Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 565
Filter
1.
J Cell Biol ; 223(9)2024 Sep 02.
Article in English | MEDLINE | ID: mdl-38787349

ABSTRACT

Cell processes require precise regulation of actin polymerization that is mediated by plus-end regulatory proteins. Detailed mechanisms that explain plus-end dynamics involve regulators with opposing roles, including factors that enhance assembly, e.g., the formin mDia1, and others that stop growth (capping protein, CP). We explore IQGAP1's roles in regulating actin filament plus-ends and the consequences of perturbing its activity in cells. We confirm that IQGAP1 pauses elongation and interacts with plus ends through two residues (C756 and C781). We directly visualize the dynamic interplay between IQGAP1 and mDia1, revealing that IQGAP1 displaces the formin to influence actin assembly. Using four-color TIRF, we show that IQGAP1's displacement activity extends to formin-CP "decision complexes," promoting end-binding protein turnover at plus-ends. Loss of IQGAP1 or its plus-end activities disrupts morphology and migration, emphasizing its essential role. These results reveal a new role for IQGAP1 in promoting protein turnover on filament ends and provide new insights into how plus-end actin assembly is regulated in cells.


Subject(s)
Actin Cytoskeleton , Actins , Formins , ras GTPase-Activating Proteins , ras GTPase-Activating Proteins/metabolism , ras GTPase-Activating Proteins/genetics , Actins/metabolism , Humans , Actin Cytoskeleton/metabolism , Formins/metabolism , Actin Capping Proteins/metabolism , Actin Capping Proteins/genetics , Protein Binding , Animals , Cell Movement , HeLa Cells
2.
Int J Mol Sci ; 25(8)2024 Apr 21.
Article in English | MEDLINE | ID: mdl-38674130

ABSTRACT

IQGAP3 (IQ Motif Containing GTPase Activating Protein 3) is member of the IQGAP family of scaffold proteins, which are essential for assembling multiprotein complexes that coordinate various intracellular signaling pathways. Previous research has shown that IQGAP3 is overexpressed in psoriatic skin lesions. Given its involvement in processes like cell proliferation and chemokine signaling, we sought to explore its molecular role in driving the psoriatic phenotype of keratinocytes. By conducting transcriptome profiling of HaCaT keratinocytes, we identified numerous psoriasis-associated pathways that were affected when IQGAP3 was knocked down. These included alterations in NFkB signaling, EGFR signaling, activation of p38/MAPK and ERK1/ERK2, lipid metabolism, cytokine production, and the response to inflammatory cytokine stimulation. Real-time analysis further revealed changes in cell growth dynamics, including proliferation and wound healing. The balance between cell proliferation and apoptosis was altered, as were skin barrier functions and the production of IL-6 and IFNγ. Despite these significant findings, the diversity of the alterations observed in the knockdown cells led us to conclude that IQGAP3 may not be the best target for the therapeutic inhibition to normalize the phenotype of keratinocytes in psoriasis.


Subject(s)
Cell Proliferation , GTPase-Activating Proteins , Keratinocytes , Psoriasis , Humans , Keratinocytes/metabolism , Keratinocytes/pathology , Psoriasis/metabolism , Psoriasis/pathology , Psoriasis/genetics , ras GTPase-Activating Proteins/metabolism , ras GTPase-Activating Proteins/genetics , Signal Transduction , HaCaT Cells , Cytokines/metabolism , Apoptosis , Skin/metabolism , Skin/pathology , Cell Line , Gene Expression Profiling
3.
Am J Intellect Dev Disabil ; 129(3): 199-214, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38657965

ABSTRACT

This study aimed to describe the behavioral profile of individuals with SYNGAP1-ID. Parents/carers of 30 individuals aged 3-18 years old with a diagnosis of SYNGAP1-ID and 21 typically developing individuals completed the Vineland-3 Adaptive Behavior Scale and the Child Behavior Checklist. We found that those with SYNGAP1-ID showed fewer adaptive behaviors and higher levels of internalizing and externalizing behaviors across almost all domains compared to typically developing controls. There was some evidence that these differences were greatest in older children, and more apparent in those with co-occuring epilepsy. This characterization of the phenotype of SYNGAP1-ID significantly aids our understanding of the behavioral profile of this population and is a step towards the development of tailored interventions.


Subject(s)
Intellectual Disability , ras GTPase-Activating Proteins , Humans , Child , Male , Female , Child, Preschool , ras GTPase-Activating Proteins/genetics , Adolescent , Adaptation, Psychological/physiology , Child Behavior/physiology , Epilepsy
4.
Clin Cancer Res ; 30(10): 2206-2224, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38470497

ABSTRACT

PURPOSE: Microvascular invasion (MVI) is a major unfavorable prognostic factor for intrahepatic metastasis and postoperative recurrence of hepatocellular carcinoma (HCC). However, the intervention and preoperative prediction for MVI remain clinical challenges due to the absent precise mechanism and molecular marker(s). Herein, we aimed to investigate the mechanisms underlying vascular invasion that can be applied to clinical intervention for MVI in HCC. EXPERIMENTAL DESIGN: The histopathologic characteristics of clinical MVI+/HCC specimens were analyzed using multiplex immunofluorescence staining. The liver orthotopic xenograft mouse model and mechanistic experiments on human patient-derived HCC cell lines, including coculture modeling, RNA-sequencing, and proteomic analysis, were used to investigate MVI-related genes and mechanisms. RESULTS: IQGAP3 overexpression was correlated significantly with MVI status and reduced survival in HCC. Upregulation of IQGAP3 promoted MVI+-HCC cells to adopt an infiltrative vessel co-optive growth pattern and accessed blood capillaries by inducing detachment of activated hepatic stellate cells (HSC) from the endothelium. Mechanically, IQGAP3 overexpression contributed to HCC vascular invasion via a dual mechanism, in which IQGAP3 induced HSC activation and disruption of the HSC-endothelial interaction via upregulation of multiple cytokines and enhanced the trans-endothelial migration of MVI+-HCC cells by remodeling the cytoskeleton by sustaining GTPase Rac1 activity. Importantly, systemic delivery of IQGAP3-targeting small-interfering RNA nanoparticles disrupted the infiltrative vessel co-optive growth pattern and reduced the MVI of HCC. CONCLUSIONS: Our results revealed a plausible mechanism underlying IQGAP3-mediated microvascular invasion in HCC, and provided a potential target to develop therapeutic strategies to treat HCC with MVI.


Subject(s)
Carcinoma, Hepatocellular , Gene Expression Regulation, Neoplastic , Liver Neoplasms , Neoplasm Invasiveness , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/pathology , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Humans , Animals , Mice , Cell Line, Tumor , ras GTPase-Activating Proteins/genetics , ras GTPase-Activating Proteins/metabolism , Microvessels/pathology , Microvessels/metabolism , Male , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology , Neovascularization, Pathologic/metabolism , Xenograft Model Antitumor Assays , Female , Cell Proliferation , Prognosis , Hepatic Stellate Cells/metabolism , Hepatic Stellate Cells/pathology , Cell Movement/genetics
5.
Epilepsia ; 65(5): 1428-1438, 2024 May.
Article in English | MEDLINE | ID: mdl-38470175

ABSTRACT

OBJECTIVE: To delineate the comprehensive phenotypic spectrum of SYNGAP1-related disorder in a large patient cohort aggregated through a digital registry. METHODS: We obtained de-identified patient data from an online registry. Data were extracted from uploaded medical records. We reclassified all SYNGAP1 variants using American College of Medical Genetics criteria and included patients with pathogenic/likely pathogenic (P/LP) single nucleotide variants or microdeletions incorporating SYNGAP1. We analyzed neurodevelopmental phenotypes, including epilepsy, intellectual disability (ID), autism spectrum disorder (ASD), behavioral disorders, and gait dysfunction for all patients with respect to variant type and location within the SynGAP1 protein. RESULTS: We identified 147 patients (50% male, median age 8 years) with P/LP SYNGAP1 variants from 151 individuals with data available through the database. One hundred nine were truncating variants and 22 were missense. All patients were diagnosed with global developmental delay (GDD) and/or ID, and 123 patients (84%) were diagnosed with epilepsy. Of those with epilepsy, 73% of patients had GDD diagnosed before epilepsy was diagnosed. Other prominent features included autistic traits (n = 100, 68%), behavioral problems (n = 100, 68%), sleep problems (n = 90, 61%), anxiety (n = 35, 24%), ataxia or abnormal gait (n = 69, 47%), sensory problems (n = 32, 22%), and feeding difficulties (n = 69, 47%). Behavioral problems were more likely in those patients diagnosed with anxiety (odds ratio [OR] 3.6, p = .014) and sleep problems (OR 2.41, p = .015) but not necessarily those with autistic traits. Patients with variants in exons 1-4 were more likely to have the ability to speak in phrases vs those with variants in exons 5-19, and epilepsy occurred less frequently in patients with variants in the SH3 binding motif. SIGNIFICANCE: We demonstrate that the data obtained from a digital registry recapitulate earlier but smaller studies of SYNGAP1-related disorder and add additional genotype-phenotype relationships, validating the use of the digital registry. Access to data through digital registries broadens the possibilities for efficient data collection in rare diseases.


Subject(s)
Autism Spectrum Disorder , Epilepsy , Phenotype , ras GTPase-Activating Proteins , Humans , Male , Female , Child , Epilepsy/genetics , ras GTPase-Activating Proteins/genetics , Child, Preschool , Adolescent , Autism Spectrum Disorder/genetics , Autism Spectrum Disorder/epidemiology , Registries , Intellectual Disability/genetics , Intellectual Disability/epidemiology , Adult , Young Adult , Developmental Disabilities/genetics , Infant , Cohort Studies , Autistic Disorder/genetics
6.
Cell Rep ; 43(4): 113989, 2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38536816

ABSTRACT

Attachment of circulating tumor cells to the endothelial cells (ECs) lining blood vessels is a critical step in cancer metastatic colonization, which leads to metastatic outgrowth. Breast and prostate cancers are common malignancies in women and men, respectively. Here, we observe that ß1-integrin is required for human prostate and breast cancer cell adhesion to ECs under shear-stress conditions in vitro and to lung blood vessel ECs in vivo. We identify IQGAP1 and neural Wiskott-Aldrich syndrome protein (NWASP) as regulators of ß1-integrin transcription and protein expression in prostate and breast cancer cells. IQGAP1 and NWASP depletion in cancer cells decreases adhesion to ECs in vitro and retention in the lung vasculature and metastatic lung nodule formation in vivo. Mechanistically, NWASP and IQGAP1 act downstream of Cdc42 to increase ß1-integrin expression both via extracellular signal-regulated kinase (ERK)/focal adhesion kinase signaling at the protein level and by myocardin-related transcription factor/serum response factor (SRF) transcriptionally. Our results identify IQGAP1 and NWASP as potential therapeutic targets to reduce early metastatic dissemination.


Subject(s)
Integrin beta1 , Neoplasm Metastasis , Serum Response Factor , ras GTPase-Activating Proteins , Humans , Integrin beta1/metabolism , Integrin beta1/genetics , ras GTPase-Activating Proteins/metabolism , ras GTPase-Activating Proteins/genetics , Cell Line, Tumor , Serum Response Factor/metabolism , Male , Female , Prostatic Neoplasms/pathology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/genetics , Animals , Trans-Activators/metabolism , Cell Adhesion , Wiskott-Aldrich Syndrome Protein, Neuronal/metabolism , Wiskott-Aldrich Syndrome Protein, Neuronal/genetics , Breast Neoplasms/pathology , Breast Neoplasms/metabolism , Breast Neoplasms/genetics , Mice , Focal Adhesion Kinase 1/metabolism , Focal Adhesion Kinase 1/genetics , Gene Expression Regulation, Neoplastic , cdc42 GTP-Binding Protein/metabolism
7.
Aging (Albany NY) ; 16(7): 6054-6067, 2024 Mar 27.
Article in English | MEDLINE | ID: mdl-38546389

ABSTRACT

Lower-grade gliomas (GBMLGG) are common, fatal, and difficult-to-treat cancers. The current treatment choices have impressive efficacy constraints. As a result, the development of effective treatments and the identification of new therapeutic targets are urgent requirements. Disulfide metabolism is the cause of the non-apoptotic programmed cell death known as disulfideptosis, which was only recently discovered. The mRNA expression data and related clinical information of GBMLGG patients downloaded from public databases were used in this study to investigate the prognostic significance of genes involved in disulfideptosis. In the Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) cohort, our findings showed that many disulfidptosis-related genes were expressed differently in normal and GBMLGG tissues. It was discovered that IQ motif-containing GTPase-activating protein 1 (IQGAP1) is a key gene that influences the outcome of GBMLGG. Besides, a nomogram model was built to foresee the visualization of GBMLGG patients. In addition, in vivo and in vitro validation of IQGAP1's cancer-promoting function was done. In conclusion, we discovered a gene signature associated with disulfideptosis that can effectively predict OS in GBMLGG patients. As a result, treating disulfideptosis may be a viable alternative for GBMLGG patients.


Subject(s)
Brain Neoplasms , Disulfides , Glioma , Humans , Glioma/genetics , Glioma/pathology , Prognosis , Brain Neoplasms/genetics , Brain Neoplasms/pathology , ras GTPase-Activating Proteins/genetics , ras GTPase-Activating Proteins/metabolism , Gene Expression Regulation, Neoplastic , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Animals , Nomograms , Gene Expression Profiling , Female , Transcriptome , Male , Neoplasm Grading
8.
Science ; 383(6686): eadk1291, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38422154

ABSTRACT

SynGAP is an abundant synaptic GTPase-activating protein (GAP) critical for synaptic plasticity, learning, memory, and cognition. Mutations in SYNGAP1 in humans result in intellectual disability, autistic-like behaviors, and epilepsy. Heterozygous Syngap1-knockout mice display deficits in synaptic plasticity, learning, and memory and exhibit seizures. It is unclear whether SynGAP imparts structural properties at synapses independently of its GAP activity. Here, we report that inactivating mutations within the GAP domain do not inhibit synaptic plasticity or cause behavioral deficits. Instead, SynGAP modulates synaptic strength by physically competing with the AMPA-receptor-TARP excitatory receptor complex in the formation of molecular condensates with synaptic scaffolding proteins. These results have major implications for developing therapeutic treatments for SYNGAP1-related neurodevelopmental disorders.


Subject(s)
Cognition , Neuronal Plasticity , ras GTPase-Activating Proteins , Animals , Humans , Mice , Autistic Disorder/genetics , GTPase-Activating Proteins/genetics , Learning , Mice, Knockout , Neuronal Plasticity/genetics , ras GTPase-Activating Proteins/genetics , ras GTPase-Activating Proteins/metabolism , Catalysis
9.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 41(1): 25-31, 2024 Jan 10.
Article in Chinese | MEDLINE | ID: mdl-38171555

ABSTRACT

OBJECTIVE: To explore the early neurodevelopmental features of young children with SYNGAP1 variants and their genotype-phenotype correlation. METHODS: Young children with neurodevelopmental disorders (NDDs) (< 5 years old) who were referred to the Children's Hospital Affiliated to the Capital Institute of Pediatrics between January 2019 and July 2022 were selected as the study subjects. All children had undergone whole-exome sequencing, comprehensive pediatric neuropsychological assessment, familial segregation analysis, and pathogenicity classification. Meanwhile, young Chinese NDD children (< 5 years old) with pathogenic/likely pathogenic SYNGAP1 variants were retrieved from the literature, with information including detailed clinical and genetic testing, neurodevelopmental quotient (DQ) of the Children Neuropsychological and Behavior Scale-Revision 2016 (CNBS-R2016). Children who did not have a detailed DQ but had their developmental status assessed by a medical professional were also included. The correlation between neurodevelopmental severity, comorbidity and SYNGAP1 variants were summarized. RESULTS: Four young NDD children carrying SYNGAP1 variants were recruited (1 male and 3 females, with a mean age of 34.0 ± 18.2 months), among whom one harboring a novel variant (c.437C>G, p.S146*). Combined with 19 similar cases retrieved from the literature, 23 Chinese NDD young children were included in our study (8 males and 10 females, 5 with unknown sex, with a mean age of 37.1 ± 14.2 months). A loss of function (LOF) variant was found in 19 (82.6%) children. All of the children had presented global developmental delay (GDD) before the age of two. In addition, 16 (69.6%) had seizure/epilepsy at the age of 27.0 ± 12.1 months, among whom 15 had occurred independent of the global developmental delay. Myoclonic and absence were common types of seizures. Compared with those with variants of exons 8 to 15, the severity of developmental delay was milder among children with variants in exons 1 to 5. CONCLUSION: The early neurodevelopment features of the SYNGAP1 variants for young children (< 5 years old) have included global developmental delay and seizure/epilepsy. All of the children may present GDD before the age of two. The severity of developmental delay may be related to the type and location of the SYNGAP1 variants.


Subject(s)
Epilepsy , Neurodevelopmental Disorders , Child, Preschool , Female , Humans , Infant , Male , Epilepsy/genetics , Genetic Testing , Genotype , Neurodevelopmental Disorders/genetics , ras GTPase-Activating Proteins/genetics , Seizures/genetics
10.
Epilepsy Behav ; 151: 109636, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38232560

ABSTRACT

Atypical absence seizures are generalized non-convulsive seizures that often occur in children with cognitive impairment. They are common in refractory epilepsy and have been recognized as one of the hallmarks of developmental epileptic encephalopathies. Notably, pathogenic variants associated with AAS, such as GABRG2, GABRG3, SLC6A1, CACNB4, SCN8A, and SYNGAP1, are also linked to developmental epileptic encephalopathies. Atypical absences differ from typical absences in that they are frequently drug-resistant and the prognosis is dependent on the etiology or related epileptic syndromes. To improve clinicians' understanding of atypical absences and provide novel perspectives for clinical treatment, we have reviewed the electro-clinical characteristics, etiologies, treatment, and prognosis of atypical absences, with a focus on the etiology of advancements in gene variants, shedding light on potential avenues for improved clinical management.


Subject(s)
Drug Resistant Epilepsy , Epilepsy, Absence , Epilepsy, Generalized , Humans , Child , Epilepsy, Absence/genetics , Epilepsy, Absence/drug therapy , Seizures , ras GTPase-Activating Proteins/genetics , Electroencephalography
11.
Lab Med ; 55(2): 204-208, 2024 Mar 07.
Article in English | MEDLINE | ID: mdl-37467311

ABSTRACT

OBJECTIVE: Intellectual developmental disorder (IDD) type 5 is an autosomal dominant (AD) disorder and is characterized by intellectual disability (ID), psychomotor developmental delay, variable autism phenotypes, microcephaly, and seizure. IDD can be caused by mutations in the SYNGAP1 gene, which encodes a Ras GTPase-activating protein. This study revealed a novel de novo nonsense variant in SYNGAP1. The identification of such variants is essential for genetic counseling in patients and their families. METHODS: Exome sequencing implicated the causative variant. Sanger sequencing and cosegregation analyses were used to confirm the variant. Multiple in silico analysis tools were applied to interpret the variant using the American College of Medical Genetics and Genomics and the Association for Molecular Pathology guidelines. RESULTS: The de novo NM_006772.3(SYNGAP1):c.3685C>T variant was identified in an 11-year-old boy with severe intellectual disability, neurodevelopmental delay, speech disorder, ataxia, specific dysmorphic facial features, and aggressive behavior. CONCLUSION: The current study findings expand the existing knowledge of variants in SYNGAP1 that have been previously associated with nonsyndromic intellectual disability and autism, extending the spectrum of phenotypes associated with this gene. The data have implications for genetic diagnosis and counseling in similar phenotypic presentations.


Subject(s)
Intellectual Disability , ras GTPase-Activating Proteins , Child , Humans , Male , Genomics , Intellectual Disability/diagnosis , Intellectual Disability/genetics , Iran , Mutation , Phenotype , ras GTPase-Activating Proteins/genetics
12.
J Appl Genet ; 65(2): 303-308, 2024 May.
Article in English | MEDLINE | ID: mdl-37987971

ABSTRACT

Noonan syndrome (NS; OMIM 163950) is an autosomal dominant RASopathy with variable clinical expression and genetic heterogeneity. Clinical manifestations include characteristic facial features, short stature, and cardiac anomalies. Variants in protein-tyrosine phosphatase, non-receptor-type 11 (PTPN11), encoding SHP-2, account for about half of NS patients, SOS1 in approximately 13%, RAF1 in 10%, and RIT1 each in 9%. Other genes have been reported to cause NS in less than 5% of cases including SHOC2, RASA2, LZTR1, SPRED2, SOS2, CBL, KRAS, NRAS, MRAS, PRAS, BRAF, PPP1CB, A2ML1, MAP2K1, and CDC42. Several additional genes associated with a Noonan syndrome-like phenotype have been identified. Clinical presentation and variants in patients with Noonan syndrome are this study's objectives. We performed Sanger sequencing of PTPN11 hotspot (exons 3, 8, and 13). We report molecular analysis of 61 patients with NS phenotype belonging to 58 families. We screened for hotspot variants (exons 3, 8, and 13) in PTPN11 gene by Sanger sequencing. Twenty-seven patients were carrying heterozygous pathogenic variants of PTPN11 gene with a similar frequency (41.4%) compared to the literature. Our findings expand the variant spectrum of Moroccan patients with NS phenotype in whom the analysis of hotspot variants showed a high frequency of exons 3 and 8. This screening test allowed us to establish a molecular diagnosis in almost half of the patients with a good benefit-cost ratio, with appropriate management and genetic counseling.


Subject(s)
Noonan Syndrome , Protein Tyrosine Phosphatase, Non-Receptor Type 11 , alpha-Macroglobulins , Humans , Exons , Intracellular Signaling Peptides and Proteins/genetics , Mutation , Noonan Syndrome/genetics , Noonan Syndrome/diagnosis , Noonan Syndrome/pathology , Phenotype , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , ras GTPase-Activating Proteins/genetics , Repressor Proteins/genetics , Transcription Factors/genetics
13.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-1009348

ABSTRACT

OBJECTIVE@#To explore the early neurodevelopmental features of young children with SYNGAP1 variants and their genotype-phenotype correlation.@*METHODS@#Young children with neurodevelopmental disorders (NDDs) (< 5 years old) who were referred to the Children's Hospital Affiliated to the Capital Institute of Pediatrics between January 2019 and July 2022 were selected as the study subjects. All children had undergone whole-exome sequencing, comprehensive pediatric neuropsychological assessment, familial segregation analysis, and pathogenicity classification. Meanwhile, young Chinese NDD children (< 5 years old) with pathogenic/likely pathogenic SYNGAP1 variants were retrieved from the literature, with information including detailed clinical and genetic testing, neurodevelopmental quotient (DQ) of the Children Neuropsychological and Behavior Scale-Revision 2016 (CNBS-R2016). Children who did not have a detailed DQ but had their developmental status assessed by a medical professional were also included. The correlation between neurodevelopmental severity, comorbidity and SYNGAP1 variants were summarized.@*RESULTS@#Four young NDD children carrying SYNGAP1 variants were recruited (1 male and 3 females, with a mean age of 34.0 ± 18.2 months), among whom one harboring a novel variant (c.437C>G, p.S146*). Combined with 19 similar cases retrieved from the literature, 23 Chinese NDD young children were included in our study (8 males and 10 females, 5 with unknown sex, with a mean age of 37.1 ± 14.2 months). A loss of function (LOF) variant was found in 19 (82.6%) children. All of the children had presented global developmental delay (GDD) before the age of two. In addition, 16 (69.6%) had seizure/epilepsy at the age of 27.0 ± 12.1 months, among whom 15 had occurred independent of the global developmental delay. Myoclonic and absence were common types of seizures. Compared with those with variants of exons 8 to 15, the severity of developmental delay was milder among children with variants in exons 1 to 5.@*CONCLUSION@#The early neurodevelopment features of the SYNGAP1 variants for young children (< 5 years old) have included global developmental delay and seizure/epilepsy. All of the children may present GDD before the age of two. The severity of developmental delay may be related to the type and location of the SYNGAP1 variants.


Subject(s)
Child, Preschool , Female , Humans , Infant , Male , Epilepsy/genetics , Genetic Testing , Genotype , Neurodevelopmental Disorders/genetics , ras GTPase-Activating Proteins/genetics , Seizures/genetics
14.
PLoS Genet ; 19(12): e1011049, 2023 Dec.
Article in English | MEDLINE | ID: mdl-38091360

ABSTRACT

Neural regulation of sleep and metabolic homeostasis are critical in many aspects of human health. Despite extensive epidemiological evidence linking sleep dysregulation with obesity, diabetes, and metabolic syndrome, little is known about the neural and molecular basis for the integration of sleep and metabolic function. The RAS GTPase-activating gene Neurofibromin (Nf1) has been implicated in the regulation of sleep and metabolic rate, raising the possibility that it serves to integrate these processes, but the effects on sleep consolidation and physiology remain poorly understood. A key hallmark of sleep depth in mammals and flies is a reduction in metabolic rate during sleep. Here, we examine multiple measures of sleep quality to determine the effects of Nf1 on sleep-dependent changes in arousal threshold and metabolic rate. Flies lacking Nf1 fail to suppress metabolic rate during sleep, raising the possibility that loss of Nf1 prevents flies from integrating sleep and metabolic state. Sleep of Nf1 mutant flies is fragmented with a reduced arousal threshold in Nf1 mutants, suggesting Nf1 flies fail to enter deep sleep. The effects of Nf1 on sleep can be localized to a subset of neurons expressing the GABAA receptor Rdl. Sleep loss has been associated with changes in gut homeostasis in flies and mammals. Selective knockdown of Nf1 in Rdl-expressing neurons within the nervous system increases gut permeability and reactive oxygen species (ROS) in the gut, raising the possibility that loss of sleep quality contributes to gut dysregulation. Together, these findings suggest Nf1 acts in GABA-sensitive neurons to modulate sleep depth in Drosophila.


Subject(s)
Drosophila Proteins , Nerve Tissue Proteins , ras GTPase-Activating Proteins , Sleep , Animals , Drosophila melanogaster , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , ras GTPase-Activating Proteins/genetics , ras GTPase-Activating Proteins/metabolism , Sleep Duration , Male , Brain/metabolism , Intestines/metabolism , Diet
15.
Nat Neurosci ; 26(12): 2090-2103, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37946050

ABSTRACT

Genes involved in synaptic function are enriched among those with autism spectrum disorder (ASD)-associated rare genetic variants. Dysregulated cortical neurogenesis has been implicated as a convergent mechanism in ASD pathophysiology, yet it remains unknown how 'synaptic' ASD risk genes contribute to these phenotypes, which arise before synaptogenesis. Here, we show that the synaptic Ras GTPase-activating (RASGAP) protein 1 (SYNGAP1, a top ASD risk gene) is expressed within the apical domain of human radial glia cells (hRGCs). In a human cortical organoid model of SYNGAP1 haploinsufficiency, we find dysregulated cytoskeletal dynamics that impair the scaffolding and division plane of hRGCs, resulting in disrupted lamination and accelerated maturation of cortical projection neurons. Additionally, we confirmed an imbalance in the ratio of progenitors to neurons in a mouse model of Syngap1 haploinsufficiency. Thus, SYNGAP1-related brain disorders may arise through non-synaptic mechanisms, highlighting the need to study genes associated with neurodevelopmental disorders (NDDs) in diverse human cell types and developmental stages.


Subject(s)
Autism Spectrum Disorder , Neurodevelopmental Disorders , Animals , Mice , Humans , Autism Spectrum Disorder/genetics , ras GTPase-Activating Proteins/genetics , Neurodevelopmental Disorders/genetics , Phenotype , Neurogenesis/genetics
17.
Medicina (Kaunas) ; 59(10)2023 Oct 17.
Article in English | MEDLINE | ID: mdl-37893562

ABSTRACT

Background and Objectives: This study aims to identify the minor allele of the single nucleotide polymorphisms (SNPs) DAB2IP rs7025486, IL6R rs2228145, CDKN2BAS rs10757278, LPA rs3798220, LRP1 rs1466535, and SORT1 rs599839 in order to assess the risk of abdominal aortic aneurysm (AAA) formation and define the linkage among these SNPs. Materials and Methods: A case-control study with AAA patients (AAA group) and non-AAA controls (control group) was carried out in a study population. DNA was isolated from whole blood samples; the SNPs were amplified using PCR and sequenced. Results: In the AAA group of 148 patients, 87.2% of the patients were male, 64.2% had a history of smoking, and 18.2% had relatives with AAA. The mean ± SD of age, BMI, and aneurysmal diameter in the AAA group were 74.8 ± 8.3 years, 27.6 ± 4.6 kg/m2, and 56.2 ± 11.8 mm, respectively. In comparison with 50 non-AAA patients, there was a significantly elevated presence of the SNPs DAB2IP rs7025486[A], CDKN2BAS rs10757278[G], and SORT1 rs599839[G] in the AAA group (p-values 0.040, 0.024, 0.035, respectively), while LPA rs3798220[C] was significantly higher in the control group (p = 0.049). A haplotype investigation showed that the SNPs DAB2IP, CDKN2BAS, and IL6R rs2228145[C] were significantly elevated in the AAA group (p = 0.037, 0.037, and 0.046) with minor allele frequencies (MAF) of 25.5%, 10.6%, and 15.4%, respectively. Only DAB2IP and CDKN2BAS showed significantly higher occurrences of a mutation (p = 0.028 and 0.047). Except for LPA, all SNPs were associated with a large aortic diameter in AAA (p < 0.001). Linkage disequilibrium detection showed that LPA to DAB2IP, to IL6R, to CDKN2BAS, and to LRP1 rs1466535[T] had D' values of 70.9%, 80.4%, 100%, and 100%, respectively. IL6R to LRP1 and to SORT1 had values for the coefficient of determination (r2) of 3.9% and 2.2%, respectively. Conclusions: In the investigated study population, the SNPs CDKN2BAS rs10757278, LPA rs3798220, SORT1 rs599839, DAB2IP rs7025486, and IL6R rs2228145 were associated with the development of abdominal aortic aneurysms. Individuals with risk factors for atherosclerosis and/or a family history of AAA should be evaluated using genetic analysis.


Subject(s)
Aortic Aneurysm, Abdominal , Genetic Predisposition to Disease , Humans , Male , Aged , Aged, 80 and over , Female , Case-Control Studies , Polymorphism, Single Nucleotide/genetics , Aortic Aneurysm, Abdominal/genetics , Risk Factors , Inflammation , Apoptosis , Cholesterol , ras GTPase-Activating Proteins/genetics
18.
Funct Integr Genomics ; 23(4): 326, 2023 Oct 25.
Article in English | MEDLINE | ID: mdl-37880458

ABSTRACT

Renal cell carcinoma (RCC) is the most lethal of the urologic malignancies. We previously discovered that DAB2IP, a novel Ras GTPase-activating protein, was frequently epigenetically silenced in RCC, and DAB2IP loss was correlated with the overall survival of RCC patients. In this study, we determined the biological functions of DAB2IP in clear cell RCC (ccRCC) and its potential mechanisms of action. Correlations between DAB2IP expression level and ccRCC tumor size and patient survival were analyzed, and the results showed that ccRCC patients with high DAB2IP mRNA level exhibited smaller tumor size and better survival than the patients with low DAB2IP. Compared to control, DAB2IP knockdown significantly increased cell proliferation, promoted cell cycle progression in G1/S phase, and decreased p27 expression. Mechanism studies demonstrated that loss of DAB2IP promoted p27 protein phosphorylation, cytosolic sequestration, and subsequently ubiquitination-mediated degradation in ccRCC cells. Further studies confirmed that the proline-rich domain in C terminal (CPR) of DAB2IP suppressed AKT phosphorylation and p27 phosphorylation on S10. Hence, DAB2IP is essential for p27 protein stabilization in ccRCC, which is at less partly mediated by PI3K/AKT signaling pathway.


Subject(s)
Carcinoma, Renal Cell , Kidney Neoplasms , Humans , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/pathology , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction/genetics , Cell Proliferation/genetics , Kidney Neoplasms/genetics , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Cell Line, Tumor , ras GTPase-Activating Proteins/genetics , ras GTPase-Activating Proteins/metabolism
19.
DNA Cell Biol ; 42(10): 617-637, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37610843

ABSTRACT

Recent studies have shown that several members of the G-protein-coupled receptors (GPCR) superfamily play crucial roles in the maintenance of ion-water homeostasis of the sperm and Sertoli cells, development of the germ cells, formation of the blood barrier, and maturation of sperm. The GPCR, guanyl-nucleotide exchange factor, membrane traffic protein, and small GTPase genes were analyzed by microarray and bioinformatics (3513 sperm and Sertoli cell genes). In the microarray analyses of three human cases with different nonobstructive azoospermia sperm, the expression of GOLGA8IP, OR2AT4, PHKA1, A2M, OR56A1, SEMA3G, LRRC17, APP, ARHGAP33, RABGEF1, NPY2R, GHRHR, LTB4R2, GRIK5, OR6K6, NAPG, OR6C65, VPS35, FPR3, and ARL4A was upregulated, while expression of MARS, SIRPG, OGFR, GPR150, LRRK1, and NGEF was downregulated. There was an increase in GBP3, GBP3, TNF, TGFB3, and CLTC expression in the Sertoli cells of three human cases with NOA, whereas expression of PAQR4, RRAGD, RAC2, SERPINB8, IRPB1, MRGPRF, RASA2, SIRPG, RGS2, RAP2A, RAB2B, ARL17, SERINC4, XIAP, DENND4C, ANKRA2, CSTA, STX18, and SNAP23 were downregulated. A combined analysis of Enrich Shiny Gene Ontology (GO), STRING, and Cytoscape was used to predict proteins' molecular interactions and then to recognize master pathways. Functional enrichment analysis showed that the biological process (BP), regulation of protein metabolic process, regulation of small GTPase-mediated signal transduction were significantly expressed in up-/downregulated differentially expressed genes (DEGs) in sperm. In molecular function (MF) experiments of DEGs that were up-/downregulated, it was found that GPCR activity, guanyl ribonucleotide binding, GTPase activity and nucleoside-triphosphatase activity were overexpressed. An analysis of GO enrichment findings of Sertoli cells showed BP and MF to be common DEGs. When these gene mutations have been validated, they can be used to create new GPCR antagonists or agonists that are receptor-selective.


Subject(s)
Azoospermia , Monomeric GTP-Binding Proteins , Humans , Male , Testis/metabolism , Azoospermia/genetics , Azoospermia/metabolism , Semen/metabolism , Gene Expression , Monomeric GTP-Binding Proteins/genetics , Monomeric GTP-Binding Proteins/metabolism , GTP-Binding Proteins/genetics , ras GTPase-Activating Proteins/genetics , Ankyrins/genetics , Ankyrins/metabolism , ADP-Ribosylation Factors/genetics , ADP-Ribosylation Factors/metabolism , rap GTP-Binding Proteins/genetics , rap GTP-Binding Proteins/metabolism
20.
J Neurosci ; 43(35): 6212-6226, 2023 08 30.
Article in English | MEDLINE | ID: mdl-37558489

ABSTRACT

SYNGAP1 haploinsufficiency in humans leads to severe neurodevelopmental disorders characterized by intellectual disability, autism, epilepsy, and sensory processing deficits. However, the circuit mechanisms underlying these disorders are not well understood. In mice, a decrease of SynGAP levels results in cognitive deficits by interfering with the development of excitatory glutamatergic connections. Recent evidence suggests that SynGAP also plays a crucial role in the development and function of GABAergic inhibitory interneurons. Nevertheless, it remains uncertain whether and to what extent the expression of SYNGAP1 in inhibitory interneurons contributes to cortical circuit function and related behaviors. The activity of cortical neurons has not been measured simultaneously with behavior. To address these gaps, we recorded from layer 2/3 neurons in the primary whisker somatosensory cortex (wS1) of mice while they learned to perform a whisker tactile detection task. Our results demonstrate that mice with interneuron-specific SYNGAP1 haploinsufficiency exhibit learning deficits characterized by heightened behavioral responses in the absence of relevant sensory input and premature responses to unrelated sensory stimuli not associated with reward acquisition. These behavioral deficits are accompanied by specific circuit abnormalities within wS1. Interneuron-specific SYNGAP1 haploinsufficiency increases detrimental neuronal correlations directly related to task performance and enhances responses to irrelevant sensory stimuli unrelated to the reward acquisition. In summary, our findings indicate that a reduction of SynGAP in inhibitory interneurons impairs sensory representation in the primary sensory cortex by disrupting neuronal correlations, which likely contributes to the observed cognitive deficits in mice with pan-neuronal SYNGAP1 haploinsufficiency.SIGNIFICANCE STATEMENT SYNGAP1 haploinsufficiency leads to severe neurodevelopmental disorders. The exact nature of neural circuit dysfunction caused by SYNGAP1 haploinsufficiency remains poorly understood. SynGAP plays a critical role in the function of GABAergic inhibitory interneurons as well as glutamatergic pyramidal neurons in the neocortex. Whether and how decreasing SYNGAP1 level in inhibitory interneurons disrupts a behaviorally relevant circuit remains unclear. We measure neural activity and behavior in mice learning a perceptual task. Mice with interneuron-targeted disruption of SYNGAP1 display increased detrimental neuronal correlations and elevated responses to irrelevant sensory inputs, which are related to impaired task performance. These results show that cortical interneuron dysfunction contributes to sensory deficits in SYNGAP1 haploinsufficiency with important implications for identifying therapeutic targets.


Subject(s)
Neocortex , Humans , Mice , Animals , Neocortex/metabolism , Interneurons/physiology , Neurons/physiology , Pyramidal Cells/metabolism , Learning , ras GTPase-Activating Proteins/genetics , ras GTPase-Activating Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...