Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
1.
Genome Res ; 29(7): 1057-1066, 2019 07.
Article in English | MEDLINE | ID: mdl-31160375

ABSTRACT

Germline mutations in fundamental epigenetic regulatory molecules including DNA methyltransferase 3 alpha (DNMT3A) are commonly associated with growth disorders, whereas somatic mutations are often associated with malignancy. We profiled genome-wide DNA methylation patterns in DNMT3A c.2312G > A; p.(Arg771Gln) carriers in a large Amish sibship with Tatton-Brown-Rahman syndrome (TBRS), their mosaic father, and 15 TBRS patients with distinct pathogenic de novo DNMT3A variants. This defined widespread DNA hypomethylation at specific genomic sites enriched at locations annotated as genes involved in morphogenesis, development, differentiation, and malignancy predisposition pathways. TBRS patients also displayed highly accelerated DNA methylation aging. These findings were most marked in a carrier of the AML-associated driver mutation p.Arg882Cys. Our studies additionally defined phenotype-related accelerated and decelerated epigenetic aging in two histone methyltransferase disorders: NSD1 Sotos syndrome overgrowth disorder and KMT2D Kabuki syndrome growth impairment. Together, our findings provide fundamental new insights into aberrant epigenetic mechanisms, the role of epigenetic machinery maintenance, and determinants of biological aging in these growth disorders.


Subject(s)
Aging/genetics , DNA (Cytosine-5-)-Methyltransferases/genetics , Epigenesis, Genetic , Growth Disorders/genetics , Mutation , Abnormalities, Multiple/genetics , Adolescent , Adult , Amish/genetics , Child , DNA Methylation , DNA Methyltransferase 3A , Face/abnormalities , Hematologic Diseases/genetics , Humans , Intellectual Disability/genetics , Leukemia, Myeloid, Acute/genetics , Male , Methyltransferases , Morphogenesis/genetics , Syndrome , Vestibular Diseases/genetics , Young Adult
2.
Brain ; 140(4): 940-952, 2017 Apr 01.
Article in English | MEDLINE | ID: mdl-28334956

ABSTRACT

PRUNE is a member of the DHH (Asp-His-His) phosphoesterase protein superfamily of molecules important for cell motility, and implicated in cancer progression. Here we investigated multiple families from Oman, India, Iran and Italy with individuals affected by a new autosomal recessive neurodevelopmental and degenerative disorder in which the cardinal features include primary microcephaly and profound global developmental delay. Our genetic studies identified biallelic mutations of PRUNE1 as responsible. Our functional assays of disease-associated variant alleles revealed impaired microtubule polymerization, as well as cell migration and proliferation properties, of mutant PRUNE. Additionally, our studies also highlight a potential new role for PRUNE during microtubule polymerization, which is essential for the cytoskeletal rearrangements that occur during cellular division and proliferation. Together these studies define PRUNE as a molecule fundamental for normal human cortical development and define cellular and clinical consequences associated with PRUNE mutation.


Subject(s)
Brain/growth & development , Carrier Proteins/genetics , Developmental Disabilities/genetics , Microcephaly/genetics , Adolescent , Cell Differentiation/genetics , Cell Movement/genetics , Cerebral Cortex/growth & development , Child , Child, Preschool , Cytoskeleton/genetics , Cytoskeleton/ultrastructure , Female , Genes, Recessive , Heredodegenerative Disorders, Nervous System/genetics , Humans , Infant , Male , Microtubules/genetics , Microtubules/ultrastructure , Mutation/genetics , Pedigree , Phosphoric Monoester Hydrolases , Young Adult
3.
PLoS Genet ; 13(1): e1006470, 2017 Jan.
Article in English | MEDLINE | ID: mdl-28081210

ABSTRACT

Orofacial clefting is amongst the most common of birth defects, with both genetic and environmental components. Although numerous studies have been undertaken to investigate the complexities of the genetic etiology of this heterogeneous condition, this factor remains incompletely understood. Here, we describe mutations in the HYAL2 gene as a cause of syndromic orofacial clefting. HYAL2, encoding hyaluronidase 2, degrades extracellular hyaluronan, a critical component of the developing heart and palatal shelf matrix. Transfection assays demonstrated that the gene mutations destabilize the molecule, dramatically reducing HYAL2 protein levels. Consistent with the clinical presentation in affected individuals, investigations of Hyal2-/- mice revealed craniofacial abnormalities, including submucosal cleft palate. In addition, cor triatriatum sinister and hearing loss, identified in a proportion of Hyal2-/- mice, were also found as incompletely penetrant features in affected humans. Taken together our findings identify a new genetic cause of orofacial clefting in humans and mice, and define the first molecular cause of human cor triatriatum sinister, illustrating the fundamental importance of HYAL2 and hyaluronan turnover for normal human and mouse development.


Subject(s)
Cell Adhesion Molecules/genetics , Cleft Lip/genetics , Cleft Palate/genetics , Cor Triatriatum/genetics , Hyaluronoglucosaminidase/genetics , Mutation , Adolescent , Animals , Child , Child, Preschool , Cleft Lip/pathology , Cleft Palate/pathology , Cor Triatriatum/pathology , Female , GPI-Linked Proteins/genetics , Humans , Male , Mice , Mice, Inbred C57BL , Pedigree , Penetrance , Syndrome
4.
Brain ; 140(3): 547-554, 2017 03 01.
Article in English | MEDLINE | ID: mdl-28052917

ABSTRACT

Mutations in genes involved in lipid metabolism have increasingly been associated with various subtypes of hereditary spastic paraplegia, a highly heterogeneous group of neurodegenerative motor neuron disorders characterized by spastic paraparesis. Here, we report an unusual autosomal recessive neurodegenerative condition, best classified as a complicated form of hereditary spastic paraplegia, associated with mutation in the ethanolaminephosphotransferase 1 (EPT1) gene (now known as SELENOI), responsible for the final step in Kennedy pathway forming phosphatidylethanolamine from CDP-ethanolamine. Phosphatidylethanolamine is a glycerophospholipid that, together with phosphatidylcholine, constitutes more than half of the total phospholipids in eukaryotic cell membranes. We determined that the mutation defined dramatically reduces the enzymatic activity of EPT1, thereby hindering the final step in phosphatidylethanolamine synthesis. Additionally, due to central nervous system inaccessibility we undertook quantification of phosphatidylethanolamine levels and species in patient and control blood samples as an indication of liver phosphatidylethanolamine biosynthesis. Although this revealed alteration to levels of specific phosphatidylethanolamine fatty acyl species in patients, overall phosphatidylethanolamine levels were broadly unaffected indicating that in blood EPT1 inactivity may be compensated for, in part, via alternate biochemical pathways. These studies define the first human disorder arising due to defective CDP-ethanolamine biosynthesis and provide new insight into the role of Kennedy pathway components in human neurological function.


Subject(s)
Ethanolaminephosphotransferase/genetics , Ethanolaminephosphotransferase/metabolism , Mutation/genetics , Phospholipids/biosynthesis , Signal Transduction/genetics , Spastic Paraplegia, Hereditary/genetics , Adolescent , Child , Child, Preschool , Chromatography, Liquid , Consanguinity , DNA Mutational Analysis , Family Health , Female , Gene Expression , Humans , Infant , Male , Mass Spectrometry , Oman , Phospholipids/blood , Saccharomyces cerevisiae , Spastic Paraplegia, Hereditary/diagnostic imaging , Spastic Paraplegia, Hereditary/enzymology , Spastic Paraplegia, Hereditary/pathology
5.
Hum Mutat ; 37(11): 1157-1161, 2016 11.
Article in English | MEDLINE | ID: mdl-27492651

ABSTRACT

Hereditary spastic paraplegias (HSPs) are genetically and clinically heterogeneous axonopathies primarily affecting upper motor neurons and, in complex forms, additional neurons. Here, we report two families with distinct recessive mutations in TFG, previously suggested to cause HSP based on findings in a single small family with complex HSP. The first carried a homozygous c.317G>A (p.R106H) variant and presented with pure HSP. The second carried the same homozygous c.316C>T (p.R106C) variant previously reported and displayed a similarly complex phenotype including optic atrophy. Haplotyping and bisulfate sequencing revealed evidence for a c.316C>T founder allele, as well as for a c.316_317 mutation hotspot. Expression of mutant TFG proteins in cultured neurons revealed mitochondrial fragmentation, the extent of which correlated with clinical severity. Our findings confirm the causal nature of bi-allelic TFG mutations for HSP, broaden the clinical and mutational spectra, and suggest mitochondrial impairment to represent a pathomechanistic link to other neurodegenerative conditions.


Subject(s)
Mutation, Missense , Proteins/genetics , Proteins/metabolism , Spastic Paraplegia, Hereditary/pathology , Animals , Cells, Cultured , Female , Genetic Predisposition to Disease , Humans , Magnetic Resonance Imaging/methods , Male , Mice , Mitochondria/pathology , Neurons/cytology , Neurons/metabolism , Neurons/pathology , Pedigree , Sequence Analysis, DNA , Spastic Paraplegia, Hereditary/genetics , Spastic Paraplegia, Hereditary/metabolism
6.
BMC Med Genet ; 16: 104, 2015 Nov 10.
Article in English | MEDLINE | ID: mdl-26554554

ABSTRACT

BACKGROUND: The deletion of the chromosome 4p16.3 Wolf-Hirschhorn syndrome critical region (WHSCR-2) typically results in a characteristic facial appearance, varying intellectual disability, stereotypies and prenatal onset of growth retardation, while gains of the same chromosomal region result in a more variable degree of intellectual deficit and dysmorphism. Similarly the phenotype of individuals with terminal deletions of distal chromosome 3p (3p deletion syndrome) varies from mild to severe intellectual deficit, micro- and trigonocephaly, and a distinct facial appearance. METHODS AND RESULTS: We investigated a large Indian five-generation pedigree with ten affected family members in which chromosomal microarray and fluorescence in situ hybridization analyses disclosed a complex rearrangement involving chromosomal subregions 4p16.1 and 3p26.3 resulting in a 4p16.1 deletion and 3p26.3 microduplication in three individuals, and a 4p16.1 duplication and 3p26.3 microdeletion in seven individuals. A typical clinical presentation of WHS was observed in all three cases with 4p16.1 deletion and 3p26.3 microduplication. Individuals with a 4p16.1 duplication and 3p26.3 microdeletion demonstrated a range of clinical features including typical 3p microdeletion or 4p partial trisomy syndrome to more severe neurodevelopmental delay with distinct dysmorphic features. CONCLUSION: We present the largest pedigree with complex t(4p;3p) chromosomal rearrangements and diverse clinical outcomes including Wolf Hirschorn-, 3p deletion-, and 4p duplication syndrome amongst affected individuals.


Subject(s)
Chromosome Disorders/genetics , Chromosomes, Human, Pair 3 , Chromosomes, Human, Pair 4 , Chromosome Disorders/etiology , Developmental Disabilities/genetics , Female , Humans , In Situ Hybridization, Fluorescence , India , Male , Oligonucleotide Array Sequence Analysis , Pedigree , Sequence Deletion , Trisomy , Wolf-Hirschhorn Syndrome/genetics
7.
Nat Genet ; 47(7): 814-7, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26005865

ABSTRACT

The major pathway by which the brain obtains essential omega-3 fatty acids from the circulation is through a sodium-dependent lysophosphatidylcholine (LPC) transporter (MFSD2A), expressed in the endothelium of the blood-brain barrier. Here we show that a homozygous mutation affecting a highly conserved MFSD2A residue (p.Ser339Leu) is associated with a progressive microcephaly syndrome characterized by intellectual disability, spasticity and absent speech. We show that the p.Ser339Leu alteration does not affect protein or cell surface expression but rather significantly reduces, although not completely abolishes, transporter activity. Notably, affected individuals displayed significantly increased plasma concentrations of LPCs containing mono- and polyunsaturated fatty acyl chains, indicative of reduced brain uptake, confirming the specificity of MFSD2A for LPCs having mono- and polyunsaturated fatty acyl chains. Together, these findings indicate an essential role for LPCs in human brain development and function and provide the first description of disease associated with aberrant brain LPC transport in humans.


Subject(s)
Fatty Acids, Omega-3/metabolism , Microcephaly/genetics , Tumor Suppressor Proteins/genetics , Adolescent , Animals , Base Sequence , Biological Transport , Blood-Brain Barrier/metabolism , Case-Control Studies , Child , Child, Preschool , Female , Genetic Association Studies , HEK293 Cells , Humans , Infant , Lysophosphatidylcholines/blood , Male , Microcephaly/blood , Mutation, Missense , Pedigree , Sequence Analysis, DNA , Symporters , Syndrome
8.
Neurology ; 84(17): 1745-50, 2015 Apr 28.
Article in English | MEDLINE | ID: mdl-25832664

ABSTRACT

OBJECTIVE: To identify the genetic cause of pontocerebellar hypoplasia type III (PCH3). METHODS: We studied the original reported pedigree of PCH3 and performed genetic analysis including genome-wide single nucleotide polymorphism genotyping, linkage analysis, whole-exome sequencing, and Sanger sequencing. Human fetal brain RNA sequencing data were then analyzed for the identified candidate gene. RESULTS: The affected individuals presented with severe global developmental delay and seizures starting in the first year of life. Brain MRI of an affected individual showed diffuse atrophy of the cerebrum, cerebellum, and brainstem. Genome-wide single nucleotide polymorphism analysis confirmed the linkage to chromosome 7q we previously reported, and showed no other genomic areas of linkage. Whole-exome sequencing of 2 affected individuals identified a shared homozygous, nonsense variant in the PCLO (piccolo) gene. This variant segregated with the disease phenotype in the pedigree was rare in the population and was predicted to eliminate the PDZ and C2 domains in the C-terminus of the protein. RNA sequencing data of human fetal brain showed that PCLO was moderately expressed in the developing cerebral cortex. CONCLUSIONS: Here, we show that a homozygous, nonsense PCLO mutation underlies the autosomal recessive neurodegenerative disorder, PCH3. PCLO is a component of the presynaptic cytoskeletal matrix, and is thought to be involved in regulation of presynaptic proteins and synaptic vesicles. Our findings suggest that PCLO is crucial for the development and survival of a wide range of neuronal types in the human brain.


Subject(s)
Codon, Nonsense/genetics , Cytoskeletal Proteins/genetics , Neuropeptides/genetics , Cerebellar Diseases/genetics , Cerebellar Diseases/pathology , Cerebellar Diseases/physiopathology , Child , Consanguinity , Exome , Genetic Linkage , Humans , Oman , Pedigree , Polymorphism, Single Nucleotide , Sequence Analysis, RNA
9.
J Clin Invest ; 124(7): 3137-46, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24911150

ABSTRACT

Numerous human disorders, including Cockayne syndrome, UV-sensitive syndrome, xeroderma pigmentosum, and trichothiodystrophy, result from the mutation of genes encoding molecules important for nucleotide excision repair. Here, we describe a syndrome in which the cardinal clinical features include short stature, hearing loss, premature aging, telangiectasia, neurodegeneration, and photosensitivity, resulting from a homozygous missense (p.Ser228Ile) sequence alteration of the proliferating cell nuclear antigen (PCNA). PCNA is a highly conserved sliding clamp protein essential for DNA replication and repair. Due to this fundamental role, mutations in PCNA that profoundly impair protein function would be incompatible with life. Interestingly, while the p.Ser228Ile alteration appeared to have no effect on protein levels or DNA replication, patient cells exhibited marked abnormalities in response to UV irradiation, displaying substantial reductions in both UV survival and RNA synthesis recovery. The p.Ser228Ile change also profoundly altered PCNA's interaction with Flap endonuclease 1 and DNA Ligase 1, DNA metabolism enzymes. Together, our findings detail a mutation of PCNA in humans associated with a neurodegenerative phenotype, displaying clinical and molecular features common to other DNA repair disorders, which we showed to be attributable to a hypomorphic amino acid alteration.


Subject(s)
DNA Repair-Deficiency Disorders/genetics , Mutant Proteins/genetics , Mutation, Missense , Proliferating Cell Nuclear Antigen/genetics , Adolescent , Adult , Aging, Premature/genetics , Amino Acid Substitution , Child , Chromosomes, Human, Pair 20/genetics , DNA Mutational Analysis , DNA Repair-Deficiency Disorders/pathology , DNA Repair-Deficiency Disorders/physiopathology , Dwarfism/genetics , Female , Hearing Loss/genetics , Homozygote , Humans , Male , Models, Molecular , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Nerve Degeneration/genetics , Pedigree , Phenotype , Photosensitivity Disorders/genetics , Proliferating Cell Nuclear Antigen/chemistry , Proliferating Cell Nuclear Antigen/metabolism , Protein Structure, Quaternary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Syndrome , Telangiectasis/genetics
10.
Am J Hum Genet ; 94(1): 87-94, 2014 Jan 02.
Article in English | MEDLINE | ID: mdl-24239382

ABSTRACT

The proper development of neuronal circuits during neuromorphogenesis and neuronal-network formation is critically dependent on a coordinated and intricate series of molecular and cellular cues and responses. Although the cortical actin cytoskeleton is known to play a key role in neuromorphogenesis, relatively little is known about the specific molecules important for this process. Using linkage analysis and whole-exome sequencing on samples from families from the Amish community of Ohio, we have demonstrated that mutations in KPTN, encoding kaptin, cause a syndrome typified by macrocephaly, neurodevelopmental delay, and seizures. Our immunofluorescence analyses in primary neuronal cell cultures showed that endogenous and GFP-tagged kaptin associates with dynamic actin cytoskeletal structures and that this association is lost upon introduction of the identified mutations. Taken together, our studies have identified kaptin alterations responsible for macrocephaly and neurodevelopmental delay and define kaptin as a molecule crucial for normal human neuromorphogenesis.


Subject(s)
Developmental Disabilities/genetics , Megalencephaly/genetics , Microfilament Proteins/genetics , Mutation , Seizures/genetics , Actin Cytoskeleton/metabolism , Amino Acid Sequence , Female , Fluorescent Antibody Technique , Genetic Linkage , Humans , Male , Microfilament Proteins/metabolism , Molecular Sequence Data , Pedigree
11.
J Clin Invest ; 123(5): 2094-102, 2013 May.
Article in English | MEDLINE | ID: mdl-23543054

ABSTRACT

Myopia is by far the most common human eye disorder that is known to have a clear, albeit poorly defined, heritable component. In this study, we describe an autosomal-recessive syndrome characterized by high myopia and sensorineural deafness. Our molecular investigation in 3 families led to the identification of 3 homozygous nonsense mutations (p.R181X, p.S297X, and p.Q414X) in SLIT and NTRK-like family, member 6 (SLITRK6), a leucine-rich repeat domain transmembrane protein. All 3 mutant SLITRK6 proteins displayed defective cell surface localization. High-resolution MRI of WT and Slitrk6-deficient mouse eyes revealed axial length increase in the mutant (the endophenotype of myopia). Additionally, mutant mice exhibited auditory function deficits that mirrored the human phenotype. Histological investigation of WT and Slitrk6-deficient mouse retinas in postnatal development indicated a delay in synaptogenesis in Slitrk6-deficient animals. Taken together, our results showed that SLITRK6 plays a crucial role in the development of normal hearing as well as vision in humans and in mice and that its disruption leads to a syndrome characterized by severe myopia and deafness.


Subject(s)
Hearing Loss, Sensorineural/genetics , Membrane Proteins/genetics , Myopia/genetics , Adolescent , Adult , Animals , Child , Codon, Nonsense , Female , Hearing , Humans , Infant , Male , Mice , Mice, Knockout , Middle Aged , Mutation , Pedigree , Phenotype , Protein Structure, Tertiary , Young Adult
12.
J Med Genet ; 50(2): 65-73, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23243086

ABSTRACT

BACKGROUND: Deregulation of the activity of the ubiquitin ligase E6AP (UBE3A) is well recognised to contribute to the development of Angelman syndrome (AS). The ubiquitin ligase HERC2, encoded by the HERC2 gene is thought to be a key regulator of E6AP. METHODS AND RESULTS: Using a combination of autozygosity mapping and linkage analysis, we studied an autosomal-recessive neurodevelopmental disorder with some phenotypic similarities to AS, found among the Old Order Amish. Our molecular investigation identified a mutation in HERC2 associated with the disease phenotype. We establish that the encoded mutant HERC2 protein has a reduced half-life compared with its wild-type counterpart, which is associated with a significant reduction in HERC2 levels in affected individuals. CONCLUSIONS: Our data implicate a model in which disruption of HERC2 function relates to a reduction in E6AP activity resulting in neurodevelopmental delay, suggesting a previously unrecognised role of HERC2 in the pathogenesis of AS.


Subject(s)
Amish/genetics , Angelman Syndrome/genetics , Guanine Nucleotide Exchange Factors/genetics , Mutation , Adolescent , Adult , Cell Cycle Proteins/chemistry , Cell Line , Child , Child, Preschool , DNA/analysis , DNA/genetics , DNA Mutational Analysis , Female , Fibroblasts/chemistry , Fibroblasts/metabolism , Genome-Wide Association Study , Guanine Nucleotide Exchange Factors/blood , Guanine Nucleotide Exchange Factors/chemistry , Guanine Nucleotide Exchange Factors/metabolism , HEK293 Cells , Humans , Infant , Male , Models, Molecular , Nuclear Proteins/chemistry , Pedigree , Ubiquitin-Protein Ligases
15.
Am J Hum Genet ; 87(5): 655-60, 2010 Nov 12.
Article in English | MEDLINE | ID: mdl-20970105

ABSTRACT

In human mitochondria, polyadenylation of mRNA, undertaken by the nuclear-encoded mitochondrial poly(A) RNA polymerase, is essential for maintaining mitochondrial gene expression. Our molecular investigation of an autosomal-recessive spastic ataxia with optic atrophy, present among the Old Order Amish, identified a mutation of MTPAP associated with the disease phenotype. When subjected to poly(A) tail-length assays, mitochondrial mRNAs from affected individuals were shown to have severely truncated poly(A) tails. Although defective mitochondrial DNA maintenance underlies a well-described group of clinical disorders, our findings reveal a defect of mitochondrial mRNA maturation associated with human disease and imply that this disease mechanism should be considered in other complex neurodegenerative disorders.


Subject(s)
Cerebellar Ataxia/genetics , DNA-Directed RNA Polymerases/genetics , Genes, Mitochondrial , Mitochondrial Proteins/genetics , Paraparesis, Spastic/genetics , RNA, Messenger , Adolescent , Adult , Amino Acid Sequence , Base Sequence , Child , Child, Preschool , Female , Humans , Male , Molecular Sequence Data , Mutation , Optic Atrophy/genetics , Pedigree , RNA, Mitochondrial
16.
Clin Dysmorphol ; 19(4): 198-201, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20577083

ABSTRACT

Hereditary congenital facial paresis is a rare syndrome of isolated facial nerve palsy causing facial asymmetry and ptosis. Most described cases follow an autosomal dominant pattern of inheritance. It differs from Moebius syndrome, which is usually sporadic and associated with the involvement of other cranial nerves, commonly the abducens nerve in addition to orofacial and limb malformations and defects of the musculoskeletal system. We present three patients from the same family with features of congenital hereditary facial paresis. Facial asymmetry and facial weakness were the most remarkable findings. High-resolution imaging showed both facial nerves to be present but symmetrically and markedly hypoplastic with no other structural abnormality in the brainstem. This syndrome has been previously mapped to chromosome 3q21-22 but no gene has been identified as yet.


Subject(s)
Facial Paralysis , Adult , Child, Preschool , Facial Paralysis/congenital , Facial Paralysis/genetics , Facial Paralysis/pathology , Female , Humans , In Situ Hybridization, Fluorescence , Magnetic Resonance Imaging , Male
17.
Hum Mutat ; 31(4): E1251-60, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20104589

ABSTRACT

Hereditary spastic paraplegia (HSP) describes a heterogeneous group of inherited neurodegenerative disorders in which the cardinal pathological feature is upper motor neurone degeneration leading to progressive spasticity and weakness of the lower limbs. Using samples from a large Omani family we recently mapped a gene for a novel autosomal recessive form of HSP (SPG35) in which the spastic paraplegia was associated with intellectual disability and seizures. Magnetic resonance imaging of the brain of SPG35 patients showed white matter abnormalities suggestive of a leukodystrophy. Here we report homozygous mutations in the fatty acid 2-hydroxylase gene (FA2H) in the original family used to define the SPG35 locus (p.Arg235Cys) as well as in a previously unreported Pakistani family with a similar phenotype (p.Arg53_Ile58del). Measurement of enzyme activity in vitro revealed significantly reduced enzymatic function of FA2H associated with these mutations. These results demonstrate that mutations in FA2H are associated with SPG35, and that abnormal hydroxylation of myelin galactocerebroside lipid components can lead to a severe progressive phenotype, with a clinical presentation of complicated HSP and radiological features of leukodystrophy. (c) 2010 Wiley-Liss, Inc.


Subject(s)
Mixed Function Oxygenases/genetics , Mutation/genetics , Spastic Paraplegia, Hereditary/enzymology , Spastic Paraplegia, Hereditary/genetics , Adolescent , Adult , Animals , Brain/pathology , CHO Cells , Child , Child, Preschool , Chromatography, Thin Layer , Consanguinity , Cricetinae , Cricetulus , Female , Humans , Infant , Magnetic Resonance Imaging , Male , Pedigree , Pregnancy , Transfection
18.
Nat Genet ; 42(1): 27-9, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19966803

ABSTRACT

Noonan syndrome, a developmental disorder characterized by congenital heart defects, reduced growth, facial dysmorphism and variable cognitive deficits, is caused by constitutional dysregulation of the RAS-MAPK signaling pathway. Here we report that germline NRAS mutations conferring enhanced stimulus-dependent MAPK activation account for some cases of this disorder. These findings provide evidence for an obligate dependency on proper NRAS function in human development and growth.


Subject(s)
Genes, ras , Mutation , Noonan Syndrome/genetics , ras Proteins/genetics , Adolescent , Amino Acid Sequence , Animals , Base Sequence , COS Cells , Child , Child, Preschool , Chlorocebus aethiops , DNA Mutational Analysis , Female , Humans , Male , Middle Aged , Mitogen-Activated Protein Kinases/metabolism , Models, Molecular , Molecular Sequence Data , Noonan Syndrome/metabolism , Noonan Syndrome/pathology , Phosphorylation , Protein Structure, Tertiary , Sequence Homology, Amino Acid , Transfection , Young Adult , ras Proteins/chemistry
20.
Am J Hum Genet ; 82(2): 510-5, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18252231

ABSTRACT

The hereditary spastic paraplegias (HSPs) are a genetically and clinically heterogeneous group of upper-motor-neuron degenerative diseases characterized by selective axonal loss in the corticospinal tracts and dorsal columns. Although numerous mechanisms involving defective subcellular transportation, mitochondrial malfunction, and increased oxidative stress have been proposed, the pathogenic basis underlying the neuronal loss is unknown. We have performed linkage analysis to refine the extent of the SPG5 disease locus and conducted sequence analysis of the genes located within this region. This identified sequence alterations in the cytochrome P450-7B1 (CYP7B1) associated with this pure form of HSP. In the liver, CYP7B1 offers an alternative pathway for cholesterol degradation and also provides the primary metabolic route for the modification of dehydroepiandrosterone neurosteroids in the brain. These findings provide the first direct evidence of a pivotal role of altered cholesterol metabolism in the pathogenesis of motor-neuron degenerative disease and identify a potential for therapeutic intervention in this form of HSP.


Subject(s)
Cholesterol/metabolism , Cytochrome P-450 Enzyme System/genetics , Homeostasis/genetics , Spastic Paraplegia, Hereditary/genetics , Steroid Hydroxylases/genetics , Amino Acid Sequence , Base Sequence , Chromosome Mapping , Cytochrome P450 Family 7 , Humans , Liver/metabolism , Molecular Sequence Data , Pedigree , Sequence Analysis, DNA , Spastic Paraplegia, Hereditary/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL