Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
J Inherit Metab Dis ; 43(6): 1321-1332, 2020 11.
Article in English | MEDLINE | ID: mdl-32588908

ABSTRACT

We investigated seven children from six families to expand the phenotypic spectrum associated with an early infantile epileptic encephalopathy caused by biallelic pathogenic variants in the phosphatidylinositol glycan anchor biosynthesis class Q (PIGQ) gene. The affected children were all identified by clinical or research exome sequencing. Clinical data, including EEGs and MRIs, was comprehensively reviewed and flow cytometry and transfection experiments were performed to investigate PIGQ function. Pathogenic biallelic PIGQ variants were associated with increased mortality. Epileptic seizures, axial hypotonia, developmental delay and multiple congenital anomalies were consistently observed. Seizure onset occurred between 2.5 months and 7 months of age and varied from treatable seizures to recurrent episodes of status epilepticus. Gastrointestinal issues were common and severe, two affected individuals had midgut volvulus requiring surgical correction. Cardiac anomalies including arrythmias were observed. Flow cytometry using granulocytes and fibroblasts from affected individuals showed reduced expression of glycosylphosphatidylinositol (GPI)-anchored proteins. Transfection of wildtype PIGQ cDNA into patient fibroblasts rescued this phenotype. We expand the phenotypic spectrum of PIGQ-related disease and provide the first functional evidence in human cells of defective GPI-anchoring due to pathogenic variants in PIGQ.


Subject(s)
Abnormalities, Multiple/genetics , Membrane Proteins/genetics , Muscle Hypotonia/genetics , Seizures/genetics , Spasms, Infantile/genetics , Abnormalities, Multiple/diagnosis , Abnormalities, Multiple/metabolism , Child , Child, Preschool , Fatal Outcome , Female , Humans , Infant , Infant, Newborn , Male , Muscle Hypotonia/pathology , Mutation, Missense , Phenotype , Seizures/diagnosis , Seizures/metabolism , Spasms, Infantile/metabolism , Spasms, Infantile/pathology , Exome Sequencing
2.
Am J Hum Genet ; 106(4): 484-495, 2020 04 02.
Article in English | MEDLINE | ID: mdl-32220290

ABSTRACT

Glycosylphosphatidylinositol (GPI)-anchored proteins are critical for embryogenesis, neurogenesis, and cell signaling. Variants in several genes participating in GPI biosynthesis and processing lead to decreased cell surface presence of GPI-anchored proteins (GPI-APs) and cause inherited GPI deficiency disorders (IGDs). In this report, we describe 12 individuals from nine unrelated families with 10 different bi-allelic PIGK variants. PIGK encodes a component of the GPI transamidase complex, which attaches the GPI anchor to proteins. Clinical features found in most individuals include global developmental delay and/or intellectual disability, hypotonia, cerebellar ataxia, cerebellar atrophy, and facial dysmorphisms. The majority of the individuals have epilepsy. Two individuals have slightly decreased levels of serum alkaline phosphatase, while eight do not. Flow cytometric analysis of blood and fibroblasts from affected individuals showed decreased cell surface presence of GPI-APs. The overexpression of wild-type (WT) PIGK in fibroblasts rescued the levels of cell surface GPI-APs. In a knockout cell line, transfection with WT PIGK also rescued the GPI-AP levels, but transfection with the two tested mutant variants did not. Our study not only expands the clinical and known genetic spectrum of IGDs, but it also expands the genetic differential diagnosis for cerebellar atrophy. Given the fact that cerebellar atrophy is seen in other IGDs, flow cytometry for GPI-APs should be considered in the work-ups of individuals presenting this feature.


Subject(s)
Acyltransferases/genetics , Cell Adhesion Molecules/genetics , Cerebellar Diseases/genetics , Epilepsy/genetics , Genetic Variation/genetics , Muscle Hypotonia/genetics , Neurodevelopmental Disorders/genetics , Abnormalities, Multiple/genetics , Alleles , Female , Humans , Intellectual Disability/genetics , Male , Nervous System Malformations/genetics , Pedigree , Syndrome
3.
Am J Hum Genet ; 103(4): 602-611, 2018 10 04.
Article in English | MEDLINE | ID: mdl-30269814

ABSTRACT

Inherited GPI deficiencies (IGDs) are a subset of congenital disorders of glycosylation that are increasingly recognized as a result of advances in whole-exome sequencing (WES) and whole-genome sequencing (WGS). IGDs cause a series of overlapping phenotypes consisting of seizures, dysmorphic features, multiple congenital malformations, and severe intellectual disability. We present a study of six individuals from three unrelated families in which WES or WGS identified bi-allelic phosphatidylinositol glycan class S (PIGS) biosynthesis mutations. Phenotypes included severe global developmental delay, seizures (partly responding to pyridoxine), hypotonia, weakness, ataxia, and dysmorphic facial features. Two of them had compound-heterozygous variants c.108G>A (p.Trp36∗) and c.101T>C (p.Leu34Pro), and two siblings of another family were homozygous for a deletion and insertion leading to p.Thr439_Lys451delinsArgLeuLeu. The third family had two fetuses with multiple joint contractures consistent with fetal akinesia. They were compound heterozygous for c.923A>G (p.Glu308Gly) and c.468+1G>C, a splicing mutation. Flow-cytometry analyses demonstrated that the individuals with PIGS mutations show a GPI-AP deficiency profile. Expression of the p.Trp36∗ variant in PIGS-deficient HEK293 cells revealed only partial restoration of cell-surface GPI-APs. In terms of both biochemistry and phenotype, loss of function of PIGS shares features with PIGT deficiency and other IGDs. This study contributes to the understanding of the GPI-AP biosynthesis pathway by describing the consequences of PIGS disruption in humans and extending the family of IGDs.


Subject(s)
Abnormalities, Multiple/genetics , Acyltransferases/genetics , Arthrogryposis/genetics , Cerebellar Ataxia/genetics , Epilepsy, Generalized/genetics , Cell Line , Child , Child, Preschool , Developmental Disabilities/genetics , Female , HEK293 Cells , Humans , Intellectual Disability/genetics , Male , Muscle Hypotonia/genetics , Mutation , Nervous System Malformations/genetics , Pedigree , Seizures/genetics , Syndrome , Exome Sequencing/methods
4.
Am J Hum Genet ; 101(5): 856-865, 2017 Nov 02.
Article in English | MEDLINE | ID: mdl-29100095

ABSTRACT

Approximately one in every 200 mammalian proteins is anchored to the cell membrane through a glycosylphosphatidylinositol (GPI) anchor. These proteins play important roles notably in neurological development and function. To date, more than 20 genes have been implicated in the biogenesis of GPI-anchored proteins. GPAA1 (glycosylphosphatidylinositol anchor attachment 1) is an essential component of the transamidase complex along with PIGK, PIGS, PIGT, and PIGU (phosphatidylinositol-glycan biosynthesis classes K, S, T, and U, respectively). This complex orchestrates the attachment of the GPI anchor to the C terminus of precursor proteins in the endoplasmic reticulum. Here, we report bi-allelic mutations in GPAA1 in ten individuals from five families. Using whole-exome sequencing, we identified two frameshift mutations (c.981_993del [p.Gln327Hisfs∗102] and c.920delG [p.Gly307Alafs∗11]), one intronic splicing mutation (c.1164+5C>T), and six missense mutations (c.152C>T [p.Ser51Leu], c.160_161delinsAA [p.Ala54Asn], c.527G>C [p.Trp176Ser], c.869T>C [p.Leu290Pro], c.872T>C [p.Leu291Pro], and c.1165G>C [p.Ala389Pro]). Most individuals presented with global developmental delay, hypotonia, early-onset seizures, cerebellar atrophy, and osteopenia. The splicing mutation was found to decrease GPAA1 mRNA. Moreover, flow-cytometry analysis of five available individual samples showed that several GPI-anchored proteins had decreased cell-surface abundance in leukocytes (FLAER, CD16, and CD59) or fibroblasts (CD73 and CD109). Transduction of fibroblasts with a lentivirus encoding the wild-type protein partially rescued the deficiency of GPI-anchored proteins. These findings highlight the role of the transamidase complex in the development and function of the cerebellum and the skeletal system.


Subject(s)
Acyltransferases/genetics , Atrophy/genetics , Bone Diseases, Metabolic/genetics , Developmental Disabilities/genetics , Epilepsy/genetics , Membrane Glycoproteins/genetics , Mutation/genetics , Adolescent , Adult , Alleles , Cerebellum/pathology , Child , Child, Preschool , Exome/genetics , Female , Fibroblasts/pathology , Glycosylphosphatidylinositols/genetics , Humans , Male , Muscle Hypotonia/genetics , Pedigree , RNA, Messenger/genetics , Seizures/genetics
5.
Hum Mol Genet ; 26(9): 1706-1715, 2017 05 01.
Article in English | MEDLINE | ID: mdl-28334793

ABSTRACT

There are over 150 known human proteins which are tethered to the cell surface via glycosylphosphatidylinositol (GPI) anchors. These proteins play a variety of important roles in development, and particularly in neurogenesis. Not surprisingly, mutations in the GPI anchor biosynthesis and remodeling pathway cause a number of developmental disorders. This group of conditions has been termed inherited GPI deficiencies (IGDs), a subgroup of congenital disorders of glycosylation; they present with variable phenotypes, often including seizures, hypotonia and intellectual disability. Here, we report two siblings with compound heterozygous variants in the gene phosphatidylinositol glycan anchor biosynthesis, class P (PIGP) (NM_153681.2: c.74T > C;p.Met25Thr and c.456delA;p.Glu153AsnFs*34). PIGP encodes a subunit of the enzyme that catalyzes the first step of GPI anchor biosynthesis. Both children presented with early-onset refractory seizures, hypotonia, and profound global developmental delay, reminiscent of other IGD phenotypes. Functional studies with patient cells showed reduced PIGP mRNA levels, and an associated reduction of GPI-anchored cell surface proteins, which was rescued by exogenous expression of wild-type PIGP. This work associates mutations in the PIGP gene with a novel autosomal recessive IGD, and expands our knowledge of the role of PIG genes in human development.


Subject(s)
Hexosyltransferases/genetics , Membrane Proteins/genetics , Spasms, Infantile/genetics , Abnormalities, Multiple/genetics , Adult , Cell Line , Child , Developmental Disabilities/genetics , Glycosylphosphatidylinositols/deficiency , Glycosylphosphatidylinositols/genetics , Glycosylphosphatidylinositols/metabolism , Hemoglobinuria, Paroxysmal/genetics , Hexosyltransferases/metabolism , Humans , Intellectual Disability/genetics , Membrane Proteins/metabolism , Muscle Hypotonia/genetics , Mutation , Pedigree , Seizures/genetics , Spasms, Infantile/metabolism
6.
J Med Genet ; 54(3): 196-201, 2017 03.
Article in English | MEDLINE | ID: mdl-27694521

ABSTRACT

BACKGROUND: Of our 1400 exome-studied patients, 67% originate from consanguineous families. ∼80% suffer from variable degree of intellectual disability (ID). The search for disease causing genes using homozygosity mapping was progressing slowly until 2010, then markedly accelerated by the introduction of exome analysis. OBJECTIVES: To identify the disease causing mutation(s) in three patients from two unrelated families who suffered from global developmental delay, severe ID and drug-responsive seizure disorder. METHODS: Exome analysis was performed in DNA of the three patients. The identified PIGC variants were generated and transfected into PIGC-defective mouse cells and the restoration of the surface expression of mouse CD90, CD48 and FLAER was assessed using flow cytometry. The expression of these proteins was also studied on the surface of patients' leucocytes. RESULTS: Three PIGC mutations were identified; homozygous p.L189W in one family and compound heterozygosity for p.L212P/p.R21X variants in another. PIGC participates in the biosynthesis of the glycosylphosphatidylinositol (GPI) anchor which tethers proteins to plasma membrane. In cells lacking PIGC protein, which were transfected with each of the PIGC variants, we detected a clear reduction of surface expression of GPI-anchored proteins. Furthermore, analyses of patients' leucocytes showed significant and constant decrease of CD16 surface expression in granulocytes, and moderate decrease of CD14, CD55, CD59 and FLAER levels. CONCLUSIONS: PIGC joins the list of genes in which mutations result in defective biosynthesis of GPI anchoring, manifesting by global developmental delay and seizure disorder. The lack of specific biomarker dictates exome sequencing as the diagnostic procedure of choice in similar patients.


Subject(s)
Epilepsy/genetics , Hexosyltransferases/genetics , Intellectual Disability/genetics , Membrane Proteins/genetics , Abnormalities, Multiple , Amino Acid Sequence/genetics , Animals , Developmental Disabilities , Epilepsy/physiopathology , Exome/genetics , Female , Genetic Association Studies , Homozygote , Humans , Intellectual Disability/physiopathology , Male , Mice , Mutation , Pedigree
7.
BMC Infect Dis ; 13: 24, 2013 Jan 22.
Article in English | MEDLINE | ID: mdl-23336346

ABSTRACT

BACKGROUND: Reports of acquired immunodeficiency due to autoantibodies against interferon gamma in the adult population are increasing. The interleukin-12-dependent interferon-gamma axis is a major regulatory pathway of cell-mediated immunity and is critical for protection against a few intracellular organisms, including non-tuberculous mycobacteria and Salmonella spp. We report the first case of a fatal disseminated Mycobacterium colombiense/cytomegalovirus coinfection in an adult woman associated with the acquisition of autoantibodies against interferon-gamma. CASE PRESENTATION: A 49-year-old woman, born to nonconsanguineous parents in Laos, but who had lived in Canada for the past 30 years, presented with a 1-month history of weight loss, fatigue, cough, and intermittent low-grade fever. A thoracic computed tomography scan revealed an 8 × 7 cm irregular mass impacting the right superior lobar bronchus along with multiple mediastinal and hilar adenopathies. On the fourth day of admission, the patient developed fever with purulent expectorations. Treatment for a post-obstructive bacterial pneumonia was initiated while other investigations were being pursued. Almost every culture performed during the patient's hospitalization was positive for M. colombiense. Given the late presentation of symptoms - at the age of 49 years - and the absence of significant family or personal medical history, we suspected an acquired immunodeficiency due to the presence of anti-interferon-gamma autoantibodies. This was confirmed by their detection at high levels in the plasma and a STAT1 phosphorylation assay on human monocytes. The final diagnosis was immunodeficiency secondary to the production of autoantibodies against interferon-gamma, which resulted in a post-obstructive pneumonia and disseminated infection of M. colombiense. The clinical course was complicated by the presence of a multiresistant Pseudomonas aeruginosa post-endobronchial ultrasound mediastinitis, cytomegalovirus pneumonitis with dissemination, and finally, susceptible P. aeruginosa ventilator-associated pneumonia with septic shock and multiple organ failure, leading to death despite appropriate antibacterial and anti-mycobacterial treatment. CONCLUSIONS: Although rare, acquired immunodeficiency syndromes should be considered in the differential diagnosis of patients with severe, persistent, or recurrent infections. Specifically, severe non-tuberculous mycobacteria or Salmonella infections in adults without any other known risk factors may warrant examination of autoantibodies against interferon-gamma because of their increasing recognition in the literature.


Subject(s)
Autoantibodies/immunology , Coinfection , Cytomegalovirus Infections/etiology , Immunologic Deficiency Syndromes/complications , Immunologic Deficiency Syndromes/immunology , Interferon-gamma/immunology , Mycobacterium Infections/etiology , Fatal Outcome , Female , Humans , Middle Aged
8.
Genes Dev ; 24(17): 1914-26, 2010 Sep 01.
Article in English | MEDLINE | ID: mdl-20713510

ABSTRACT

Pre-mRNA processing is coupled with transcription. It is still unclear if the transcription machinery can also directly affect the cytoplasmic fate of a transcript, such as its intracellular localization. In yeast, the RNA-binding protein She2p binds several mRNAs and targets them for localization at the bud. Here we report that She2p is recruited cotranscriptionally to the nascent bud-localized ASH1, IST2, and EAR1 mRNA. She2p interacts in vivo with the elongating forms of RNA polymerase II (pol II) via the transcription elongation factor Spt4-Spt5. Mutations in either SPT4 or SPT5 reduce the cotranscriptional recruitment of She2p on the ASH1 gene, disrupt the proper localization of ASH1 mRNA at the bud tip, and affect Ash1p sorting to the daughter cell nucleus. We propose that She2p is recruited by the RNA pol II machinery prior to its transfer to nascent bud-localized mRNAs. Indeed, She2p is present with RNA pol II on genes coding for localized or nonlocalized transcripts, but is associated with nascent mRNA only on genes coding for bud-localized transcripts. Moreover, a She2p mutant defective in RNA binding still associates with RNA pol II transcribed genes. This study uncovers a novel mechanism for the cotranscriptional assembly of mRNP complexes primed for localization in the cytoplasm.


Subject(s)
Chromosomal Proteins, Non-Histone/metabolism , Nuclear Proteins/metabolism , RNA Transport , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/physiology , Transcriptional Elongation Factors/metabolism , Chromatin Immunoprecipitation , Chromosomal Proteins, Non-Histone/genetics , Mutation/genetics , Nuclear Proteins/genetics , Repressor Proteins/genetics , Repressor Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Transcriptional Elongation Factors/genetics
9.
Ann Neurol ; 57(3): 408-14, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15732101

ABSTRACT

Senataxin recently was identified as the mutated gene in ataxia-oculomotor apraxia 2, which is characterized by ataxia, oculomotor apraxia, and increased alpha-fetoprotein levels. In this study, we evaluated 24 ataxic patients from 10 French-Canadian families. All cases have a homogeneous phenotype consisting of a progressive ataxia appearing between 2 and 20 (mean age, 14.8) years of age with associated dysarthria, saccadic ocular pursuit, distal amyotrophy, sensory and motor neuropathy, and increased alpha-fetoprotein levels but absence of oculomotor apraxia. Linkage disequilibrium was observed with markers in the ataxia-oculomotor apraxia 2 locus on chromosome 9q34. We have identified four mutations in senataxin in the French-Canadian population including two novel missense mutations: the 5927T-->G mutation changes the leucine encoded by codon 1976 to an arginine in the helicase domain (L1976R), and the 193G-->A mutation changes a glutamic acid encoded by codon 65 into a lysine in the N-terminal domain of the protein (E65K). The common L1976R mutation is shared by 17 of 20 (85%) carrier chromosomes. The study of this large French-Canadian cohort better defines the phenotype of this ataxia and presents two novel mutations in senataxin including the more common founder mutation in the French-Canadian population.


Subject(s)
Ataxia/genetics , Cluster Analysis , Mutation/genetics , Oculomotor Nerve Diseases/genetics , RNA Helicases/genetics , Adult , Aged , Apraxias/complications , Apraxias/genetics , Arginine/genetics , Ataxia/complications , Chromosomes, Human, Pair 9 , DNA Helicases , DNA Mutational Analysis/methods , Family Health , Female , Glutamic Acid/genetics , Humans , Leucine/genetics , Linkage Disequilibrium , Lysine/genetics , Male , Middle Aged , Multifunctional Enzymes , Oculomotor Nerve Diseases/complications , Quebec , alpha-Fetoproteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...