Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 196
Filter
1.
Neurol Sci ; 45(5): 2253-2260, 2024 May.
Article in English | MEDLINE | ID: mdl-38055078

ABSTRACT

INTRODUCTION: PIGW-related glycosylphosphatidylinositol deficiency is a rare disease that manifests heterogeneous clinical phenotypes. METHODS: We describe a patient with PIGW deficiency and summarize the clinical characteristics of the case. In addition, we conducted a literature review of previously reported patients with pathogenic variants of PIGW. RESULTS: A Chinese girl presented with refractory epilepsy, severe intellectual disability, recurrent respiratory infections, and hyperphosphatasia. Seizures worsened during fever and infections, making her more susceptible to epileptic status. She was found to carry a heterozygous variant of PIGW and a deletion of chromosome 17q12 containing PIGW. Only six patients with homozygous or compound heterozygous pathogenic variants of PIGW have been identified in the literature thus far. Epileptic seizures were reported in all patients, and the most common types of seizures were epileptic spasms. Distinctive facial and physical features and recurrent respiratory infections are common in these patients with developmental delays. Serum alkaline phosphatase (ALP) levels were elevated in four of the six patients. CONCLUSIONS: PIGW-related glycosylphosphatidylinositol deficiency is characterized by developmental delay, epilepsy, distinctive facial features, and multiple organ anomalies. Genetic testing is an important method for diagnosing this disease, and flow cytometry and serum ALP level detection are crucial complements for genetic testing.


Subject(s)
Abnormalities, Multiple , Epilepsy , Glycosylphosphatidylinositols/deficiency , Intellectual Disability , Respiratory Tract Infections , Humans , Female , Seizures/genetics , Epilepsy/genetics , Epilepsy/diagnosis , Intellectual Disability/complications , Intellectual Disability/genetics , Intellectual Disability/diagnosis , Abnormalities, Multiple/genetics
2.
Nat Commun ; 13(1): 3107, 2022 06 03.
Article in English | MEDLINE | ID: mdl-35661110

ABSTRACT

Inherited glycosylphosphatidylinositol (GPI) deficiency (IGD) is caused by mutations in GPI biosynthesis genes. The mechanisms of its systemic, especially neurological, symptoms are not clarified and fundamental therapy has not been established. Here, we report establishment of mouse models of IGD caused by PIGO mutations as well as development of effective gene therapy. As the clinical manifestations of IGD are systemic and lifelong lasting, we treated the mice with adeno-associated virus for homology-independent knock-in as well as extra-chromosomal expression of Pigo cDNA. Significant amelioration of neuronal phenotypes and growth defect was achieved, opening a new avenue for curing IGDs.


Subject(s)
Glycosylphosphatidylinositols , Seizures , Animals , Disease Models, Animal , Genetic Therapy , Glycosylphosphatidylinositols/deficiency , Glycosylphosphatidylinositols/genetics , Immunoglobulin D/genetics , Mice , Seizures/genetics
3.
BMC Pediatr ; 22(1): 222, 2022 04 25.
Article in English | MEDLINE | ID: mdl-35468813

ABSTRACT

BACKGROUND: Mutations in PIGN, resulting in a glycosylphosphatidylinositol (GPI) anchor deficiency, typically leads to multiple congenital anomalies-hypotonia-seizures syndrome. However, the link between PIGN and epilepsy or paroxysmal non-kinesigenic dyskinesia (PNKD) is not well-described. This study reported a patient with PIGN mutation leading to developmental and epileptic encephalopathy and PNKD, to expand upon the genotype-phenotype correlation of PIGN. CASE PRESENTATION: During the first 10 days of life, a girl exhibited paroxysmal staring episodes with durations that ranged from several minutes to hours. These episodes occurred 2-5 times daily and always occurred during wakefulness. Ictal electroencephalography revealed no abnormalities, and PNKD was diagnosed. The patient also exhibited severely delayed psychomotor development and generalized seizures at the age of 4 months. Results of brain magnetic resonance imaging and metabolic screenings were normal, but trio-based whole-exome sequencing identified two novel compound heterozygous PIGN mutations (NM_176787; c.163C > T [p.R55 > X] and c.283C > T [p.R95W]). Flow cytometry analysis of the patient's granulocytes revealed dramatically reduced expression of GPI-anchored proteins. This indicated that the mutations compromised GPI functions. The patient got seizure-free for 1 year, and her dyskinesia episodes reduced significantly (1-2 times/month) after treatment with levetiracetam (600 mg/day) and clonazepam (1.5 mg/day). No progress was observed with respect to psychomotor development; however, no craniofacial dysmorphic features, cleft lip/palate, brachytelephalangy with nail hypoplasia, and internal malformations have been observed until now (6 years of age). CONCLUSION: This is the first study to document developmental and epileptic encephalopathy with PNKD in a human with PIGN mutations. This report expanded our understanding of the genotype-phenotype correlation of PIGN, and PIGN may be considered a potentially relevant gene when investigating cases of epilepsy or PNKD.


Subject(s)
Cleft Lip , Cleft Palate , Dyskinesias , Epilepsy , Epilepsy/drug therapy , Epilepsy/genetics , Female , Glycosylphosphatidylinositols/deficiency , Humans , Mutation , Phosphotransferases/genetics , Seizures
4.
Expert Rev Hematol ; 15(2): 175-181, 2022 02.
Article in English | MEDLINE | ID: mdl-35119350

ABSTRACT

OBJECTIVES: We examined the frequencies and sizes of glycosylphosphatidylinositol (GPI)-deficient cells as per the International Clinical Cytometry Society/European Society for Clinical Cell Analysis (ICCS/ESCCA) consensus guidelines for the high-sensitivity detection of GPI-deficient cells. METHODS: In 2018, the ICCS/ESCCA guidelines for the high-sensitivity detection of GPI-deficient cells were published. We evaluated frequencies and sizes of GPI-deficient red blood cells (RBCs), neutrophils, and monocytes as determined using the ICCS/ESCCA guidelines and Clinical and Laboratory Standards Institute (CLSI) guidelines in patients with a hematologic malignancy, aplastic anemia, or cytopenia. RESULTS: A total of 106 (38.7%) patients exhibited GPI deficiency in at least one blood cell lineage. GPI-deficient cells of one or more lineages were found in 62.7% of patients with a hematologic malignancy, 51.1% of patients with aplastic anemia, and 23.4% of patients with cytopenia. GPI-deficient monocytes were most frequently detected in all three groups. By population size, GPI-deficient clones (>1%) in monocytes were mostly detected in patients with a hematologic malignancy or aplastic anemia. Rare cells with GPI deficiency (<0.1%) in monocytes were most common among patients with cytopenia. CONCLUSION: High-sensitive flow cytometry analysis including monocytes may be necessary for patients with a hematologic disorder.


Subject(s)
Anemia, Aplastic , Hematologic Neoplasms , Hemoglobinuria, Paroxysmal , Thrombocytopenia , Anemia, Aplastic/diagnosis , Anemia, Aplastic/genetics , Flow Cytometry , Glycosylphosphatidylinositols/deficiency , Hemoglobinuria, Paroxysmal/diagnosis , Hemoglobinuria, Paroxysmal/genetics , Humans , Seizures
5.
Dev Med Child Neurol ; 64(6): 789-798, 2022 06.
Article in English | MEDLINE | ID: mdl-35080266

ABSTRACT

AIM: To investigate the short-term efficacy and safety of high-dose pyridoxine and pyridoxal 5-phosphate (P5P) in the treatment of inherited glycosylphosphatidylinositol (GPI) deficiency-associated epilepsy. METHOD: Participants with genetically confirmed GPI deficiency were treated with oral pyridoxine or P5P as compassionate use in an agreed-upon clinical regimen. Pyridoxine (20-30 mg/kg/day) was used for 3 months. Baseline evaluation included 4 weeks of prospective seizure data and one video electroencephalogram (EEG). Seizure frequency was captured daily. The EEG was repeated after reaching maximum dosage of pyridoxine. Pyridoxine was switched to P5P (20-30 mg/kg/day) if seizure burden was unchanged after 3 months' treatment. Another EEG was done after 3 months of P5P treatment. Primary outcome measures were reduction of seizure frequency and EEG improvements. RESULTS: Seven participants (one female, six males; age range 5-23 year; mean age 11 years 10 months, SD 5 year 2 months) were included. The genetic causes of inherited GPI deficiency were phosphatidylinositol N-acetylglucosaminyltransferase subunit A/T/V deficiency. All had drug-resistant epilepsy and neurodevelopmental impairment. We observed more than 50% seizure frequency reduction in 2 out of 7 and less than 50% reduction in another 3 out of 7 participants. No participants reached seizure freedom. No remarkable changes in electrophysiological findings were observed in 6 out of 7 participants treated with pyridoxine or P5P when comparing the baseline and follow-up EEGs. INTERPRETATION: We observed no long-lasting electrophysiological improvements during treatment but pyridoxine may reduce seizure frequency or burden in inherited GPI deficiency. WHAT THIS PAPER ADDS: Inherited glycosylphosphatidylinositol (GPI) deficiency often causes early-onset and drug-resistant epilepsy. Vitamin B6 is a potential disease-specific treatment; however, efficacy and safety are ill-defined. Pyridoxine may reduce seizure frequency or burden in inherited GPI deficiency. Pyridoxine and P5P could prove to be a useful treatment in some individuals with inherited GPI deficiency and epilepsy.


Subject(s)
Drug Resistant Epilepsy , Epilepsy , Cohort Studies , Drug Resistant Epilepsy/drug therapy , Epilepsy/complications , Epilepsy/drug therapy , Epilepsy/genetics , Female , Glycosylphosphatidylinositols/deficiency , Glycosylphosphatidylinositols/therapeutic use , Humans , Infant , Male , Phosphates/therapeutic use , Prospective Studies , Pyridoxal Phosphate/therapeutic use , Pyridoxine/therapeutic use , Seizures/drug therapy , Seizures/etiology
6.
Brain Dev ; 43(6): 680-687, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33824024

ABSTRACT

OBJECTIVE: We aimed to assess the efficacy and safety of high-dose pyridoxine treatment for seizures and its effects on development in patients with inherited glycosylphosphatidylinositol deficiencies (IGDs). METHODS: In this prospective open-label multicenter pilot study, we enrolled patients diagnosed with IGDs using flow cytometry and/or genetic tests. The patients received oral pyridoxine (20-30 mg/kg/day) for 1 year, in addition to previous treatment. RESULTS: All nine enrolled patients (mean age: 66.3 ± 44.3 months) exhibited marked decreases in levels of CD16, a glycosylphosphatidylinositol-anchored protein, on blood granulocytes. The underlying genetic causes of IGDs were PIGO, PIGL, and unknown gene mutations in two, two, and five patients, respectively. Six patients experienced seizures, while all patients presented with developmental delay (mean developmental age: 11.1 ± 8.1 months). Seizure frequencies were markedly (>50%) and drastically (>90%) reduced in three and one patients who experienced seizures, respectively. None of the patients presented with seizure exacerbation. Eight of nine patients exhibited modest improvements in development (P = 0.14). No adverse events were observed except for mild transient diarrhea in one patient. CONCLUSION: One year of daily high-dose pyridoxine treatment was effective in the treatment of seizures in more than half of our patients with IGDs and modestly improved development in the majority of them. Moreover, such treatment was reasonably safe. These findings indicate that high-dose pyridoxine treatment may be effective against seizures in patients with IGDs, although further studies are required to confirm our findings. (University Hospital Medical Information Network Clinical Trials Registry [UMIN-CTR] number: UMIN000024185.).


Subject(s)
Glycosylphosphatidylinositols/deficiency , Pyridoxine/pharmacology , Seizures/drug therapy , Vitamin B Complex/pharmacology , Adolescent , Child , Child, Preschool , Female , Glycosylphosphatidylinositols/genetics , Humans , Infant , Male , Outcome Assessment, Health Care , Pilot Projects , Prospective Studies , Pyridoxine/administration & dosage , Seizures/complications , Seizures/etiology , Seizures/genetics , Vitamin B Complex/administration & dosage
7.
Blood ; 137(26): 3660-3669, 2021 07 01.
Article in English | MEDLINE | ID: mdl-33763700

ABSTRACT

Glycosylphosphatidylinositol (GPI) is a glycolipid that anchors >150 proteins to the cell surface. Pathogenic variants in several genes that participate in GPI biosynthesis cause inherited GPI deficiency disorders. Here, we reported that homozygous null alleles of PIGG, a gene involved in GPI modification, are responsible for the rare Emm-negative blood phenotype. Using a panel of K562 cells defective in both the GPI-transamidase and GPI remodeling pathways, we show that the Emm antigen, whose molecular basis has remained unknown for decades, is carried only by free GPI and that its epitope is composed of the second and third ethanolamine of the GPI backbone. Importantly, we show that the decrease in Emm expression in several inherited GPI deficiency patients is indicative of GPI defects. Overall, our findings establish Emm as a novel blood group system, and they have important implications for understanding the biological function of human free GPI.


Subject(s)
Blood Group Antigens , Developmental Disabilities , Glycosylphosphatidylinositols/deficiency , Glycosylphosphatidylinositols/metabolism , Phosphotransferases (Alcohol Group Acceptor) , Seizures , Blood Group Antigens/genetics , Blood Group Antigens/metabolism , Developmental Disabilities/enzymology , Developmental Disabilities/genetics , Glycosylphosphatidylinositols/genetics , Humans , K562 Cells , Phosphotransferases (Alcohol Group Acceptor)/genetics , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Seizures/enzymology , Seizures/genetics
8.
Hum Mol Genet ; 30(9): 758-770, 2021 05 28.
Article in English | MEDLINE | ID: mdl-33607654

ABSTRACT

Posttranslational modification of a protein with glycosylphosphatidylinositol (GPI) is a conserved mechanism exists in all eukaryotes. Thus far, >150 human GPI-anchored proteins have been discovered and ~30 enzymes have been reported to be involved in the biosynthesis and maturation of mammalian GPI. Phosphatidylinositol glycan biosynthesis class A protein (PIGA) catalyzes the very first step of GPI anchor biosynthesis. Patients carrying a mutation of the PIGA gene usually suffer from inherited glycosylphosphatidylinositol deficiency (IGD) with intractable epilepsy and intellectual developmental disorder. We generated three mouse models with PIGA deficits specifically in telencephalon excitatory neurons (Ex-M-cko), inhibitory neurons (In-M-cko) or thalamic neurons (Th-H-cko), respectively. Both Ex-M-cko and In-M-cko mice showed impaired long-term fear memory and were more susceptible to kainic acid-induced seizures. In addition, In-M-cko demonstrated a severe limb-clasping phenotype. Hippocampal synapse changes were observed in Ex-M-cko mice. Our Piga conditional knockout mouse models provide powerful tools to understand the cell-type specific mechanisms underlying inherited GPI deficiency and to test different therapeutic modalities.


Subject(s)
Glycosylphosphatidylinositols , Kainic Acid , Animals , Cognition , Glycosylphosphatidylinositols/deficiency , Humans , Kainic Acid/metabolism , Mammals , Mice , Mice, Knockout , Mutation , Neurons/metabolism , Seizures/genetics , Seizures/metabolism
9.
Hum Genet ; 140(6): 879-884, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33386993

ABSTRACT

DOORS syndrome is characterized by deafness, onychodystrophy, osteodystrophy, intellectual disability, and seizures. In this study, we report two unrelated individuals with DOORS syndrome without deafness. Exome sequencing revealed a homozygous missense variant in PIGF (NM_173074.3:c.515C>G, p.Pro172Arg) in both. We demonstrate impaired glycosylphosphatidylinositol (GPI) biosynthesis through flow cytometry analysis. We thus describe the causal role of a novel disease gene, PIGF, in DOORS syndrome and highlight the overlap between this condition and GPI deficiency disorders. For each gene implicated in DOORS syndrome and/or inherited GPI deficiencies, there is considerable clinical variability so a high index of suspicion is warranted even though not all features are noted.


Subject(s)
Craniofacial Abnormalities/genetics , Glycosylphosphatidylinositols/deficiency , Hand Deformities, Congenital/genetics , Hearing Loss, Sensorineural/genetics , Intellectual Disability/genetics , Membrane Proteins/genetics , Mutation, Missense , Nails, Malformed/genetics , Seizures/genetics , Adolescent , Amino Acid Sequence , Animals , Consanguinity , Craniofacial Abnormalities/metabolism , Craniofacial Abnormalities/pathology , Female , Gene Expression , Glycosylphosphatidylinositols/genetics , Glycosylphosphatidylinositols/metabolism , HEK293 Cells , Hand Deformities, Congenital/metabolism , Hand Deformities, Congenital/pathology , Hearing Loss, Sensorineural/metabolism , Hearing Loss, Sensorineural/pathology , Homozygote , Humans , Infant , Intellectual Disability/metabolism , Intellectual Disability/pathology , Male , Membrane Proteins/deficiency , Nails, Malformed/metabolism , Nails, Malformed/pathology , Seizures/metabolism , Seizures/pathology , Sequence Alignment , Exome Sequencing
10.
Hum Genet ; 140(5): 791-803, 2021 May.
Article in English | MEDLINE | ID: mdl-33392778

ABSTRACT

PIGK gene, encoding a key component of glycosylphosphatidylinositol (GPI) transamidase, was recently reported to be associated with inherited GPI deficiency disorders (IGDs). However, little is known about the specific downstream effects of PIGK on neurodevelopment due to the rarity of the disease and the lack of in vivo study. Here, we described 2 patients in a Chinese family presented with profound global developmental delay, severe hypotonia, seizures, and postnatal progressive global brain atrophy including hemisphere, cerebellar and corpus callosum atrophy. Two novel compound heterozygous variants in PIGK were identified via genetic analysis, which was proved to cause significant decrease of PIGK protein and reduced cell surface presence of GPI-APs in the patients. To explore the role of Pigk on embryonic and neuronal development, we constructed Pigk knock-down zebrafish and knock-in mouse models. Zebrafish injected with a small dose of morpholino oligonucleotides displayed severe developmental defects including small eyes, deformed head, curly spinal cord, and unconsumed yolk sac. Primary motor neuronal dysplasia and extensive neural cell apoptosis were further observed. Meanwhile, the mouse models, carrying the two variants respectively homologous with the patients, both resulted in complete embryonic lethality of the homozygotes, which suggested the intolerable effect caused by amino acid substitution of Asp204 as well as the truncated mutation. Our findings provide the in vivo evidence for the essential role of PIGK during the embryonic and neuronal development. Based on these data, we propose a basis for further study of pathological and molecular mechanisms of PIGK-related neurodevelopmental defects.


Subject(s)
Brain Diseases/genetics , Cell Adhesion Molecules/genetics , Glycosylphosphatidylinositols/deficiency , Nervous System Malformations/genetics , Neurogenesis/genetics , Seizures/genetics , Abnormalities, Multiple/genetics , Animals , Apoptosis/genetics , Cell Line , Child, Preschool , Disease Models, Animal , Embryonic Development/genetics , Gene Knock-In Techniques , Glycosylphosphatidylinositols/genetics , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Zebrafish
11.
Proc Natl Acad Sci U S A ; 118(2)2021 01 12.
Article in English | MEDLINE | ID: mdl-33402532

ABSTRACT

Pathogenic germline mutations in PIGV lead to glycosylphosphatidylinositol biosynthesis deficiency (GPIBD). Individuals with pathogenic biallelic mutations in genes of the glycosylphosphatidylinositol (GPI)-anchor pathway exhibit cognitive impairments, motor delay, and often epilepsy. Thus far, the pathophysiology underlying the disease remains unclear, and suitable rodent models that mirror all symptoms observed in human patients have not been available. Therefore, we used CRISPR-Cas9 to introduce the most prevalent hypomorphic missense mutation in European patients, Pigv:c.1022C > A (p.A341E), at a site that is conserved in mice. Mirroring the human pathology, mutant Pigv341E mice exhibited deficits in motor coordination, cognitive impairments, and alterations in sociability and sleep patterns, as well as increased seizure susceptibility. Furthermore, immunohistochemistry revealed reduced synaptophysin immunoreactivity in Pigv341E mice, and electrophysiology recordings showed decreased hippocampal synaptic transmission that could underlie impaired memory formation. In single-cell RNA sequencing, Pigv341E-hippocampal cells exhibited changes in gene expression, most prominently in a subtype of microglia and subicular neurons. A significant reduction in Abl1 transcript levels in several cell clusters suggested a link to the signaling pathway of GPI-anchored ephrins. We also observed elevated levels of Hdc transcripts, which might affect histamine metabolism with consequences for circadian rhythm. This mouse model will not only open the doors to further investigation into the pathophysiology of GPIBD, but will also deepen our understanding of the role of GPI-anchor-related pathways in brain development.


Subject(s)
Glycosylphosphatidylinositols/genetics , Glycosylphosphatidylinositols/metabolism , Mannosyltransferases/metabolism , Abnormalities, Multiple/genetics , Amino Acid Sequence , Amino Acids/genetics , Animals , CRISPR-Cas Systems , Disease Models, Animal , Epilepsy/genetics , Glycosylphosphatidylinositols/deficiency , Hippocampus/metabolism , Intellectual Disability/genetics , Mannosyltransferases/physiology , Mice , Mice, Inbred C57BL , Mutation , Mutation, Missense , Phenotype , Protein Engineering/methods , Seizures/genetics , Seizures/physiopathology
12.
Clin Genet ; 98(5): 468-476, 2020 11.
Article in English | MEDLINE | ID: mdl-32725661

ABSTRACT

PIGT is one of over 29 glycosylphosphatidylinositol biosynthesis defect genes. Mutations cause genetically determined disorders characterized mainly by epilepsy with fever-sensitivity, central hypotonia, psychomotor delay and congenital malformations. The disease is known as multiple congenital anomalies-hypotonia-seizures syndrome 3 (MCAHS3) or glycosylphosphatidylinositol biosynthesis defect-7. Twenty-eight cases have been reported until today. We present seven novel Polish patients, all harboring 1582G>A variant in a homozygous or compound heterozygous state which seems to cause a milder phenotype of the disease.


Subject(s)
Acyltransferases/genetics , Epilepsy/genetics , Glycosylphosphatidylinositols/deficiency , Intellectual Disability/genetics , Psychomotor Disorders/genetics , Seizures/genetics , Child , Child, Preschool , Developmental Disabilities/complications , Developmental Disabilities/genetics , Developmental Disabilities/pathology , Epilepsy/complications , Epilepsy/pathology , Female , Flow Cytometry , Glycosylphosphatidylinositols/genetics , Homozygote , Humans , Infant , Intellectual Disability/complications , Intellectual Disability/pathology , Male , Mutation/genetics , Nervous System Malformations/complications , Nervous System Malformations/genetics , Nervous System Malformations/pathology , Pedigree , Phenotype , Poland , Psychomotor Disorders/pathology , Seizures/complications , Seizures/pathology
13.
Mol Genet Metab ; 130(1): 49-57, 2020 05.
Article in English | MEDLINE | ID: mdl-32165008

ABSTRACT

BACKGROUND: Mutations in the ARV1 Homolog, Fatty Acid Homeostasis Modulator (ARV1), have recently been described in association with early infantile epileptic encephalopathy 38. Affected individuals presented with epilepsy, ataxia, profound intellectual disability, visual impairment, and central hypotonia. In S. cerevisiae, Arv1 is thought to be involved in sphingolipid metabolism and glycophosphatidylinositol (GPI)-anchor synthesis. The function of ARV1 in human cells, however, has not been elucidated. METHODS: Mutations were discovered through whole exome sequencing and alternate splicing was validated on the cDNA level. Expression of the variants was determined by qPCR and Western blot. Expression of GPI-anchored proteins on neutrophils and fibroblasts was analyzed by FACS and immunofluorescence microscopy, respectively. RESULTS: Here we describe seven patients from two unrelated families with biallelic splice mutations in ARV1. The patients presented with early onset epilepsy, global developmental delays, profound hypotonia, delayed speech development, cortical visual impairment, and severe generalized cerebral and cerebellar atrophy. The splice variants resulted in decreased ARV1 expression and significant decreases in GPI-anchored protein on the membranes of neutrophils and fibroblasts, indicating that the loss of ARV1 results in impaired GPI-anchor synthesis. CONCLUSION: Loss of GPI-anchored proteins on our patients' cells confirms that the yeast Arv1 function of GPI-anchor synthesis is conserved in humans. Overlap between the phenotypes in our patients and those reported for other GPI-anchor disorders suggests that ARV1-deficiency is a GPI-anchor synthesis disorder.


Subject(s)
Abnormalities, Multiple/genetics , Carrier Proteins/genetics , Carrier Proteins/metabolism , Developmental Disabilities/genetics , Epilepsy/genetics , Glycosylphosphatidylinositols/deficiency , Intellectual Disability/genetics , Membrane Proteins/genetics , Membrane Proteins/metabolism , Abnormalities, Multiple/physiopathology , Adolescent , Alternative Splicing/genetics , Child, Preschool , Developmental Disabilities/physiopathology , Epilepsy/physiopathology , Female , Fibroblasts/metabolism , GPI-Linked Proteins/metabolism , Glycosylphosphatidylinositols/biosynthesis , Homozygote , Humans , Infant , Infant, Newborn , Male , Mutation , Neutrophils/metabolism , Pedigree , Exome Sequencing
14.
Am J Med Genet A ; 182(6): 1477-1482, 2020 06.
Article in English | MEDLINE | ID: mdl-32198969

ABSTRACT

Inherited glycosylphosphatidylinositol (GPI) deficiencies are a group of clinically and genetically heterogeneous conditions belonging to the congenital disorders of glycosylation. PIGW is involved in GPI biosynthesis and modification, and biallelic pathogenic variants in this gene cause autosomal recessive GPI biosynthesis defect 11. Only five patients and two fetuses have been reported in the literature thus far. Here we describe a new patient with a novel homozygous missense variant in PIGW, who presented with hypotonia, severe intellectual disability, early-onset epileptic seizures, brain abnormalities, nystagmus, hand stereotypies, recurrent respiratory infections, distinctive facial features, and hyperphosphatasia. Our report expands the phenotype of GPI biosynthesis defect 11 to include stereotypies and recurrent respiratory infections. A detailed and long-term analysis of the electroclinical characteristics and review of the literature suggest that early-onset epileptic seizures are a key manifestation of GPI biosynthesis defect 11. West syndrome and focal-onset epileptic seizures are the most common seizure types, and the fronto-temporal regions may be the most frequently involved areas in these patients.


Subject(s)
Acyltransferases/genetics , Glycosylphosphatidylinositols/deficiency , Glycosylphosphatidylinositols/genetics , Intellectual Disability/genetics , Membrane Proteins/genetics , Seizures/genetics , Brain/abnormalities , Brain/pathology , Child , Child, Preschool , Female , Glycosylphosphatidylinositols/biosynthesis , Humans , Infant , Intellectual Disability/complications , Intellectual Disability/pathology , Male , Muscle Hypotonia/complications , Muscle Hypotonia/genetics , Muscle Hypotonia/pathology , Mutation, Missense/genetics , Seizures/complications , Seizures/pathology , Seizures/physiopathology
15.
Hum Mol Genet ; 29(7): 1205-1217, 2020 05 08.
Article in English | MEDLINE | ID: mdl-32179897

ABSTRACT

The glycosylphosphatidylinositol (GPI) anchor is a post-translational modification added to approximately 150 different proteins to facilitate proper membrane anchoring and trafficking to lipid rafts. Biosynthesis and remodeling of the GPI anchor requires the activity of over 20 distinct genes. Defects in the biosynthesis of GPI anchors in humans lead to inherited glycosylphosphatidylinositol deficiency (IGD). IGD patients display a wide range of phenotypes though the central nervous system (CNS) appears to be the most commonly affected tissue. A full understanding of the etiology of these phenotypes has been hampered by the lack of animal models due to embryonic lethality of GPI biosynthesis gene null mutants. Here we model IGD by genetically ablating GPI production in the CNS with a conditional mouse allele of phosphatidylinositol glycan anchor biosynthesis, class A (Piga) and Nestin-Cre. We find that the mutants do not have structural brain defects but do not survive past weaning. The mutants show progressive decline with severe ataxia consistent with defects in cerebellar development. We show that the mutants have reduced myelination and defective Purkinje cell development. Surprisingly, we found that Piga was expressed in a fairly restricted pattern in the early postnatal brain consistent with the defects we observed in our model. Thus, we have generated a novel mouse model of the neurological defects of IGD which demonstrates a critical role for GPI biosynthesis in cerebellar and white matter development.


Subject(s)
Central Nervous System Diseases/genetics , Central Nervous System/metabolism , Cerebellar Ataxia/genetics , Glycosylphosphatidylinositols/deficiency , Seizures/genetics , Animals , Central Nervous System/pathology , Central Nervous System Diseases/pathology , Cerebellar Ataxia/metabolism , Cerebellar Ataxia/pathology , Disease Models, Animal , Glycosylphosphatidylinositols/genetics , Humans , Mice , Mortality, Premature , Mutation/genetics , Phenotype , Seizures/pathology , White Matter/metabolism , White Matter/pathology
16.
Br J Haematol ; 189(5): 954-966, 2020 06.
Article in English | MEDLINE | ID: mdl-32103498

ABSTRACT

A retrospective analysis of presentation clinical, laboratory and immunophenotypic features of 1 081 patients with paroxysmal nocturnal haemoglobinuria (PNH) clones [glycosylphosphatidylinositol (GPI)-deficient blood cells] identified at our hospital by flow cytometry over the past 25 years was undertaken. Three distinct clusters of patients were identified and significant correlations between presentation disease type and PNH clone sizes were evident. Smaller PNH clones predominate in cytopenic and myelodysplastic subtypes; large PNH clones were associated with haemolytic, thrombotic and haemolytic/thrombotic subtypes. Rare cases with an associated chronic myeloproliferative disorder had either large or small PNH clones. Cytopenia was a frequent finding, highlighting bone marrow failure as the major underlying feature associated with the detection of PNH clones in the peripheral blood. Red cell PNH clones showed significant correlations between the presence of type II (partial GPI deficiency) red cells and thrombotic disease. Haemolytic PNH was associated with type III (complete GPI deficiency) red cell populations of >20%. Those with both haemolytic and thrombotic features had major type II and type III red cell populations. Distinct patterns of presentation age decade were evident for clinical subtypes with a peak incidence of haemolytic PNH in the 30-49 year age group and a biphasic age distribution for the cytopenia group.


Subject(s)
Glycosylphosphatidylinositols/deficiency , Hemoglobinuria, Paroxysmal/blood , Adolescent , Adult , Aged , Aged, 80 and over , Anemia, Aplastic/etiology , Anemia, Hemolytic/etiology , CD55 Antigens/deficiency , CD59 Antigens/deficiency , Child , Child, Preschool , Clonal Evolution , Clone Cells/pathology , Disease Progression , Female , Flow Cytometry , Hemoglobinuria, Paroxysmal/complications , Hemoglobinuria, Paroxysmal/genetics , Hemoglobinuria, Paroxysmal/pathology , Humans , Immunophenotyping , Infant , Lymphocytes/pathology , Male , Middle Aged , Myeloproliferative Disorders/etiology , Neutrophils/pathology , Receptors, Transferrin/blood , Retrospective Studies , Thrombosis/etiology , Young Adult
17.
Eur J Med Genet ; 63(4): 103822, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31805394

ABSTRACT

We report that recessive inheritance of a post-GPI attachment to proteins 2 (PGAP2) gene variant results in the hyperphosphatasia with neurologic deficit (HPMRS) phenotype described by Mabry et al., in 1970. HPMRS, or Mabry syndrome, is now known to be one of 21 inherited glycosylphosphatidylinositol (GPI) deficiencies (IGDs), or GPI biosynthesis defects (GPIBDs). Bi-allelic mutations in at least six genes result in HPMRS phenotypes. Disruption of four phosphatidylinositol glycan (PIG) biosynthesis genes, PIGV, PIGO, PIGW and PIGY, expressed in the endoplasmic reticulum, result in HPMRS 1, 2, 5 and 6; disruption of the PGAP2 and PGAP3 genes, necessary for stabilizing the association of GPI anchored proteins (AP) with the Golgi membrane, result in HPMRS 3 and 4. We used exome sequencing to identify a novel homozygous missense PGAP2 variant NM_014489.3:c.881C > T, p.Thr294Met in two index patients and targeted sequencing to identify this variant in an unrelated patient. Rescue assays were conducted in two PGAP2 deficient cell lines, PGAP2 KO cells generated by CRISPR/Cas9 and PGAP2 deficient CHO cells, in order to examine the pathogenicity of the PGAP2 variant. First, we used the CHO rescue assay to establish that the wild type PGAP2 isoform 1, translated from transcript 1, is less active than the wild type PGAP2 isoform 8, translated from transcript 12 (alternatively spliced to omit exon 3). As a result, in our variant rescue assays, we used the more active NM_001256240.2:c.698C > T, p.Thr233Met isoform 8 instead of NM_014489.3:c.881C > T, p.Thr294Met isoform 1. Flow cytometric analysis showed that restoration of cell surface CD59 and CD55 with variant PGAP2 isoform 8, driven by the weak (pTA FLAG) promoter, was less efficient than wild type isoform 8. Therefore, we conclude that recessive inheritance of c.881C > T PGAP2, expressed as the hypomorphic PGAP2 c.698C > T, p.Thr233Met isoform 8, results in prototypical Mabry phenotype, HPMRS3 (GPIBD 8 [MIM: 614207]). This study highlights the need for long-term follow up of individuals with rare diseases in order to ensure that they benefit from innovations in diagnosis and treatment.


Subject(s)
Abnormalities, Multiple/genetics , Intellectual Disability/genetics , Nuclear Proteins/genetics , Phosphorus Metabolism Disorders/genetics , Adolescent , Adult , Animals , CHO Cells , Child , Cricetulus , Female , Glycosylphosphatidylinositols/deficiency , HEK293 Cells , Humans , Male , Middle Aged , Mutation, Missense , Nuclear Proteins/metabolism , Young Adult
18.
Eur J Med Genet ; 63(4): 103802, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31698102

ABSTRACT

Mabry syndrome is a glycophosphatidylinositol (GPI) deficiency characterized by intellectual disability, distinctive facial features, intractable seizures, and hyperphosphatasia. We expand the phenotypic spectrum of inherited GPI deficiencies with novel bi-allelic phosphatidylinositol glycan anchor biosynthesis class O (PIGO) variants in a neonate who presented with intractable epilepsy and complex gastrointestinal and urogenital malformations.


Subject(s)
Abnormalities, Multiple/genetics , Drug Resistant Epilepsy/genetics , Glycosylphosphatidylinositols/deficiency , Intellectual Disability/genetics , Membrane Proteins/genetics , Phosphorus Metabolism Disorders/genetics , Urogenital Abnormalities/genetics , Female , Gastrointestinal Tract/abnormalities , Genetic Variation , Humans , Infant, Newborn , Phenotype
19.
Mol Genet Metab ; 128(1-2): 151-161, 2019.
Article in English | MEDLINE | ID: mdl-31445883

ABSTRACT

Defects of the glycosylphosphatidylinositol (GPI) biosynthesis pathway constitute an emerging subgroup of congenital disorders of glycosylation with heterogeneous phenotypes. A mutation in the promoter of PIGM, resulting in a syndrome with portal vein thrombosis and persistent absence seizures, was previously described in three patients. We now report four additional patients in two unrelated families, with further clinical, biochemical and molecular delineation of this unique entity. We also describe the first prenatal diagnosis of PIGM deficiency, allowing characterization of the natural history of the disease from birth. The patients described herein expand the phenotypic spectrum of PIGM deficiency to include macrocephaly and infantile-onset cerebrovascular thrombotic events. Finally, we offer insights regarding targeted treatment of this rare disorder with sodium phenylbutyrate.


Subject(s)
Glycosylphosphatidylinositols/deficiency , Mannosyltransferases/genetics , Megalencephaly/etiology , Portal Vein/pathology , Seizures/etiology , Thrombosis/etiology , Adolescent , Child , Child, Preschool , Female , Glycosylphosphatidylinositols/genetics , Humans , Infant , Magnetic Resonance Imaging , Male , Megalencephaly/diagnostic imaging , Mutation , Promoter Regions, Genetic , Seizures/complications , Seizures/genetics
20.
Am J Hum Genet ; 105(2): 384-394, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31256876

ABSTRACT

Proteins anchored to the cell surface via glycosylphosphatidylinositol (GPI) play various key roles in the human body, particularly in development and neurogenesis. As such, many developmental disorders are caused by mutations in genes involved in the GPI biosynthesis and remodeling pathway. We describe ten unrelated families with bi-allelic mutations in PIGB, a gene that encodes phosphatidylinositol glycan class B, which transfers the third mannose to the GPI. Ten different PIGB variants were found in these individuals. Flow cytometric analysis of blood cells and fibroblasts from the affected individuals showed decreased cell surface presence of GPI-anchored proteins. Most of the affected individuals have global developmental and/or intellectual delay, all had seizures, two had polymicrogyria, and four had a peripheral neuropathy. Eight children passed away before four years old. Two of them had a clinical diagnosis of DOORS syndrome (deafness, onychodystrophy, osteodystrophy, mental retardation, and seizures), a condition that includes sensorineural deafness, shortened terminal phalanges with small finger and toenails, intellectual disability, and seizures; this condition overlaps with the severe phenotypes associated with inherited GPI deficiency. Most individuals tested showed elevated alkaline phosphatase, which is a characteristic of the inherited GPI deficiency but not DOORS syndrome. It is notable that two severely affected individuals showed 2-oxoglutaric aciduria, which can be seen in DOORS syndrome, suggesting that severe cases of inherited GPI deficiency and DOORS syndrome might share some molecular pathway disruptions.


Subject(s)
Craniofacial Abnormalities/etiology , Glycosylphosphatidylinositols/biosynthesis , Glycosylphosphatidylinositols/deficiency , Hand Deformities, Congenital/etiology , Hearing Loss, Sensorineural/etiology , Intellectual Disability/etiology , Mannosyltransferases/genetics , Metabolic Diseases/etiology , Mutation , Nails, Malformed/etiology , Peripheral Nervous System Diseases/etiology , Seizures/pathology , Adult , Child , Child, Preschool , Craniofacial Abnormalities/pathology , Female , Glycosylphosphatidylinositols/genetics , Hand Deformities, Congenital/pathology , Hearing Loss, Sensorineural/pathology , Humans , Infant , Infant, Newborn , Intellectual Disability/pathology , Male , Metabolic Diseases/pathology , Nails, Malformed/pathology , Pedigree , Peripheral Nervous System Diseases/pathology , Seizures/genetics , Severity of Illness Index , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...