± SD) from 20 control subjects. LPL activities obtained from 71 patients with documented history of major hypertriglyceridemia showed a trimodal distribution. Among the 11 patients with a very low LPL activity (<10 µmol/l/min), 5 were homozygous or compound heterozygous for LPL or GPIHBP1 deleterious mutations, 3 were compound heterozygous for APOA5 deleterious mutations and the p.S19W APOA5 susceptibility variant, and 2 were free of any mutations in the usual candidate genes. No homozygous gene alteration in LPL, GPIHBP1 and APOC2 genes was found in any of the patients with LPL activity >10 µmol/l/min. CONCLUSION: This new reproducible method is a valuable tool for routine diagnosis and reliably identifies LPL activity defects.


Subject(s)
Enzyme Assays/methods , Lipoprotein Lipase/metabolism , Lipoproteins, VLDL/metabolism , Triglycerides/blood , Adult , Aged , Aged, 80 and over , Anticoagulants/pharmacology , Apolipoproteins E/blood , Apolipoproteins E/genetics , Apolipoproteins E/metabolism , Blood Coagulation/drug effects , DNA Mutational Analysis , Fatty Acids, Nonesterified/blood , Fatty Acids, Nonesterified/metabolism , Female , Heparin/pharmacology , Humans , Hypertriglyceridemia/blood , Hypertriglyceridemia/diagnosis , Hypertriglyceridemia/genetics , Kinetics , Lipolysis/genetics , Lipoprotein Lipase/blood , Lipoprotein Lipase/genetics , Male , Middle Aged , Receptors, Lipoprotein/blood , Receptors, Lipoprotein/genetics , Receptors, Lipoprotein/metabolism , Reproducibility of Results , Sensitivity and Specificity , Substrate Specificity , Young Adult
7.
Am J Hum Genet ; 94(1): 129-34, 2014 Jan 02.
Article in English | MEDLINE | ID: mdl-24387992

ABSTRACT

APOA5 c.*158C>T (rs2266788), located in the 3' UTR, belongs to APOA5 haplotype 2 (APOA5*2), which is strongly associated with plasma triglyceride levels and modulates the occurrence of both moderate and severe hypertriglyceridemia. Individuals with APOA5*2 display reduced APOA5 expression at the posttranscriptional level. However, the functionality of this haplotype remains unclear. We hypothesized that the hypertriglyceridemic effects of APOA5*2 could involve miRNA regulation in the APOA5 3' UTR. Bioinformatic studies have identified the creation of a potential miRNA binding site for liver-expressed miR-485-5p (MIRN485-5p) in the mutant APOA5 3' UTR with the c.*158C allele. In human embryonic kidney 293T (HEK293T) cells cotransfected with an APOA5 3' UTR luciferase reporter vector and a miR485-5p precursor, c.*158C allele expression was significantly decreased. Moreover, in HuH-7 cells endogenously expressing miR-485-5p, we observed that luciferase activity was significantly lower in the presence of the c.*158C allele than in the presence of the c.*158T allele, which was completely reversed by a miR-485-5p inhibitor. We demonstrated that the rare c.*158C APOA5 allele creates a functional target site for liver-expressed miR-485-5p. Therefore, we propose that the well-documented hypertriglyceridemic effect of APOA5*2 involves an APOA5 posttranscriptional downregulation mediated by miR-485-5p.


Subject(s)
3' Untranslated Regions/genetics , Apolipoproteins A/genetics , Genetic Variation , MicroRNAs/genetics , Triglycerides/blood , Alleles , Apolipoprotein A-V , Apolipoproteins A/metabolism , Binding Sites , Computational Biology , Down-Regulation , HEK293 Cells , Haplotypes , Humans , Liver/metabolism , Luciferases/metabolism , MicroRNAs/metabolism
8.
Diagn Pathol ; 8: 54, 2013 Apr 04.
Article in English | MEDLINE | ID: mdl-23556456

ABSTRACT

BACKGROUND: Abetalipoproteinemia (ABL; OMIM 200100) is a rare monogenic disorder of lipid metabolism characterized by reduced plasma levels of total cholesterol (TC), low density lipoprotein-cholesterol (LDL-C) and almost complete absence of apolipoprotein B (apoB). ABL results from genetic deficiency in microsomal triglyceride transfer protein (MTP; OMIM 157147). In the present study we investigated two unrelated Tunisian patients, born from consanguineous marriages, with severe deficiency of plasma low-density lipoprotein (LDL) and apo B. METHODS: Intestinal biopsies were performed and The MTTP gene was amplified by Polymerase chain reaction then directly sequenced in patients presenting chronic diarrhea and retarded growth. RESULTS: First proband was homozygous for a novel nucleotide deletion (c. 2611delC) involving the exon 18 of MTTP gene predicted to cause a non functional protein of 898 amino acids (p.H871I fsX29). Second proband was homozygous for a nonsense mutation in exon 8 (c.923 G > A) predicted to cause a truncated protein of 307 amino acids (p.W308X), previously reported in ABL patients. CONCLUSIONS: We discovered a novel mutation in MTTP gene and we confirmed the diagnosis of abetalipoproteinemia in new Tunisian families. VIRTUAL SLIDES: The virtual slide(s) for this article can be found here: http://www.diagnosticpathology.diagnomx.eu/vs/8134027928652779.


Subject(s)
Abetalipoproteinemia/genetics , Carrier Proteins/genetics , Codon, Nonsense , Sequence Deletion , Abetalipoproteinemia/blood , Abetalipoproteinemia/complications , Abetalipoproteinemia/diagnosis , Adult , Apolipoprotein B-100/blood , Apolipoprotein B-100/deficiency , Biomarkers/blood , Biopsy , Chronic Disease , Consanguinity , DNA Mutational Analysis , Diarrhea/genetics , Exons , Female , Genetic Predisposition to Disease , Growth Disorders/genetics , Heredity , Homozygote , Humans , Infant , Lipoproteins, LDL/blood , Lipoproteins, LDL/deficiency , Male , Pedigree , Phenotype , Severity of Illness Index , Tunisia , Young Adult
9.
FASEB J ; 27(7): 2855-61, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23507868

ABSTRACT

There is evidence that high-density lipoproteins (HDLs) may regulate platelet function, but disparate results exist regarding the effects of oxidized HDLs on platelets. The objective of our study was to determine the role of in vivo oxidized HDLs on platelet aggregation. Platelet aggregation and redox status were investigated in 5 patients with abetalipoproteinemia (ABLP) or homozygous hypobetalipoproteinemia, two rare metabolic diseases characterized by the absence of apolipoprotein B-containing lipoproteins, compared to 5 control subjects. Platelets isolated from plasma of patients with ABLP aggregated 4 to 10 times more than control platelets, depending on the agonist. By contrast, no differences in the extent of platelet aggregation were observed between ABLP platelet-rich plasma (PRP) and control PRP, suggesting the presence of a protective factor in ABLP plasma. ABLP HDLs inhibited agonist-induced platelet aggregation by binding to SR-BI, while control HDLs had no effect. On the other hand, lipoprotein-deficient plasma from patients with ABLP did not inhibit platelet aggregation. Severe oxidative stress was evidenced in patients with ABLP. Compared to control HDLs, ABLP HDLs showed a 40% decrease of α-tocopherol and an 11-fold increased malondialdehyde concentration. These results demonstrate that in vivo oxidized HDLs do not lose their antiaggregatory properties despite oxidation.


Subject(s)
Abetalipoproteinemia/metabolism , Blood Platelets/physiology , Lipoproteins, HDL/metabolism , Platelet Aggregation/physiology , Abetalipoproteinemia/blood , Abetalipoproteinemia/genetics , Adenosine Diphosphate/pharmacology , Adult , Apolipoproteins B/genetics , Arachidonic Acid/pharmacology , Blood Platelets/drug effects , Blood Platelets/metabolism , Collagen/pharmacology , Fatty Acids, Unsaturated/metabolism , Female , Humans , Lipoproteins, HDL/chemistry , Lipoproteins, HDL/pharmacology , Malondialdehyde/metabolism , Mutation , Oxidation-Reduction , Oxidative Stress , Platelet Aggregation/drug effects , Scavenger Receptors, Class B/metabolism , Young Adult , alpha-Tocopherol/blood , alpha-Tocopherol/metabolism
10.
J Lipid Res ; 53(3): 548-555, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22236406

ABSTRACT

Abetalipoproteinemia (ABL) is an inherited disease characterized by the defective assembly and secretion of apolipoprotein B-containing lipoproteins caused by mutations in the microsomal triglyceride transfer protein large subunit (MTP) gene (MTTP). We report here a female patient with an unusual clinical and biochemical ABL phenotype. She presented with severe liver injury, low levels of LDL-cholesterol, and subnormal levels of vitamin E, but only mild fat malabsorption and no retinitis pigmentosa or acanthocytosis. Our objective was to search for MTTP mutations and to determine the relationship between the genotype and this particular phenotype. The subject exhibited compound heterozygosity for two novel MTTP mutations: one missense mutation (p.Leu435His) and an intronic deletion (c.619-5_619-2del). COS-1 cells expressing the missense mutant protein exhibited negligible levels of MTP activity. In contrast, the minigene splicing reporter assay showed an incomplete splicing defect of the intronic deletion, with 26% of the normal splicing being maintained in the transfected HeLa cells. The small amount of MTP activity resulting from the residual normal splicing in the patient explains the atypical phenotype observed. Our investigation provides an example of a functional analysis of unclassified variations, which is an absolute necessity for the molecular diagnosis of atypical ABL cases.


Subject(s)
Abetalipoproteinemia/enzymology , Carrier Proteins/genetics , Child, Preschool , DNA Mutational Analysis , Female , Genetic Predisposition to Disease , Humans , Mutation
11.
J Pediatr Endocrinol Metab ; 25(11-12): 1191-4, 2012.
Article in English | MEDLINE | ID: mdl-23329770

ABSTRACT

Chylomicron retention disease (CRD), or Anderson disease, is a rare, hereditary cause of fat malabsorption. It is one of the familial hypocholesterolaemia syndromes, along with homozygous hypobetalipoproteinaemia (HBL) and abetalipoproteinaemia (ABL). We report clinical, laboratory and histological data as well as molecular DNA analysis in the case of a 4-month-old boy with failure to thrive and steatorrhea who was diagnosed with CRD. His mother's first cousin, who was diagnosed as hypobetalipoproteinaemia 30 years ago, was also reviewed and his diagnosis was revised to CRD. Both patients were treated with a low fat diet and supplementation with fat-soluble vitamins resulting in significant improvement. In conclusion, CRD is a well-defined cause of fat malabsorption and can be distinguished from other forms of familial hypocholesterolaemia because of its specific lipid profile.


Subject(s)
Chylomicrons/metabolism , Failure to Thrive/diagnosis , Growth Disorders/diagnosis , Lipid Metabolism Disorders/diagnosis , Malabsorption Syndromes/diagnosis , Diet, Fat-Restricted , Dietary Supplements , Duodenum/pathology , Duodenum/ultrastructure , Endoscopy, Gastrointestinal , Failure to Thrive/genetics , Failure to Thrive/metabolism , Family Health , Growth Disorders/genetics , Growth Disorders/metabolism , Humans , Infant , Lipid Metabolism Disorders/genetics , Lipid Metabolism Disorders/metabolism , Malabsorption Syndromes/genetics , Malabsorption Syndromes/metabolism , Male , Steatorrhea/diagnosis , Steatorrhea/genetics , Steatorrhea/metabolism , Vitamins/administration & dosage
12.
J Neurol ; 259(6): 1222-6, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22179783

ABSTRACT

Tangier disease (TD) (OMIM#205400) is a rare autosomal recessive disorder resulting from mutations in the ABCA1 gene, leading to decreased levels of plasma high-density lipoproteins (HDL). Peripheral neuropathy is a common finding in this disease, and may present as relapsing/remitting mono/polyneuropathies or as syringomyelia-like neuropathy. We retrospectively analyzed four patients, and report here their clinical, biological, electrophysiological, imaging, and genetic findings. Three patients had a typical pseudosyringomyelic neuropathy including facial diplegia, but asymmetrical onset was observed in one patient who had first been misdiagnosed with Lewis-Sumner syndrome. Electrophysiological pattern was heterogeneous, showing both signs of demyelination and axonal degeneration. Truncating mutations of the ABCA1 gene, including two previously undescribed mutations, were constantly found. Atypical symptom onset and demyelinating features on electrophysiological examination can be misleading in case of pseudosyringomyelic neuropathy. These reports illustrate two different neurological phenotypes in TD, namely the pseudosyringomyelic type and the Lewis-Sumner-like type, and advocate for a systematic assessment of lipid profile including HDL cholesterol in demyelinating neuropathies.


Subject(s)
Peripheral Nervous System Diseases/diagnosis , Peripheral Nervous System Diseases/physiopathology , Tangier Disease/diagnosis , ATP Binding Cassette Transporter 1 , ATP-Binding Cassette Transporters/genetics , Adult , Electrophysiological Phenomena/physiology , Humans , Male , Middle Aged , Peripheral Nervous System Diseases/complications , Retrospective Studies , Tangier Disease/complications , Tangier Disease/physiopathology
13.
J Clin Endocrinol Metab ; 96(10): E1675-9, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21816778

ABSTRACT

CONTEXT: GPIHBP1 is a new endothelial binding site for lipoprotein lipase (LPL), the key enzyme for intravascular lipolysis of triglyceride-rich lipoproteins (TGRL). We have identified two new missense mutations of the GPIHBP1 gene, C89F and G175R, by systematic sequencing in a cohort of 376 hyperchylomicronemic patients without mutations on the LPL, APOC2, or APOA5 gene. OBJECTIVE: Phenotypic expression and functional consequences of these two mutations were studied. DESIGN: We performed clinical and genotypic studies of probands and their families. GPIHBP1 functional alterations were studied in CHO pgsA-745 transfected cells. RESULTS: Probands are an adult with a homozygous G175R mutation and a child with a hemizygous C89F neomutation and a deletion of the second allele. C89F mutation was associated with a C14F signal peptide polymorphism on the same haplotype. Both patients had resistant hyperchylomicronemia, low LPL activity, and history of acute pancreatitis. In CHO pgsA-745 cells, both G175R and C14F variants reduce the expression of GPIHBP1 at the cell surface. C89F mutation is responsible for a drastic LPL-binding defect to GPIHBP1. C14F may further potentiate C89F effect. CONCLUSIONS: The emergence of hyperchylomicronemia in the generation after a neomutation further establishes a critical role for GPIHBP1 in TGRL physiopathology in humans. Our results highlight the crucial role of C65-C89 disulfide bond in LPL binding by GPIHBP1 Ly6 domain. Furthermore, we first report a mutation of the hydrophobic C-terminal domain that impairs GPIHBP1 membrane targeting.


Subject(s)
Carrier Proteins/genetics , Chylomicrons/blood , Chylomicrons/genetics , Hyperlipoproteinemia Type I/blood , Hyperlipoproteinemia Type I/genetics , Adult , Animals , Apolipoprotein A-V , Apolipoprotein C-II/genetics , Apolipoprotein C-II/metabolism , Apolipoproteins A/genetics , Apolipoproteins A/metabolism , CHO Cells , Cohort Studies , Cricetinae , Cricetulus , DNA/genetics , Humans , Infant , Lipoprotein Lipase/genetics , Lipoprotein Lipase/metabolism , Liver/enzymology , Male , Mutation/genetics , Mutation/physiology , Mutation, Missense , Pancreatitis/complications , Pancreatitis/genetics , Pedigree , Receptors, Lipoprotein
14.
Hum Mutat ; 32(7): 751-9, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21394827

ABSTRACT

Abetalipoproteinemia is a rare autosomal recessive disease characterized by low lipid levels and by the absence of apoB-containing lipoproteins. It is the consequence of microsomal triglyceride transfer protein (MTTP) deficiency. We report two patients with new MTTP mutations. We studied their functional consequences on the triglyceride transfer function using duodenal biopsies. We transfected MTTP mutants in HepG2 and HeLa cells to investigate their association with protein disulfide isomerase (PDI) and their localization at the endoplasmic reticulum. These children have a severe abetalipoproteinemia. Both of them had also a mild hypogammaglobulinemia. They are compound heterozygotes with c.619G>T and c.1237-28A>G mutations within the MTTP gene. mRNA analysis revealed abnormal splicing with deletion of exon 6 and 10, respectively. Deletion of exon 6 (Δ6-MTTP) introduced a frame shift in the reading frame and a premature stop codon at position 234. Despite the fact that Δ6-MTTP and Δ10-MTTP mutants were not capable of binding PDI, both MTTP mutant proteins normally localize at the endoplasmic reticulum. However, these two mutations induce a loss of MTTP triglyceride transfer activity. These two mutations lead to abnormal truncated MTTP proteins, incapable of binding PDI and responsible for the loss of function of MTTP, thereby explaining the severe abetalipoproteinemia phenotype of these children.


Subject(s)
Abetalipoproteinemia/genetics , Abetalipoproteinemia/pathology , Carrier Proteins/genetics , Exons/genetics , Agammaglobulinemia/genetics , Alternative Splicing/genetics , Amino Acid Sequence , Carrier Proteins/metabolism , Child , Endoplasmic Reticulum/metabolism , Female , HeLa Cells , Hep G2 Cells , Humans , Infant , Male , Microsomes/metabolism , Molecular Sequence Data , Mutation/genetics , Protein Binding/genetics , Protein Disulfide-Isomerases/genetics , Protein Disulfide-Isomerases/metabolism , Triglycerides/metabolism
15.
Orphanet J Rare Dis ; 5: 24, 2010 Sep 29.
Article in English | MEDLINE | ID: mdl-20920215

ABSTRACT

Familial hypocholesterolemia, namely abetalipoproteinemia, hypobetalipoproteinemia and chylomicron retention disease (CRD), are rare genetic diseases that cause malnutrition, failure to thrive, growth failure and vitamin E deficiency, as well as other complications. Recently, the gene implicated in CRD was identified. The diagnosis is often delayed because symptoms are nonspecific. Treatment and follow-up remain poorly defined.The aim of this paper is to provide guidelines for the diagnosis, treatment and follow-up of children with CRD based on a literature overview and two pediatric centers 'experience.The diagnosis is based on a history of chronic diarrhea with fat malabsorption and abnormal lipid profile. Upper endoscopy and histology reveal fat-laden enterocytes whereas vitamin E deficiency is invariably present. Creatine kinase (CK) is usually elevated and hepatic steatosis is common. Genotyping identifies the Sar1b gene mutation.Treatment should be aimed at preventing potential complications. Vomiting, diarrhea and abdominal distension improve on a low-long chain fat diet. Failure to thrive is one of the most common initial clinical findings. Neurological and ophthalmologic complications in CRD are less severe than in other types of familial hypocholesterolemia. However, the vitamin E deficiency status plays a pivotal role in preventing neurological complications. Essential fatty acid (EFA) deficiency is especially severe early in life. Recently, increased CK levels and cardiomyopathy have been described in addition to muscular manifestations. Poor mineralization and delayed bone maturation do occur. A moderate degree of macrovesicular steatosis is common, but no cases of steatohepatitis cirrhosis. Besides a low-long chain fat diet made up uniquely of polyunsaturated fatty acids, treatment includes fat-soluble vitamin supplements and large amounts of vitamin E. Despite fat malabsorption and the absence of postprandial chylomicrons, the oral route can prevent neurological complications even though serum levels of vitamin E remain chronically low. Dietary counseling is needed not only to monitor fat intake and improve symptoms, but also to maintain sufficient caloric and EFA intake. Despite a better understanding of the pathogenesis of CRD, the diagnosis and management of the disease remain a challenge for clinicians. The clinical guidelines proposed will helpfully lead to an earlier diagnosis and the prevention of complications.


Subject(s)
Chylomicrons/metabolism , Lipid Metabolism Disorders/diagnosis , Lipid Metabolism Disorders/therapy , Malabsorption Syndromes/diagnosis , Malabsorption Syndromes/therapy , Adult , Anthropometry , Child , Child, Preschool , Cohort Studies , Diarrhea/complications , Fatty Acids/metabolism , Female , Growth Disorders/complications , Humans , Infant , Lipid Metabolism Disorders/complications , Lipid Metabolism Disorders/genetics , Malabsorption Syndromes/complications , Malabsorption Syndromes/genetics , Male , Malnutrition/complications , Monomeric GTP-Binding Proteins/genetics , Monomeric GTP-Binding Proteins/metabolism , Mutation , Nervous System Diseases/complications , Vitamin E Deficiency/complications
16.
Hum Mutat ; 31(11): E1811-24, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20809525

ABSTRACT

Autosomal Dominant Hypercholesterolemia (ADH), characterized by isolated elevation of plasmatic LDL cholesterol and premature cardiovascular complications, is associated with mutations in 3 major genes: LDLR (LDL receptor), APOB (apolipoprotein B) and PCSK9(proprotein convertase subtilisin-kexin type 9). Through the French ADH Research Network, we collected molecular data from 1358 French probands from eleven different regions in France.Mutations in the LDLR gene were identified in 1003 subjects representing 391 unique events with 46.0% missense, 14.6% frameshift, 13.6% splice, and 11.3% nonsense mutations, 9.7% major rearrangements, 3.8% small in frame deletions/insertions, and 1.0% UTR mutations. Interestingly,175 are novel mutational events and represent 45% of the unique events we identified, highlighting a specificity of the LDLR mutation spectrum in France. Furthermore, mutations in the APOB gene were identified in 89 probands and in the PCSK9 gene in 10 probands. Comparison of available clinical and biochemical data showed a gradient of severity for ADH-causing mutations:FH=PCSK9>FDB>«Others¼ genes. The respective contribution of each known gene to ADH inthis French cohort is: LDLR 73.9%, APOB 6.6%, PCSK9 0.7%. Finally, in 19.0% of the probands,no mutation was found, thus underscoring the existence of ADH mutations located in still unknown genes.


Subject(s)
Hyperlipoproteinemia Type II/genetics , Mutation , Apolipoproteins B/genetics , Cholesterol/blood , Cohort Studies , DNA Mutational Analysis , Female , France , Genetic Variation , Humans , Hyperlipoproteinemia Type II/blood , Male , Proprotein Convertase 9 , Proprotein Convertases , Receptors, LDL/chemistry , Receptors, LDL/genetics , Serine Endopeptidases/genetics
17.
Br J Nutr ; 103(3): 422-8, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19781120

ABSTRACT

Type 2 diabetes is associated with a higher cardiovascular risk and there has been a growing interest in using dietary intervention to improve lipid profile and glucose control. The present work aims at analysing the effects of the enrichment of a normal diet with beta-glucan (3.5 g/d) in free-living type 2 diabetic subjects for 2 months, using a palatable soup. This trial was a parallel, placebo-controlled, double-blinded randomised study performed in fifty-three type 2 diabetic subjects. During a 3-week run-in period, subjects daily consumed a ready meal control soup (without beta-glucan). For the following 8 weeks, subjects were randomly assigned to consume daily either a control soup or a beta-glucan soup. Changes in lipid profile (total cholesterol (TC), HDL- and LDL-cholesterol (HDLc and LDLc), apo B and TAG) and in glucose control (HbA1c and fasting glucose) were measured. There was no significant alteration in lipid profile in the two groups (TC, HDLc, LDLc and apo B). TAG decreased significantly in the beta-glucan group compared with the control group ( - 0.12 (SD 0.38) v. 0.12 (SD 0.44) mmol/l, P = 0.03). HbA1c and fasting glucose were not reduced in any group. A single daily ingestion of 3.5 g beta-glucan, as required by official dietary recommendations, for 8 weeks did not change the lipid profile and HbA1c in type 2 diabetic subjects. To improve the metabolic profile of type 2 diabetic subjects in the long term, the quantity, the food vectors and the tolerability of beta-glucan products may be re-evaluated.


Subject(s)
Cardiovascular Diseases/epidemiology , Diabetes Mellitus, Type 2/physiopathology , Diabetic Angiopathies/epidemiology , Lipids/blood , beta-Glucans/pharmacology , Aged , Avena , Blood Glucose/drug effects , Blood Glucose/metabolism , Body Weight , C-Reactive Protein/drug effects , C-Reactive Protein/metabolism , Cardiovascular Diseases/prevention & control , Cholesterol, HDL/blood , Cholesterol, HDL/drug effects , Cholesterol, LDL/blood , Cholesterol, LDL/drug effects , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/complications , Diabetic Angiopathies/prevention & control , Diet, Diabetic , Double-Blind Method , Glycated Hemoglobin/drug effects , Glycated Hemoglobin/metabolism , Humans , Middle Aged , Placebos , Risk Factors , Surveys and Questionnaires , beta-Glucans/therapeutic use
18.
Atherosclerosis ; 208(2): 324-7, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19695572

ABSTRACT

PURPOSE: Type V hyperlipidemia (HTG V) characterized by accumulation of both chylomicrons and VLDL results from a complex combination of genetic and environmental factors. However, a large proportion of sporadic cases remains largely unexplained. In a few cases, in a context of autoimmunity, auto-antibodies inhibiting lipoprotein lipase (LPL) activity have been incriminated. To establish their contribution to common type V hyperlipidemia in subjects with no apparent evidence of autoimmune background, we systematically screened the presence of these antibodies and their inhibition properties. METHODS: Screening for circulating anti-human LPL immunoglobulin G (anti-hLPL IgG) was carried out by western blotting in 63 subjects with HTG V and 77 controls. Inhibition of lipolytic activity by plasma from these patients was measured ex vivo. RESULTS: Anti-hLPL IgG was detectable in plasma from both controls and subjects with HTG V. After establishment of a threshold value corresponding to the 95th percentile of the control population, 27% of subjects with HTG V were found to have abnormal antibody levels (P<0.001). Only plasma obtained from these hyperchylomicronemic subjects with a high level of anti-hLPL IgG inhibited triglyceride hydrolysis whereas plasma from controls or HTG subjects with normal anti-hLPL IgG levels had no inhibitory effect (-13.5+/-3.4% vs 1.6+/-3.4%; P=0.04). However, no correlation was observed between anti-hLPL IgG levels, inhibitory effect and plasma triglyceride concentration. CONCLUSION: High levels of anti-hLPL immunoreactivity could be detected in only one out of four adult patients with type V hyperchylomicronemia. Furthermore, only a minority of these subjects (less than 10%) displayed both high anti-hLPL IgG levels and substantial inhibition (>20%) of plasma lipolysis. These auto-antibodies, in this setting only, might contribute to the occurrence of a minority of sporadic type V dyslipidemia cases.


Subject(s)
Autoantibodies/chemistry , Chylomicrons/chemistry , Hyperlipidemias/blood , Hyperlipidemias/immunology , Lipoprotein Lipase/blood , Lipoprotein Lipase/immunology , Autoimmunity , Blotting, Western , Humans , Hydrolysis , Hyperlipidemias/epidemiology , Immunoglobulin G/chemistry , Ligands , Lipoprotein Lipase/antagonists & inhibitors , Models, Biological , Triglycerides/chemistry
19.
Mol Genet Metab ; 97(2): 136-42, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19285442

ABSTRACT

Lipoprotein assembly is critical for the intestinal absorption of dietary lipids and of fat-soluble vitamins. Through their inhibition of chylomicron secretion, mutations of the Sar1B gene coding for Sar1 GTPase are associated with chylomicron retention disease (CRD). The aim of this study was to describe the phenotypic expression of CRD in two clinically and genetically well characterized cohorts, and to compare their long term evolution. The study in 7 children from France (X age 11.3+/-1.7 years) and 9 from Quebec, Canada (X age 12+/-2.5 years) involved data collection from medical records for growth evaluation, neurological and ophthalmological status as well as bone density over an average follow-up period of 4.9 years for the French cohort and of 10.6 years for the Canadian one. All CRD patients presented within the first few months of life with diarrhea and failure to thrive. Severe hypocholesterolemia coupled with normal triglycerides was associated with low LDL and HDL-cholesterol, as well as with low apolipoproteins A-I and B. Varying degrees of essential fatty acid and of vitamin E deficiency were observed. The earlier diagnosis in the Canadian cohort (1.3+/-0.04 years) than in the French one (6.3+/-1.3 years) was unrelated with the severity of presenting symptoms. The fact that the disease had more impact on growth and bone density in the latter group may be related to delayed diagnosis of the disease. Vitamin E deficiency led to functional neurological and ophthalmic changes in a small number of patients but only one developed areflexia. Finally, genotype-phenotype correlation is not obvious in our cohort with CRD; even if, the Canadian subjects with the allele 409G>A had a more severe degree (P<0.001) of hypocholesterolemia than the other patients, many clinical data are inconsistent with a hypothetical genotype-phenotype correlation. This study provides new insights on the phenotypic expression of CRD over time and emphasizes the need to screen the lipid profile of infants with chronic diarrhea and failure to thrive.


Subject(s)
Chylomicrons/metabolism , Monomeric GTP-Binding Proteins/genetics , Steatorrhea/metabolism , Adolescent , Anthropometry , Bone Density , Child , Cholesterol/metabolism , Cohort Studies , Diarrhea/etiology , Diarrhea/metabolism , Eye Diseases/etiology , Eye Diseases/metabolism , Fatty Acids/metabolism , Female , Humans , Intestinal Absorption/genetics , Male , Mutation , Nervous System Diseases/etiology , Nervous System Diseases/metabolism , Steatorrhea/genetics , Vitamin E/metabolism
20.
Eur J Gastroenterol Hepatol ; 21(1): 104-8, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19060634

ABSTRACT

Familial hypobetalipoproteinemia (FHBL) is an autosomal codominantly inherited disorder of lipoprotein metabolism characterized by decreased concentrations of low-density lipoprotein-cholesterol and of apolipoprotein B (apoB). Mutations of APOB gene lead to the formation of truncated forms of apoB. The study aimed at determining the truncated form of apoB responsible for FHBL associated with liver cirrhosis in a 27-year-old man. Analysis of the patient's lipoproteins has been performed by SDS-PAGE electrophoresis followed by immunoblotting with monoclonal antibodies. DNA of the family (proband, daughter, wife, father, and mother) was extracted, and PCR amplification was realized; amplicons were screened and sequenced. Electrophoresis allowed us to identify a truncated form of apoB (close to apoB 59%), associated with a new heterozygous apoB variant, 8402 C>G. This mutation creates a stop codon (TAC>TAG, Y2807X) and predicts to generate a truncated protein (apoB-61.9%). No other causes of cirrhosis were established by comprehensive clinical and biological investigations. We described here an unusual clinical observation of a patient with FHBL and early development of liver cirrhosis due to a new truncated form of apoB.


Subject(s)
Apolipoproteins B/genetics , Hypobetalipoproteinemia, Familial, Apolipoprotein B/complications , Liver Cirrhosis/genetics , Adult , Early Diagnosis , Humans , Incidental Findings , Liver Cirrhosis/diagnosis , Male
SELECTION OF CITATIONS
SEARCH DETAIL
...