Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Genes (Basel) ; 12(12)2021 11 30.
Article in English | MEDLINE | ID: mdl-34946879

ABSTRACT

Due to newborn screening for X-linked adrenoleukodystrophy (ALD), and the use of exome sequencing in clinical practice, the detection of variants of unknown significance (VUS) in the ABCD1 gene is increasing. In these cases, functional tests in fibroblasts may help to classify a variant as (likely) benign or pathogenic. We sought to establish reference ranges for these tests in ALD patients and control subjects with the aim of helping to determine the pathogenicity of VUS in ABCD1. Fibroblasts from 36 male patients with confirmed ALD, 26 healthy control subjects and 17 individuals without a family history of ALD, all with an uncertain clinical diagnosis and a VUS identified in ABCD1, were included. We performed a combination of tests: (i) a test for very-long-chain fatty acids (VLCFA) levels, (ii) a D3-C22:0 loading test to study the VLCFA metabolism and (iii) immunoblotting for ALD protein. All ALD patient fibroblasts had elevated VLCFA levels and a reduced peroxisomal ß-oxidation capacity (as measured by the D3-C16:0/D3-C22:0 ratio in the D3-C22:0 loading test) compared to the control subjects. Of the VUS cases, the VLCFA metabolism was not significantly impaired (most test results were within the reference range) in 6/17, the VLCFA metabolism was significantly impaired (most test results were within/near the ALD range) in 9/17 and a definite conclusion could not be drawn in 2/17 of the cases. Biochemical studies in fibroblasts provided clearly defined reference and disease ranges for the VLCFA metabolism. In 15/17 (88%) VUS we were able to classify the variant as being likely benign or pathogenic. This is of great clinical importance as new variants will be detected.


Subject(s)
ATP Binding Cassette Transporter, Subfamily D, Member 1/genetics , Adrenoleukodystrophy/genetics , Fibroblasts/metabolism , Mutation , ATP Binding Cassette Transporter, Subfamily D, Member 1/metabolism , Adrenoleukodystrophy/metabolism , Adult , Fatty Acids/metabolism , Humans , Male , Middle Aged , Reference Values
2.
J Med Genet ; 54(5): 330-337, 2017 05.
Article in English | MEDLINE | ID: mdl-27799409

ABSTRACT

BACKGROUND: Acyl-CoA binding domain containing protein 5 (ACBD5) is a peroxisomal membrane protein with a cytosolic acyl-CoA binding domain. Because of its acyl-CoA binding domain, ACBD5 has been assumed to function as an intracellular carrier of acyl-CoA esters. In addition, a role for ACBD5 in pexophagy has been suggested. However, the precise role of ACBD5 in peroxisomal metabolism and/or functioning has not yet been established. Previously, a genetic ACBD5 deficiency was identified in three siblings with retinal dystrophy and white matter disease. We identified a pathogenic mutation in ACBD5 in another patient and studied the consequences of the ACBD5 defect in patient material and in ACBD5-deficient HeLa cells to uncover this role. METHODS: We studied a girl who presented with progressive leukodystrophy, syndromic cleft palate, ataxia and retinal dystrophy. We performed biochemical, cell biological and molecular studies in patient material and in ACBD5-deficient HeLa cells generated by CRISPR-Cas9 genome editing. RESULTS: We identified a homozygous deleterious indel mutation in ACBD5, leading to complete loss of ACBD5 protein in the patient. Our studies showed that ACBD5 deficiency leads to accumulation of very long-chain fatty acids (VLCFAs) due to impaired peroxisomal ß-oxidation. No effect on pexophagy was found. CONCLUSIONS: Our investigations strongly suggest that ACBD5 plays an important role in sequestering C26-CoA in the cytosol and thereby facilitates transport into the peroxisome and subsequent ß-oxidation. Accordingly, ACBD5 deficiency is a novel single peroxisomal enzyme deficiency caused by impaired VLCFA metabolism, leading to retinal dystrophy and white matter disease.


Subject(s)
Adaptor Proteins, Signal Transducing/deficiency , Fatty Acids/metabolism , Membrane Proteins/deficiency , Peroxisomes/metabolism , Acyl Coenzyme A/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Autophagy , Child, Preschool , DNA, Complementary/genetics , Female , Fibroblasts/metabolism , Genetic Complementation Test , HeLa Cells , Humans , Infant , Magnetic Resonance Imaging , Membrane Proteins/metabolism , Skin/pathology
3.
Orphanet J Rare Dis ; 7: 90, 2012 Nov 22.
Article in English | MEDLINE | ID: mdl-23181892

ABSTRACT

BACKGROUND: D-bifunctional protein (DBP) deficiency is typically apparent within the first month of life with most infants demonstrating hypotonia, psychomotor delay and seizures. Few children survive beyond two years of age. Among patients with prolonged survival all demonstrate severe gross motor delay, absent language development, and severe hearing and visual impairment. DBP contains three catalytically active domains; an N-terminal dehydrogenase, a central hydratase and a C-terminal sterol carrier protein-2-like domain. Three subtypes of the disease are identified based upon the domain affected; DBP type I results from a combined deficiency of dehydrogenase and hydratase activity; DBP type II from isolated hydratase deficiency and DBP type III from isolated dehydrogenase deficiency. Here we report two brothers (16½ and 14 years old) with DBP deficiency characterized by normal early childhood followed by sensorineural hearing loss, progressive cerebellar and sensory ataxia and subclinical retinitis pigmentosa. METHODS AND RESULTS: Biochemical analysis revealed normal levels of plasma VLCFA, phytanic acid and pristanic acid, and normal bile acids in urine; based on these results no diagnosis was made. Exome analysis was performed using the Agilent SureSelect 50Mb All Exon Kit and the Illumina HiSeq 2000 next-generation-sequencing (NGS) platform. Compound heterozygous mutations were identified by exome sequencing and confirmed by Sanger sequencing within the dehydrogenase domain (c.101C>T; p.Ala34Val) and hydratase domain (c.1547T>C; p.Ile516Thr) of the 17ß-hydroxysteroid dehydrogenase type 4 gene (HSD17B4). These mutations have been previously reported in patients with severe-forms of DBP deficiency, however each mutation was reported in combination with another mutation affecting the same domain. Subsequent studies in fibroblasts revealed normal VLCFA levels, normal C26:0 but reduced pristanic acid beta-oxidation activity. Both DBP hydratase and dehydrogenase activity were markedly decreased but detectable. CONCLUSIONS: We propose that the DBP phenotype seen in this family represents a distinct and novel subtype of DBP deficiency, which we have termed type IV based on the presence of a missense mutation in each of the domains of DBP resulting in markedly reduced but detectable hydratase and dehydrogenase activity of DBP. Given that the biochemical testing in plasma was normal in these patients, this is likely an underdiagnosed form of DBP deficiency.


Subject(s)
17-Hydroxysteroid Dehydrogenases/deficiency , 17-Hydroxysteroid Dehydrogenases/genetics , Hydro-Lyases/deficiency , Hydro-Lyases/genetics , Cerebellar Ataxia/blood , Cerebellar Ataxia/genetics , Cerebellar Ataxia/urine , Fatty Acids/blood , Fatty Acids/urine , Hearing Loss, Sensorineural/blood , Hearing Loss, Sensorineural/genetics , Hearing Loss, Sensorineural/urine , Heterozygote , Mutation , Peroxisomal Multifunctional Protein-2 , Phytanic Acid/blood , Polyneuropathies/blood , Polyneuropathies/genetics , Polyneuropathies/urine , Retinitis Pigmentosa/blood , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/urine
4.
Brain ; 132(Pt 2): 482-92, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19022859

ABSTRACT

Peroxisomes are organelles responsible for multiple metabolic pathways including, the biosynthesis of plasmalogens, a class of phospholipids, and the beta-oxidation of very-long-chain fatty acids (VLCFA). Lack of peroxisomes or dysfunction in any of their normal functions is the cellular basis for human peroxisomal disorders. Here we used mouse models to understand and define the biochemical and cellular determinants that mediate the pathophysiological consequences caused by peroxisomal dysfunctions. We investigated the role and effects of cellular plasmalogens and VLCFA accumulation in liver, testis and nervous tissue using Pex7 and Abcd1 knockout (KO) mice. In addition, we also generated a Pex7:Abcd1 double KO mouse to investigate how different peroxisomal dysfunctions modulate cellular function and pathology. We found that plasmalogens function as fundamental structural phospholipids and protect cells from damage caused by VLCFA accumulation. In testis, plasmalogens protect spermatocytes from VLCFA-induced degeneration and apoptosis. In nervous tissue, we found that gliosis, inflammatory demyelination and axonopathy caused by accumulation of VLCFA are modulated by plasmalogens. Our findings demonstrate the importance of normal peroxisomal functioning and allow the understanding of the pathological causality of peroxisomal dysfunctions. Nervous tissue deficient in plasmalogens is more prone to damage, illustrating the importance of plasmalogens in peroxisomal disorders including Zellweger syndrome and X-linked adrenoleukodystrophy.


Subject(s)
Adrenoleukodystrophy/metabolism , Brain/metabolism , Fatty Acids/metabolism , Plasmalogens/metabolism , Adrenoleukodystrophy/pathology , Animals , Astrocytes/metabolism , Astrocytes/pathology , Axons/pathology , Blotting, Western , Brain/pathology , Chemokine CCL22/genetics , Demyelinating Diseases , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia/metabolism , Microglia/pathology , Peroxisomal Targeting Signal 2 Receptor , Peroxisomes/pathology , Receptors, Cytoplasmic and Nuclear/genetics
5.
Am J Physiol Endocrinol Metab ; 296(1): E211-21, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18854420

ABSTRACT

Peroxisomes are essential organelles exerting key functions in fatty acid metabolism such as the degradation of very long-chain fatty acids (VLCFAs). VLCFAs accumulate in X-adrenoleukodystrophy (X-ALD), a disease caused by deficiency of the Abcd1 peroxisomal transporter. Its closest homologue, Abcd2, exhibits a high degree of functional redundancy on the catabolism of VLCFA, being able to prevent X-ALD-related neurodegeneration in the mouse. In the search for specific roles of Abcd2, we screened fatty acid profiles in organs and primary neurons of mutant knockout mice lacking Abcd2 in basal conditions and under dietary challenges. Our results indicate that ABCD2 plays a role in the degradation of long-chain saturated and omega9-monounsaturated fatty acids and in the synthesis of docosahexanoic acid (DHA). Also, we demonstrated a defective VLCFA beta-oxidation ex vivo in brain slices of Abcd1 and Abcd2 knockouts, using radiolabeled hexacosanoic acid and the precursor of DHA as substrates. As DHA levels are inversely correlated with the incidence of Alzheimer's and several degenerative conditions, we suggest that ABCD2 may act as modulator/modifier gene and therapeutic target in rare and common human disorders.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Dietary Fats/metabolism , Fatty Acids/metabolism , Peroxisomes/metabolism , ATP Binding Cassette Transporter, Subfamily D , ATP-Binding Cassette Transporters/genetics , Adrenal Glands/metabolism , Animals , Fatty Acids/analysis , Female , Homeostasis , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Peroxisomes/genetics , Phylogeny , Sciatic Nerve/metabolism , Spinal Cord/metabolism
6.
Hum Mutat ; 30(1): 93-8, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18712838

ABSTRACT

Proteins destined for the peroxisomal matrix are targeted by virtue of a peroxisomal targeting sequence type 1 (PTS1) or type 2 (PTS2). In humans, targeting of either class of proteins relies on a cytosolic receptor protein encoded by the PEX5 gene. Alternative splicing of PEX5 results in two protein variants, PEX5S and PEX5L. PEX5S is exclusively involved in PTS1 protein import, whereas PEX5L mediates the import of both PTS1 and PTS2 proteins. Genetic complementation testing with over 500 different fibroblast cell lines from patients diagnosed with a peroxisome biogenesis disorder (PBD) identified 11 cell lines with a defect in PEX5. The aim of this study was to characterize these cell lines at a biochemical and genetic level. To this end, the cultured fibroblasts were analyzed for very long chain fatty acid (VLCFA) concentrations, peroxisomal beta-and alpha-oxidation, dihydroxyacetone-phosphate acyltransferase (DHAPAT) activity, peroxisomal thiolase, and catalase immunofluorescence. Mutation analysis of the PEX5 gene revealed 11 different mutations, eight of which are novel. PTS1- and PTS2-protein import capacity was assessed by transfection of the cells with green fluorescent protein (GFP) tagged with either PTS1 or PTS2. Six cell lines showed a defect in both PTS1 and PTS2 protein import, whereas four cell lines only showed a defect in PTS1 protein import. The location of the different mutations within the PEX5 amino acid sequence correlates rather well with the peroxisomal protein import defect observed in the cell lines.


Subject(s)
Genotype , Peroxisomal Disorders/genetics , Peroxisomes/metabolism , Phenotype , Receptors, Cytoplasmic and Nuclear/genetics , Amino Acid Sequence , Cell Line , DNA Mutational Analysis , Fibroblasts/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Models, Biological , Molecular Sequence Data , Mutation , Peroxisomal Disorders/metabolism , Peroxisome-Targeting Signal 1 Receptor , Protein Transport , Receptors, Cytoplasmic and Nuclear/metabolism , Transfection
7.
N Engl J Med ; 356(17): 1736-41, 2007 Apr 26.
Article in English | MEDLINE | ID: mdl-17460227

ABSTRACT

We report on a newborn girl with microcephaly, abnormal brain development, optic atrophy and hypoplasia, persistent lactic acidemia, and a mildly elevated plasma concentration of very-long-chain fatty acids. We found a defect of the fission of both mitochondria and peroxisomes, as well as a heterozygous, dominant-negative mutation in the dynamin-like protein 1 gene (DLP1). The DLP1 protein has previously been implicated, in vitro, in the fission of both these organelles. Overexpression of the mutant DLP1 in control cells reproduced the fission defect. Our findings are representative of a class of disease characterized by defects in both mitochondria and peroxisomes.


Subject(s)
Abnormalities, Multiple/genetics , GTP Phosphohydrolases/genetics , Microtubule-Associated Proteins/genetics , Mitochondria/physiology , Mitochondrial Proteins/genetics , Mutation , Peroxisomes/physiology , Acidosis, Lactic/genetics , Dynamins , Eye Abnormalities/genetics , Fatal Outcome , Fatty Acids/blood , Female , Fibroblasts/metabolism , Fibroblasts/pathology , GTP Phosphohydrolases/metabolism , Humans , Infant, Newborn , Male , Microcephaly/genetics , Microtubule-Associated Proteins/metabolism , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Muscle Hypotonia/genetics , Open Reading Frames/genetics , Peroxisomes/metabolism , Sequence Analysis, DNA
8.
Ann Neurol ; 59(1): 92-104, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16278854

ABSTRACT

OBJECTIVE: D-bifunctional protein deficiency is an autosomal recessive inborn error of peroxisomal fatty acid oxidation. Although case reports and small series of patients have been published, these do not give a complete and balanced picture of the clinical and biochemical spectrum associated with this disorder. METHODS: To improve early recognition, diagnosis, prognosis, and management of this disorder and to provide markers for life expectancy, we performed extensive biochemical studies in a large cohort of D-bifunctional protein-deficient patients and sent out questionnaires about clinical signs and symptoms to the responsible physicians. RESULTS: Virtually all children presented with neonatal hypotonia and seizures and died within the first 2 years of life without achieving any developmental milestones. However, within our cohort, 12 patients survived beyond the age of 2 years, and detailed information on 5 patients with prolonged survival (> or =7.5 years) is provided. INTERPRETATION: Biochemical analyses showed that there is a clear correlation between several biochemical parameters and survival of the patient, with C26:0 beta-oxidation activity in cultured skin fibroblasts being the best predictive marker for life expectancy. Remarkably, three patients were identified without biochemical abnormalities in plasma, stressing that D-bifunctional protein deficiency cannot be excluded when all peroxisomal parameters in plasma are normal.


Subject(s)
3-Hydroxyacyl CoA Dehydrogenases/deficiency , Enoyl-CoA Hydratase/deficiency , Isomerases/deficiency , Lipid Metabolism, Inborn Errors , Multienzyme Complexes/deficiency , Peroxisomal Disorders , Blood Chemical Analysis , Bone and Bones/anatomy & histology , Bone and Bones/pathology , Brain/anatomy & histology , Brain/pathology , Child , Child, Preschool , Cohort Studies , Fibroblasts/cytology , Fibroblasts/metabolism , Humans , Infant , Kidney/anatomy & histology , Kidney/pathology , Life Expectancy , Liver/anatomy & histology , Liver/pathology , Magnetic Resonance Imaging , Peroxisomal Bifunctional Enzyme , Peroxisomal Disorders/classification , Peroxisomal Disorders/pathology , Peroxisomal Disorders/physiopathology , Surveys and Questionnaires
9.
Hum Mol Genet ; 14(23): 3565-77, 2005 Dec 01.
Article in English | MEDLINE | ID: mdl-16223892

ABSTRACT

ATP-binding cassette (ABC) transporters facilitate unidirectional translocation of chemically diverse substances, ranging from peptides to lipids, across cell or organelle membranes. In peroxisomes, a subfamily of four ABC transporters (ABCD1 to ABCD4) has been related to fatty acid transport, because patients with mutations in ABCD1 (ALD gene) suffer from X-linked adrenoleukodystrophy (X-ALD), a disease characterized by an accumulation of very-long-chain fatty acids (VLCFAs). Inactivation in the mouse of the abcd1 gene leads to a late-onset neurodegenerative condition, comparable to the late-onset form of X-ALD [Pujol, A., Hindelang, C., Callizot, N., Bartsch, U., Schachner, M. and Mandel, J.L. (2002) Late onset neurological phenotype of the X-ALD gene inactivation in mice: a mouse model for adrenomyeloneuropathy. Hum. Mol. Genet., 11, 499-505.]. In the present work, we have generated and characterized a mouse deficient for abcd2, the closest paralog to abcd1. The main pathological feature in abcd2-/- mice is a late-onset cerebellar and sensory ataxia, with loss of cerebellar Purkinje cells and dorsal root ganglia cell degeneration, correlating with accumulation of VLCFAs in the latter cellular population. Axonal degeneration was present in dorsal and ventral columns in spinal cord. We have identified mitochondrial, Golgi and endoplasmic reticulum damage as the underlying pathological mechanism, thus providing evidence of a disturbed organelle cross-talk, which may be at the origin of the pathological cascade.


Subject(s)
ATP-Binding Cassette Transporters/genetics , Spinocerebellar Degenerations/genetics , ATP Binding Cassette Transporter, Subfamily D , Animals , Behavior, Animal , Cerebellum/pathology , Disease Models, Animal , Endoplasmic Reticulum/pathology , Golgi Apparatus/pathology , Mice , Mice, Knockout , Mitochondria/pathology , Spinal Cord/pathology , Spinal Cord/physiopathology , Spinocerebellar Degenerations/pathology , Spinocerebellar Degenerations/physiopathology
10.
Mol Genet Metab ; 84(2): 144-51, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15670720

ABSTRACT

X-linked adrenoleukodystrophy (X-ALD) is a progressive neurodegenerative disorder characterized by the accumulation of saturated and mono-unsaturated very long-chain fatty acids (VLCFA) and reduced peroxisomal VLCFA beta-oxidation activity. In this study, we investigated the role of VLCFA biosynthesis in X-ALD fibroblasts. Our data demonstrate that elongation of both saturated and mono-unsaturated VLCFAs is enhanced in fibroblasts from patients with peroxisomal beta-oxidation defects including X-ALD, and peroxisome biogenesis disorders. These data indicate that enhanced VLCFA elongation is a general phenomenon associated with an impairment in peroxisomal beta-oxidation, and not specific for X-ALD alone. Analysis of plasma samples from patients with X-ALD and different peroxisomal beta-oxidation deficiencies revealed increased concentrations of VLCFAs up to 32 carbons. We infer that enhanced elongation does not result from impaired peroxisomal beta-oxidation alone, but is due to the additional effect of unchecked chain elongation. We demonstrate that elongated VLCFAs are incorporated into complex lipids. The role of chain elongation was also studied retrospectively in samples from patients with X-ALD previously treated with "Lorenzo's oil." We found that the decrease in plasma C26:0 previously found is offset by the increase of mono-unsaturated VLCFAs, not measured previously during the trial. We conclude that evaluation of treatment protocols for disorders of peroxisomal beta-oxidation making use of plasma samples should include the measurement of saturated and unsaturated VLCFAs of chain lengths above 26 carbon atoms. We also conclude that chain elongation offers an interesting target to be studied as a possible mode of treatment for X-ALD and other peroxisomal beta-oxidation disorders.


Subject(s)
Adrenoleukodystrophy/metabolism , Fatty Acids/metabolism , Adrenoleukodystrophy/drug therapy , Drug Combinations , Erucic Acids/therapeutic use , Fatty Acids/biosynthesis , Humans , Oxidation-Reduction , Peroxisomes/metabolism , Triolein/therapeutic use
12.
Hum Mol Genet ; 12(18): 2255-67, 2003 Sep 15.
Article in English | MEDLINE | ID: mdl-12915479

ABSTRACT

Rhizomelic chondrodysplasia punctata is a human autosomal recessive disorder characterized by skeletal, eye and brain abnormalities. The disorder is caused by mutations in the PEX7 gene, which encodes the receptor for a class of peroxisomal matrix enzymes. We describe the generation and characterization of a Pex7 mouse knockout (Pex7(-/-)). Pex7(-/-) mice are born severely hypotonic and have a growth impairment. Mortality in Pex7(-/-) mice is highest in the perinatal period although some Pex7(-/-) mice survived beyond 18 months. Biochemically Pex7(-/-) mice display the abnormalities related to a Pex7 deficiency, i.e. a severe depletion of plasmalogens, impaired alpha-oxidation of phytanic acid and impaired beta-oxidation of very-long-chain fatty acids. In the intermediate zone of the developing cerebral cortex Pex7(-/-) mice have an increase in neuronal density. In vivo neuronal birthdating revealed that Pex7(-/-) mice have a delay in neuronal migration. Analysis of bone ossification in newborn Pex7(-/-) mice revealed a defect in ossification of distal bone elements of the limbs as well as parts of the skull and vertebrae. These findings demonstrate that Pex7 knockout mice provide an important model to study the role of peroxisomal functioning in the pathogenesis of the human disorder.


Subject(s)
Cell Movement , Chondrodysplasia Punctata, Rhizomelic/genetics , Neurons/metabolism , Osteogenesis , Receptors, Cytoplasmic and Nuclear/genetics , Acetyl-CoA C-Acetyltransferase/biosynthesis , Animals , Animals, Newborn , Brain/enzymology , Cells, Cultured , Chondrodysplasia Punctata, Rhizomelic/mortality , Chondrodysplasia Punctata, Rhizomelic/pathology , Diet , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Targeting , Liver/enzymology , Mice , Mice, Knockout , Mutation , Osteoblasts/ultrastructure , Osteoclasts/ultrastructure , Oxidation-Reduction , Peroxisomal Targeting Signal 2 Receptor , Peroxisomes/metabolism , Phytanic Acid/metabolism , Phytol/metabolism , Plasmalogens/biosynthesis , Receptors, Cytoplasmic and Nuclear/deficiency , Time Factors
13.
Am J Hum Genet ; 72(4): 1013-7, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12618959

ABSTRACT

Hydrops-ectopic calcification-"moth-eaten" (HEM) or Greenberg skeletal dysplasia is an autosomal recessive chondrodystrophy with a lethal course, characterized by fetal hydrops, short limbs, and abnormal chondro-osseous calcification. We found elevated levels of cholesta-8,14-dien-3beta-ol in cultured skin fibroblasts of an 18-wk-old fetus with HEM, compatible with a deficiency of the cholesterol biosynthetic enzyme 3beta-hydroxysterol delta(14)-reductase. Sequence analysis of two candidate genes encoding putative human sterol delta(14)-reductases (TM7SF2 and LBR) identified a homozygous 1599-1605TCTTCTA-->CTAGAAG substitution in exon 13 of the LBR gene encoding the lamin B receptor, which results in a truncated protein. Functional complementation of the HEM cells by transfection with control LBR cDNA confirmed that LBR encoded the defective sterol delta(14)-reductase. Mutations in LBR recently have been reported also to cause Pelger-Huët anomaly, an autosomal dominant trait characterized by hypolobulated nuclei and abnormal chromatin structure in granulocytes. The fact that the healthy mother of the fetus showed hypolobulated nuclei in 60% of her granulocytes confirms that classic Pelger-Huët anomaly represents the heterozygous state of 3beta-hydroxysterol delta(14)-reductase deficiency.


Subject(s)
Bone Diseases/genetics , Genes, Recessive , Mutation , Oxidoreductases/genetics , Receptors, Cytoplasmic and Nuclear/genetics , Base Sequence , Bone Diseases/pathology , Cholesterol/biosynthesis , DNA Primers , Humans , Molecular Sequence Data , Oxidoreductases/deficiency , Sequence Analysis, DNA , Lamin B Receptor
SELECTION OF CITATIONS
SEARCH DETAIL
...