Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.886
Filter
1.
Mol Genet Genomic Med ; 12(10): e70016, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39359128

ABSTRACT

OBJECTIVE: To investigate the clinical value of whole-exome sequencing (WES) in the diagnosis of foetuses with central nervous system (CNS) abnormalities but having a normal karyotyping and chromosomal microarray result. METHOD: During the period of 2016-2022, there were a total of 149 foetuses with CNS abnormalities but having negative karyotyping and chromosomal microarray analysis results; WES was performed on these foetuses and their parents. Variants were classified according to ACMG guidelines, and the association of pathogenic variants with specific types of CNS abnormalities was explored. RESULTS: Among these 149 foetuses, three categories of abnormalities, namely, single CNS abnormality, multiple CNS abnormalities, CNS abnormalities along with other organ system abnormalities were identified, for which the detection rate of P/LP variants is 17.4% (12/69), 28.6% (14/49) and 54.8% (17/31), respectively. CONCLUSION: WES brought about an increase of 28.9% in diagnostic yield in the prenatal evaluation of foetuses with CNS abnormalities but having negative karyotyping and chromosome array results. WES may also be of benefit for the diagnosis of foetuses with isolated CNS abnormalities, as well as for making more informed interpretations of imaging findings and for providing better genetic counselling.


Subject(s)
Exome Sequencing , Prenatal Diagnosis , Humans , Female , Exome Sequencing/methods , Pregnancy , Prenatal Diagnosis/methods , Nervous System Malformations/genetics , Nervous System Malformations/diagnosis , Adult , Genetic Testing/methods , Fetus/abnormalities , Central Nervous System/abnormalities , Karyotyping/methods
2.
Proc Natl Acad Sci U S A ; 121(38): e2411747121, 2024 Sep 17.
Article in English | MEDLINE | ID: mdl-39254994

ABSTRACT

Detection of cytosolic DNA by the cyclic GMP-AMP (cGAMP) synthase (cGAS)-stimulator of interferon genes (STING) pathway provides immune defense against pathogens and cancer but can also cause autoimmunity when overactivated. The exonuclease three prime repair exonuclease 1 (TREX1) degrades DNA in the cytosol and prevents cGAS activation by self-DNA. Loss-of-function mutations of the TREX1 gene are linked to autoimmune diseases such as Aicardi-Goutières syndrome, and mice deficient in TREX1 develop lethal inflammation in a cGAS-dependent manner. In order to determine the type of cells in which cGAS activation drives autoinflammation, we generated conditional cGAS knockout mice on the Trex1-/- background. Here, we show that genetic ablation of the cGAS gene in classical dendritic cells (cDCs), but not in macrophages, was sufficient to rescue Trex1-/- mice from all observed disease phenotypes including lethality, T cell activation, tissue inflammation, and production of antinuclear antibodies and interferon-stimulated genes. These results show that cGAS activation in cDC causes autoinflammation in response to self-DNA accumulated in the absence of TREX1.


Subject(s)
Autoimmunity , Dendritic Cells , Exodeoxyribonucleases , Mice, Knockout , Nucleotidyltransferases , Phosphoproteins , Animals , Exodeoxyribonucleases/genetics , Exodeoxyribonucleases/deficiency , Exodeoxyribonucleases/metabolism , Nucleotidyltransferases/genetics , Nucleotidyltransferases/metabolism , Nucleotidyltransferases/deficiency , Dendritic Cells/immunology , Phosphoproteins/genetics , Phosphoproteins/metabolism , Phosphoproteins/immunology , Mice , Autoimmunity/immunology , Nervous System Malformations/genetics , Nervous System Malformations/immunology , Autoimmune Diseases of the Nervous System/immunology , Autoimmune Diseases of the Nervous System/genetics , Autoimmune Diseases of the Nervous System/pathology , Inflammation/immunology , Inflammation/genetics , Macrophages/immunology , Macrophages/metabolism , Autoimmune Diseases/immunology , Autoimmune Diseases/genetics
3.
Handb Clin Neurol ; 204: 95-109, 2024.
Article in English | MEDLINE | ID: mdl-39322397

ABSTRACT

Intracranial calcification (ICC) occurs in many neurologic disorders both acquired and genetic. In some inherited white matter disorders, it is a common or even invariable feature where the presence and pattern of calcification provides an important pointer to the specific diagnosis. This is particularly the case for the Aicardi-Goutières syndrome (AGS) and for Coats plus (CP) and leukoencephalopathy with calcifications and cysts (LCC), which are discussed in detail in this chapter. AGS is a genetic disorder of type 1 interferon regulation, caused by mutations in any of the nine genes identified to date. In its classic form, AGS has very characteristic clinical and neuroimaging features which will be discussed here. LCC is a purely neurologic disorder caused by mutations in the SNORD118 gene, whereas CP is a multisystem disorder of telomere function that may result from mutations in the CTC1, POT1, or STN genes. In spite of the different pathogenetic basis for LCC and CP, they share remarkably similar neuroimaging and neuropathologic features. Cockayne syndrome, in which ICC is usually present, is discussed elsewhere in this volume. ICC may occur as an occasional feature of many other white matter diseases, including Alexander disease, Krabbe disease, X-ALD, and occulodentodigital dysplasia.


Subject(s)
Calcinosis , Leukoencephalopathies , Nervous System Malformations , Humans , Calcinosis/genetics , Calcinosis/diagnostic imaging , Calcinosis/pathology , Leukoencephalopathies/genetics , Leukoencephalopathies/diagnostic imaging , Leukoencephalopathies/pathology , Nervous System Malformations/genetics , Nervous System Malformations/diagnostic imaging , Nervous System Malformations/pathology , Child , Central Nervous System Cysts/genetics , Central Nervous System Cysts/diagnostic imaging , Central Nervous System Cysts/pathology , Autoimmune Diseases of the Nervous System
4.
Mol Genet Metab ; 143(1-2): 108578, 2024.
Article in English | MEDLINE | ID: mdl-39332260

ABSTRACT

OBJECTIVE: Aicardi Goutières Syndrome (AGS) is a rare genetic interferonopathy associated with diverse multisystemic complications. A critical gap exists in our understanding of its longitudinal, systemic disease burden, complicated by delayed diagnosis. To address this need, real-world data extracted from existing medical records were used to characterize the longitudinal disease burden. METHODS: All subjects (n = 167) with genetically confirmed AGS enrolled in the Myelin Disorders Biorepository Project (MDBP) were included. As available in medical records, information was collected on subject demographics, age of onset, and disease complications. Information from published cases of AGS (2007-2022; n = 129) with individual-level data was also collected. Neurologic severity at the last available encounter was determined by retrospectively assigning the AGS Severity Scale [severe (0-3), moderate (4-8), and mild (9-11)]. RESULTS: The genotype frequency in the natural history cohort was TREX1 (n = 26, 15.6 %), RNASEH2B (n = 50, 29.9 %), RNASEH2C (n = 3, 1.8 %), RNASEH2A (n = 7, 4.2 %), SAMHD1 (n = 25, 15.0 %), ADAR (n = 34, 20.4 %), IFIH1 (n = 19, 11.4 %), and RNU7-1 (n = 3, 1.8 %). The median age of systemic onset was 0.15 years [IQR = 0.67 years; median range by genotype: 0 (TREX1) - 0.62 (ADAR) years], while the median neurological onset was 0.33 years [IQR = 0.82 years; median range by genotype: 0.08 (TREX1) - 0.90 (ADAR) year]. The most common early systemic complications were gastrointestinal, including dysphagia or feeding intolerance (n = 124) and liver abnormalities (n = 67). Among postnatal complications, thrombocytopenia appeared earliest (n = 29, median 0.06 years). Tone abnormalities (axial hypotonia: n = 145, 86.8 %; dystonia: n = 123, 73.7 %), irritability (n = 115, 68.9 %), and gross motor delay (n = 112, 7.1 %) emerged as the most prevalent neurological symptoms. Previously published case reports demonstrated similar patterns. The median AGS score for the entire cohort was 4 (IQR = 7). The most severe neurologic phenotype occurred in TREX1-related AGS (n = 19, median AGS severity score 2, IQR = 2). Time to feeding tube placement, chilblains, early gross motor delay, early cognitive delay, and motor regression were significantly associated with genotype (Fleming-Harrington log-rank: p = 0.0002, p < 0.0001, p = 0.0038, p < 0.0001, p = 0.0001, respectively). Microcephaly, feeding tube placement, and seizures were associated with lower AGS scores (All: Wilcoxon rank sum test, p < 0.0001). Among the qualifying case reports (n = 129), tone abnormalities were the most prevalent disease feature, with spastic quadriplegia reported in 37 of 96 cases (38.5 %) and dystonia in 30 of 96 cases (31.2 %). CONCLUSIONS: AGS is a heterogeneous disease with multi-organ system dysfunction that compounds throughout the clinical course, resulting in profound neurological and extra-neurological disease impact. Systemic symptoms precede neurologic disease features in most cases. Disease onset before the age of one year, microcephaly, feeding tube placement, and seizures were associated with worse neurological outcomes. This work will inform evidence-based clinical monitoring guidelines and clinical trial design.


Subject(s)
Autoimmune Diseases of the Nervous System , Nervous System Malformations , Humans , Nervous System Malformations/genetics , Nervous System Malformations/complications , Nervous System Malformations/epidemiology , Female , Male , Autoimmune Diseases of the Nervous System/genetics , Autoimmune Diseases of the Nervous System/complications , Child, Preschool , Infant , Child , Phosphoproteins/genetics , Exodeoxyribonucleases/genetics , Retrospective Studies , Adolescent , Ribonuclease H/genetics , SAM Domain and HD Domain-Containing Protein 1/genetics , Genotype , Severity of Illness Index , Mutation , Interferon-Induced Helicase, IFIH1/genetics
5.
Nucleic Acids Res ; 52(18): 11014-11028, 2024 Oct 14.
Article in English | MEDLINE | ID: mdl-39217460

ABSTRACT

Removal of ribonucleotides from DNA by RNaseH2 is essential for genome stability, and its impacted function causes the neurodegenerative disease, Aicardi Goutières Syndrome. We have created a zebrafish rnaseh2a mutant to model this process. Surprisingly, RNaseH2a knockouts show little phenotypic abnormality at adulthood in the first generation, unlike mouse knockout models, which are early embryonic lethal. However, the second generation offspring show reduced development, increased ribonucleotide incorporation and upregulation of key inflammatory markers, resulting in both maternal and paternal embryonic lethality. Thus, neither fathers or mothers can generate viable offspring even when crossed to wild-type partners. Despite their survival, rnaseh2a-/- adults show an accumulation of ribonucleotides in both the brain and testes that is not present in early development. Our data suggest that homozygotes possess RNaseH2 independent compensatory mechanisms that are inactive or overwhelmed by the inherited ribonucleotides in their offspring, or that zebrafish have a yet unknown tolerance mechanism. Additionally, we identify ribodysgenesis, the rapid removal of rNMPs and subsequently lethal fragmentation of DNA as responsible for maternal and paternal embryonic lethality.


Subject(s)
Ribonuclease H , Zebrafish , Animals , Zebrafish/genetics , Zebrafish/embryology , Ribonuclease H/genetics , Ribonuclease H/metabolism , Female , Male , Gene Knockout Techniques , Ribonucleotides/metabolism , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism , Nervous System Malformations/genetics , Testis/metabolism , Autoimmune Diseases of the Nervous System/genetics , Brain/metabolism , Brain/embryology , Disease Models, Animal
6.
Ann Med ; 56(1): 2399317, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39239799

ABSTRACT

OBJECTIVE: Clinical validity of genome sequencing (GS) (>30×) has been preliminarily verified in the post-natal setting. This study is to investigate the potential utility of trio-GS as a prenatal test for diagnosis of central nervous system (CNS) anomalies. METHODS: We performed trio-based GS on a prospective cohort of 17 foetuses with CNS abnormalities. Single nucleotide variation (SNV), small insertion and deletion (Indel), copy number variation (CNV), structural variant (SV), and regions with absence of heterozygosity (AOH) were analyzed and classified according to ACMG guidelines. RESULTS: Trio-GS identified diagnostic findings in 29.4% (5/17) of foetuses, with pathogenic variants found in SON, L1CAM, KMT2D, and ASPM. Corpus callosum (CC) and cavum septum pellucidum (CSP) abnormalities were the most frequent CNS abnormalities (47.1%, 8/17) with a diagnostic yield of 50%. A total of 29.4% (5/17) foetuses had variants of uncertain significance (VUS). Particularly, maternal uniparental disomy 16 and a de novo mosaic 4p12p11 duplication were simultaneously detected in one foetus with abnormal sulcus development. In addition, parentally inherited chromosomal inversions were identified in two foetuses. CONCLUSION: GS demonstrates its feasibility in providing genetic diagnosis for foetal CNS abnormalities and shows the potential to expand the application to foetuses with other ultrasound anomalies in prenatal diagnosis.


Subject(s)
DNA Copy Number Variations , Prenatal Diagnosis , Humans , Female , Pregnancy , Prospective Studies , Prenatal Diagnosis/methods , Whole Genome Sequencing , Adult , Nervous System Malformations/genetics , Nervous System Malformations/diagnosis , Nervous System Malformations/diagnostic imaging , Fetus/abnormalities , Fetus/diagnostic imaging , Central Nervous System/abnormalities , Central Nervous System/diagnostic imaging , Central Nervous System/embryology , Male
7.
Taiwan J Obstet Gynecol ; 63(5): 771-776, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39266164

ABSTRACT

OBJECTIVE: To present the ultrasound imaging and genetic diagnosis of a fetus with prenatal lethal form of Gaucher disease. CASE REPORT: A 37-year-old primiparous woman was pregnant at her 23 weeks of gestation and the prenatal fetal ultrasound revealed hydrops fetalis, cerebellum hypoplasia, and fetal immobility. The pregnancy was terminated due to major fetal anomaly, and whole exome sequencing (WES) analysis of fetal tissue and parental blood unveiled a pathogenic variant in exon 10 of the GBA gene (NM_001005741.3: c.1265T > G: p.L422R) originating from the mother. Additionally, a novel CNV (chr1: 155204785-155205635 deletion, 0.85 kb) spanning exon 10-12 in the GBA gene was identified from the father. This compound heterozygosity confirmed the diagnosis of prenatal lethal form of Gaucher disease and was informative for genetic counseling. CONCLUSION: WES is a powerful tool to detect pathogenic variants among fetuses with nonimmune hydrops fetalis and complex abnormality from prenatal ultrasound. Compound heterozygosity consisted of single nucleotide variants (SNV) and copy number variations (CNVs) may lead rare inherited metabolic disorders including prenatal lethal form of Gaucher disease.


Subject(s)
Cerebellum , DNA Copy Number Variations , Exome Sequencing , Gaucher Disease , Hydrops Fetalis , Ultrasonography, Prenatal , Humans , Female , Gaucher Disease/genetics , Gaucher Disease/diagnosis , Gaucher Disease/complications , Pregnancy , Adult , Hydrops Fetalis/genetics , Hydrops Fetalis/diagnosis , Cerebellum/abnormalities , Cerebellum/diagnostic imaging , Heterozygote , Nervous System Malformations/genetics , Nervous System Malformations/diagnosis , Polymorphism, Single Nucleotide , Glucosylceramidase/genetics , Developmental Disabilities
8.
Best Pract Res Clin Rheumatol ; 38(3): 101975, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39122631

ABSTRACT

The horror autoinflammaticus derived from aberrant type I interferon secretion determines a special group of autoinflammatory diseases named interferonopathies. Diverse mechanisms involved in nucleic acids sensing, metabolizing or the lack of interferon signaling retro-control are responsible for the phenotypes associated to Aicardi-Goutières Syndrome (AGS), Proteasome-Associated Autoinflammatory Diseases (PRAAS), STING-Associated Vasculopathy with Infancy Onset (SAVI) and certain forms of monogenic Systemic lupus erythematosus (SLE). This review approaches interferonopathies from the basic immunogenetic concept to diagnosis and treatment.


Subject(s)
Autoimmune Diseases of the Nervous System , Interferon Type I , Lupus Erythematosus, Systemic , Nervous System Malformations , Humans , Nervous System Malformations/immunology , Nervous System Malformations/genetics , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/genetics , Interferon Type I/immunology , Autoimmune Diseases of the Nervous System/immunology , Autoimmune Diseases of the Nervous System/genetics , Autoimmune Diseases of the Nervous System/physiopathology , Autoimmune Diseases of the Nervous System/diagnosis , Hereditary Autoinflammatory Diseases/immunology , Hereditary Autoinflammatory Diseases/genetics , Hereditary Autoinflammatory Diseases/physiopathology , Membrane Proteins/genetics , Membrane Proteins/immunology
9.
Early Hum Dev ; 197: 106109, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39208705

ABSTRACT

BACKGROUND: Congenital defects of the central nervous system are the second cause of disability in childhood, representing up to 20 % of structural malformations diagnosed prenatally. The accurate prenatal diagnosis of fetal neurological anomalies and the assessment of cortical development are critical for early intervention and improved long-term outcomes. Neurosonography plays a vital role in this process, providing detailed insights into the structural and functional development of the fetal brain. This systematic review aims to synthesize current knowledge on neurosonographic indicators for prenatal diagnosis, with a special focus on cortical development and its impact in cases of fetal growth defects. MATERIAL AND METHODS: We conducted a comprehensive search for primary literature in PubMed database were searched for English and Spanish-language, peer-reviewed literature published in the last 15 years. Additional articles were identified by scrutinizing others search platforms (Cochrane Library, UpToDate). Inclusion criteria were single pregnancy and no known feto-maternal pathologies at the beginning of the study. RESULTS: Of the 361 published abstracts identified, 35 met criteria for inclusion. The review highlighted the importance of detailed neurosonographic assessments, including the evaluation of cerebral fissures such as the Sylvian fissure, parieto-occipital fissure, and calcarine fissure. Targeted ultrasound techniques were found to provide comprehensive insights comparable to fetal magnetic resonance imaging. We underscored the significant impact of intrauterine growth restriction on cortical development, with early intervention being crucial. Genetic and congenital infection screenings were emphasized as essential components of prenatal assessment. CONCLUSION: The assessment of fetal brain maturation patterns according to gestational age allows us to rule out a delay in cortical development. The heterogeneity of methods and evaluable parameters in fetal neurodevelopment makes it necessary to standardize the evaluation of the main structures of interest both for screening and for the diagnosis of cortical development anomalies, even with the aim of trying to improve upgrade prognostic advice.


Subject(s)
Nervous System Malformations , Ultrasonography, Prenatal , Female , Humans , Pregnancy , Cerebral Cortex/abnormalities , Cerebral Cortex/diagnostic imaging , Cerebral Cortex/growth & development , Fetal Development , Nervous System Malformations/diagnostic imaging , Ultrasonography, Prenatal/methods , Ultrasonography, Prenatal/standards
10.
Genes (Basel) ; 15(6)2024 May 23.
Article in English | MEDLINE | ID: mdl-38927610

ABSTRACT

Pathogenic variants in the FKBP10 gene lead to a spectrum of rare autosomal recessive phenotypes, including osteogenesis imperfecta (OI) Type XI, Bruck syndrome Type I (BS I), and the congenital arthrogryposis-like phenotype (AG), each with variable clinical manifestations that are crucial for diagnosis. This study analyzed the clinical-genetic characteristics of patients with these conditions, focusing on both known and newly identified FKBP10 variants. We examined data from 15 patients, presenting symptoms of OI and joint contractures. Diagnostic methods included genealogical analysis, clinical assessments, radiography, whole exome sequencing, and direct automated Sanger sequencing. We diagnosed 15 patients with phenotypes due to biallelic FKBP10 variants-4 with OI Type XI, 10 with BS I, and 1 with the AG-like phenotype-demonstrating polymorphism in disease severity. Ten pathogenic FKBP10 variants were identified, including three novel ones, c.1373C>T (p.Pro458Leu), c.21del (p.Pro7fs), and c.831_832insCG (p.Gly278Argfs), and a recurrent variant, c.831dup (p.Gly278Argfs). Variant c.1490G>A (p.Trp497Ter) was found in two unrelated patients, causing OI XI in one and BS I in the other. Additionally, two unrelated patients with BS I and epidermolysis bullosa shared identical homozygous FKBP10 and KRT14 variants. This observation illustrates the diversity of FKBP10-related pathology and the importance of considering the full spectrum of phenotypes in clinical diagnostics.


Subject(s)
Arthrogryposis , Osteogenesis Imperfecta , Phenotype , Tacrolimus Binding Proteins , Humans , Tacrolimus Binding Proteins/genetics , Male , Female , Arthrogryposis/genetics , Arthrogryposis/pathology , Arthrogryposis/diagnosis , Osteogenesis Imperfecta/genetics , Osteogenesis Imperfecta/pathology , Child , Child, Preschool , Pedigree , Exome Sequencing , Adolescent , Mutation , Infant , Adult , Nervous System Malformations/genetics
11.
JCI Insight ; 9(11)2024 Jun 10.
Article in English | MEDLINE | ID: mdl-38855871

ABSTRACT

Human cytomegalovirus (HCMV) infection in infants infected in utero can lead to a variety of neurodevelopmental disorders. However, mechanisms underlying altered neurodevelopment in infected infants remain poorly understood. We have previously described a murine model of congenital HCMV infection in which murine CMV (MCMV) spreads hematogenously and establishes a focal infection in all regions of the brain of newborn mice, including the cerebellum. Infection resulted in disruption of cerebellar cortical development characterized by reduced cerebellar size and foliation. This disruption was associated with altered cell cycle progression of the granule cell precursors (GCPs), which are the progenitors that give rise to granule cells (GCs), the most abundant neurons in the cerebellum. In the current study, we have demonstrated that MCMV infection leads to prolonged GCP cell cycle, premature exit from the cell cycle, and reduced numbers of GCs resulting in cerebellar hypoplasia. Treatment with TNF-α neutralizing antibody partially normalized the cell cycle alterations of GCPs and altered cerebellar morphogenesis induced by MCMV infection. Collectively, our results argue that virus-induced inflammation altered the cell cycle of GCPs resulting in a reduced numbers of GCs and cerebellar cortical hypoplasia, thus providing a potential mechanism for altered neurodevelopment in fetuses infected with HCMV.


Subject(s)
Cell Cycle , Cerebellum , Cytomegalovirus Infections , Disease Models, Animal , Animals , Cytomegalovirus Infections/virology , Cytomegalovirus Infections/pathology , Mice , Cerebellum/virology , Cerebellum/pathology , Cerebellum/growth & development , Cerebellum/abnormalities , Female , Cytomegalovirus , Neural Stem Cells/virology , Muromegalovirus/physiology , Animals, Newborn , Humans , Neurons/virology , Tumor Necrosis Factor-alpha/metabolism , Developmental Disabilities , Nervous System Malformations
12.
JCI Insight ; 9(14)2024 Jun 17.
Article in English | MEDLINE | ID: mdl-38885315

ABSTRACT

IFN-signaling gene (ISG) expression scores are potential markers of inflammation with significance from cancer to genetic syndromes. In Aicardi Goutières Syndrome (AGS), a disorder of abnormal DNA and RNA metabolism, this score has potential as a diagnostic biomarker, although the approach to ISG calculation has not been standardized or validated. To optimize ISG calculation and validate ISG as a diagnostic biomarker, mRNA levels of 36 type I IFN response genes were quantified from 997 samples (including 334 AGS), and samples were randomized into training and test data sets. An independent validation cohort (n = 122) was also collected. ISGs were calculated using all potential combinations up to 6 genes. A 4-gene approach (IFI44L, IFI27, USP18, IFI6) was the best-performing model (AUC of 0.8872 [training data set], 0.9245 [test data set]). The majority of top-performing gene combinations included IFI44L. Performance of IFI44L alone was 0.8762 (training data set) and 0.9580 (test data set) by AUC. The top approaches were able to discriminate individuals with genetic interferonopathy from control samples. This study validates the context of use for the ISG score as a diagnostic biomarker and underscores the importance of IFI44L in diagnosis of genetic interferonopathies.


Subject(s)
Autoimmune Diseases of the Nervous System , Biomarkers , Nervous System Malformations , Signal Transduction , Humans , Biomarkers/metabolism , Male , Female , Nervous System Malformations/genetics , Nervous System Malformations/diagnosis , Signal Transduction/genetics , Autoimmune Diseases of the Nervous System/genetics , Autoimmune Diseases of the Nervous System/diagnosis , Child , Interferons/genetics , Interferons/metabolism , Ubiquitin Thiolesterase/genetics , Child, Preschool , Interferon Type I/genetics , Interferon Type I/metabolism , Membrane Proteins/genetics , Adult , Adolescent , RNA, Messenger/metabolism , RNA, Messenger/genetics , Tumor Suppressor Proteins
13.
Neurology ; 103(1): e209541, 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38857477

ABSTRACT

BACKGROUND AND OBJECTIVES: Aicardi Goutières syndrome (AGS) is type I interferonopathy characterized by severe neurologic impairment. Although many children with AGS demonstrate motor and expressive language deficits, the magnitude of receptive language impairment is uncharacterized. We sought to characterize cognitive function in AGS-affected children using assessment tools with reduced dependence on motor abilities and compare cognitive testing outcomes with overall severity and parental assessment of adaptive behavior. METHODS: We performed a cross-sectional study. Children were recruited as part of the Myelin Disorders Biorepository Project at the Children's Hospital of Philadelphia. We included individuals with a confirmed diagnosis of AGS. We administered the Leiter International Performance Scale, third edition (Leiter-3), and the Vineland Adaptive Behavior Scale, third edition (VABS-3), in the context of research encounters. Motor skills were categorized by AGS Severity Scale mobility levels. Descriptive statistics and Spearman's rank correlation were used to compare assessments. Mann-Whitney and Kruskal-Wallis tests with correction with Dunn's multiple comparison test were used to compare test performance between mobility groups. RESULTS: Cognitive and adaptive behavior performance was captured in 57 children. The mean age at encounters was 8.51 (SD 5.15) years. The median (IQR) Leiter-3 score was 51 (interquartile range [IQR] 60), with administration failure in 20 of 57 (35%) individuals. On the VABS-3, the Motor Domain (median 29, IQR 36.25) was more impacted than the Communication (median 50, IQR 52), Daily Living Skills (median 52, IQR 31), and Socialization (median 54, IQR 40) Domains (p < 0.0001). The AGS Scale correlated with VABS-3 (r = 0.86, p < 0.0001) and Leiter-3 (r = 0.87, p < 0.0001). There was correlation between VABS-3 Domains and Leiter-3 (r-range 0.83-0.97). Gross motor and fine motor categories, respectively, correlated with VABS-3 (H = 39.37, p < 0.0001; U = 63, p < 0.0001) and Leiter-3 (H = 40.43, p < 0.0001; U = 66, p < 0.0001). Within each gross motor and fine motor category of the AGS Scale, a subset of children scored within normal IQ range. DISCUSSION: Parental assessment of function by the VABS-3 correlated with directly assessed performance measures. Our data underscore the potential value of VABS-3 and Leiter-3 as tools to assess psychometric function in AGS. With a deeper understanding of our patients' abilities, we can better guide clinicians and families to provide appropriate support and personalized interventions to empower children with leukodystrophies to maximize their communication and educational potential.


Subject(s)
Autoimmune Diseases of the Nervous System , Nervous System Malformations , Humans , Female , Male , Cross-Sectional Studies , Child , Autoimmune Diseases of the Nervous System/physiopathology , Autoimmune Diseases of the Nervous System/diagnosis , Autoimmune Diseases of the Nervous System/complications , Autoimmune Diseases of the Nervous System/psychology , Child, Preschool , Nervous System Malformations/psychology , Nervous System Malformations/complications , Cognition/physiology , Adolescent , Neuropsychological Tests , Adaptation, Psychological , Motor Skills , Severity of Illness Index
14.
Immunity ; 57(7): 1696-1709.e10, 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38878770

ABSTRACT

Aicardi-Goutières syndrome (AGS) is an autoinflammatory disease characterized by aberrant interferon (IFN)-α production. The major cause of morbidity in AGS is brain disease, yet the primary source and target of neurotoxic IFN-α remain unclear. Here, we demonstrated that the brain was the primary source of neurotoxic IFN-α in AGS and confirmed the neurotoxicity of intracerebral IFN-α using astrocyte-driven Ifna1 misexpression in mice. Using single-cell RNA sequencing, we demonstrated that intracerebral IFN-α-activated receptor (IFNAR) signaling within cerebral endothelial cells caused a distinctive cerebral small vessel disease similar to that observed in individuals with AGS. Magnetic resonance imaging (MRI) and single-molecule ELISA revealed that central and not peripheral IFN-α was the primary determinant of microvascular disease in humans. Ablation of endothelial Ifnar1 in mice rescued microvascular disease, stopped the development of diffuse brain disease, and prolonged lifespan. These results identify the cerebral microvasculature as a primary mediator of IFN-α neurotoxicity in AGS, representing an accessible target for therapeutic intervention.


Subject(s)
Brain , Interferon-alpha , Microvessels , Nervous System Malformations , Receptor, Interferon alpha-beta , Animals , Humans , Mice , Interferon-alpha/metabolism , Brain/metabolism , Brain/pathology , Receptor, Interferon alpha-beta/metabolism , Receptor, Interferon alpha-beta/genetics , Microvessels/pathology , Nervous System Malformations/genetics , Autoimmune Diseases of the Nervous System/immunology , Endothelial Cells/metabolism , Mice, Knockout , Male , Female , Signal Transduction , Mice, Inbred C57BL , Astrocytes/metabolism , Disease Models, Animal
15.
BMJ Case Rep ; 17(6)2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38834308

ABSTRACT

Ehlers-Danlos syndrome is a group of connective tissue disorders with 14 subtypes, involving joint hyperlaxity, tissue fragility, hypertensive skin and other systemic organs with an incidence of 1 in 1 000 000 worldwide. We report a middle childhood female born of second degree consanguineous marriage with limping gait with muscle weakness, with normal development and IQ. Examination revealed microcornea, distal joint laxity of fingers and wrist, hypotonia and broad-based limping gait. Fracture dislocation right hip was managed by fixation. With the atypical neuroimaging finding of cerebellar vermis hypoplasia, exome sequencing was ordered and confirmed as Ehlers-Danlos syndrome (musculocontractural type-1). Hence, genetic counselling was done and prognosis of the child was explained.


Subject(s)
Cerebellum , Ehlers-Danlos Syndrome , Female , Humans , Cerebellum/abnormalities , Cerebellum/diagnostic imaging , Consanguinity , Developmental Disabilities , Ehlers-Danlos Syndrome/complications , Ehlers-Danlos Syndrome/diagnosis , Joint Instability/diagnostic imaging , Nervous System Malformations/complications , Child, Preschool
16.
Neuropediatrics ; 55(5): 337-340, 2024 10.
Article in English | MEDLINE | ID: mdl-38714209

ABSTRACT

Aicardi-Goutières syndrome (AGS) is a rare genetic early-onset progressive encephalopathy with variable clinical manifestations. The IFIH1 mutation has been confirmed to be responsible for type I interferon production and activation of the Janus kinase signaling pathway. We herein stress neurological observations and neuroimaging findings in a severe case report of an infant with AGS type 7 due to an IFIH1 mutation who was diagnosed in the first month of life. We also review neurological characteristics of IFIH1 mutations through recent literature.


Subject(s)
Autoimmune Diseases of the Nervous System , Interferon-Induced Helicase, IFIH1 , Mutation , Nervous System Malformations , Humans , Interferon-Induced Helicase, IFIH1/genetics , Interferon-Induced Helicase, IFIH1/immunology , Autoimmune Diseases of the Nervous System/genetics , Autoimmune Diseases of the Nervous System/diagnosis , Autoimmune Diseases of the Nervous System/complications , Autoimmune Diseases of the Nervous System/physiopathology , Nervous System Malformations/genetics , Nervous System Malformations/diagnostic imaging , Nervous System Malformations/complications , Infant , Male
17.
BMC Neurosci ; 25(1): 25, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38773385

ABSTRACT

INTRODUCTION: Incontinentia pigmenti (IP) is a rare neuroectodermal dysplasia caused by a defect in the IKBKG gene. The pathogenesis of central nervous system injury is believed to be related to microvascular ischemia. Currently, few treatment strategies are available for the inflammatory phase. MATERIALS AND METHODS: This retrospective descriptive analysis included the clinical data of 41 children with IP collected from 2007 to 2021 in Xi'an, China, comprising clinical characteristics, imaging findings, blood cell analysis, skin histopathology, and genetic data. RESULTS: Fourteen children (34%) aged 4 days to 5 months exhibited clinical signs and symptoms, including convulsions, delayed psychomotor development following neurological damage, and revealed significant MRI abnormalities, including ischemia, hypoxia, cerebral hypoperfusion, hemorrhage, encephalomalacia, and cerebral atrophy. Eight of the 24 patients (33%) presented with retinal vascular tortuosity and telangiectasis, accompanied by neovascularization and hemorrhage. Thirty-eight children (93%) had elevated eosinophils (mean: 3.63 ± 4.46 × 109), and 28 children (68%) had significantly elevated platelets (mean: 420.16 ± 179.43 × 109). Histopathology of skin revealed microvascular extravasation and vasodilation with perivascular and intravascular eosinophilic infiltration. CONCLUSION: Brain injury in IP occurs during infancy until 5 months of age, which is also the acute dermatitis phase accompanied by eosinophilia and an increased platelet count. This study provides evidence of microvascular damage to the skin and fundus during the inflammatory phase. The mechanism of microvascular damage may be similar to that in the brain.


Subject(s)
Incontinentia Pigmenti , Nervous System Malformations , Child, Preschool , Female , Humans , Infant , Infant, Newborn , Male , Brain/pathology , Brain/diagnostic imaging , Central Nervous System Diseases/congenital , Central Nervous System Diseases/genetics , China , East Asian People , Incontinentia Pigmenti/pathology , Incontinentia Pigmenti/genetics , Magnetic Resonance Imaging , Nervous System Malformations/genetics , Retrospective Studies
18.
Hum Mol Genet ; 33(18): 1555-1566, 2024 Sep 03.
Article in English | MEDLINE | ID: mdl-38796715

ABSTRACT

The cGAS-STING pathway detects cytosolic DNA and activates a signaling cascade that results in a type I interferon (IFN) response. The endoplasmic reticulum (ER)-associated exonuclease TREX1 suppresses cGAS-STING by eliminating DNA from the cytosol. Mutations that compromise TREX1 function are linked to autoinflammatory disorders, including systemic lupus erythematosus (SLE) and Aicardi-Goutières syndrome (AGS). Despite key roles in regulating cGAS-STING and suppressing excessive inflammation, the impact of many disease-associated TREX1 mutations-particularly those outside of the core catalytic domains-remains poorly understood. Here, we characterize a recessive AGS-linked TREX1 P61Q mutation occurring within the poorly characterized polyproline helix (PPII) motif. In keeping with its position outside of the catalytic core or ER targeting motifs, neither the P61Q mutation, nor aggregate proline-to-alanine PPII mutation, disrupts TREX1 exonuclease activity, subcellular localization, or cGAS-STING regulation in overexpression systems. Introducing targeted mutations into the endogenous TREX1 locus revealed that PPII mutations destabilize the protein, resulting in impaired exonuclease activity and unrestrained cGAS-STING activation. Overall, these results demonstrate that TREX1 PPII mutations, including P61Q, impair proper immune regulation and lead to autoimmune disease through TREX1 destabilization.


Subject(s)
Autoimmune Diseases of the Nervous System , Exodeoxyribonucleases , Membrane Proteins , Mutation , Nervous System Malformations , Nucleotidyltransferases , Phosphoproteins , Humans , Amino Acid Motifs , Autoimmune Diseases of the Nervous System/genetics , Autoimmune Diseases of the Nervous System/metabolism , Exodeoxyribonucleases/genetics , Exodeoxyribonucleases/metabolism , HEK293 Cells , Lupus Erythematosus, Systemic/genetics , Membrane Proteins/genetics , Membrane Proteins/metabolism , Nervous System Malformations/genetics , Nucleotidyltransferases/genetics , Nucleotidyltransferases/metabolism , Phosphoproteins/genetics , Phosphoproteins/metabolism , Signal Transduction/genetics
19.
Radiologie (Heidelb) ; 64(5): 410-419, 2024 May.
Article in German | MEDLINE | ID: mdl-38639917

ABSTRACT

CLINICAL ISSUE: Malformations of the central nervous system belong to the most common developmental disorders in humans. The clinical presentation of brain malformations is nonspecific including developmental delay, hypotonia, and/or epilepsy. The great heterogeneity concerning etiology, mechanisms of development and morphology is challenging for diagnosis and classification of brain malformations. Thereby recognizing specific malformations is essential for optimal patient management and prognostic evaluation. The aim of this article is to give an overview of several clinically relevant brain malformations occurring from different disrupted developmental processes in brain formation. STANDARD RADIOLOGICAL METHODS: Several brain malformations are already diagnosed during routine ultrasound in pregnancy. However pre- and postnatal magnetic resonance imaging remains the gold standard in detecting the partially subtle changes and to classify the malformations. METHODICAL INNOVATIONS: Advances in pre- and postnatal neuroimaging techniques and increasing investigation of genetic mechanisms underlying brain formation and its abnormalities have led to a better understanding of embryologic development and pathogeneses of brain malformations. CONCLUSION: Besides patient's history and clinical phenotype, neuroimaging plays a key role in diagnosis. Not always a specific diagnosis can be made, but neuroimaging patterns often enable a focused genetic testing and therefore are revolutionary for etiologic and prognostic assignment. Basic knowledge of brain development facilitates understanding and classifying of structural brain abnormalities.


Subject(s)
Brain , Female , Humans , Infant, Newborn , Pregnancy , Brain/abnormalities , Brain/diagnostic imaging , Brain/pathology , Magnetic Resonance Imaging , Nervous System Malformations/diagnosis , Nervous System Malformations/diagnostic imaging , Nervous System Malformations/genetics , Nervous System Malformations/pathology , Nervous System Malformations/classification , Neuroimaging/methods
SELECTION OF CITATIONS
SEARCH DETAIL