Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Hum Genet ; 141(10): 1595-1613, 2022 Oct.
Article in English | MEDLINE | ID: mdl-34549350

ABSTRACT

Whole-exome and whole-genome sequencing studies in autism spectrum disorder (ASD) have identified hundreds of thousands of exonic variants. Only a handful of them, primarily loss-of-function variants, have been shown to increase the risk for ASD, while the contributory roles of other variants, including most missense variants, remain unknown. New approaches that combine tissue-specific molecular profiles with patients' genetic data can thus play an important role in elucidating the functional impact of exonic variation and improve understanding of ASD pathogenesis. Here, we integrate spatio-temporal gene co-expression networks from the developing human brain and protein-protein interaction networks to first reach accurate prioritization of ASD risk genes based on their connectivity patterns with previously known high-confidence ASD risk genes. We subsequently integrate these gene scores with variant pathogenicity predictions to further prioritize individual exonic variants based on the positive-unlabeled learning framework with gene- and variant-score calibration. We demonstrate that this approach discriminates among variants between cases and controls at the high end of the prediction range. Finally, we experimentally validate our top-scoring de novo mutation NP_001243143.1:p.Phe309Ser in the sodium/potassium-transporting ATPase ATP1A3 to disrupt protein binding with different partners.


Subject(s)
Autism Spectrum Disorder , Autistic Disorder , Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/metabolism , Autism Spectrum Disorder/genetics , Autistic Disorder/genetics , Genetic Predisposition to Disease , Humans , Mutation , Potassium/metabolism , Sodium/metabolism , Sodium-Potassium-Exchanging ATPase/genetics
3.
Cell Rep ; 36(9): 109631, 2021 08 31.
Article in English | MEDLINE | ID: mdl-34469739

ABSTRACT

Alternative splicing plays an important role in brain development, but its global contribution to human neurodevelopmental diseases (NDDs) requires further investigation. Here we examine the relationships between splicing isoform expression in the brain and de novo loss-of-function mutations from individuals with NDDs. We analyze the full-length isoform transcriptome of the developing human brain and observe differentially expressed isoforms and isoform co-expression modules undetectable by gene-level analyses. These isoforms are enriched in loss-of-function mutations and microexons, are co-expressed with a unique set of partners, and have higher prenatal expression. We experimentally test the effect of splice-site mutations and demonstrate exon skipping in five NDD risk genes, including SCN2A, DYRK1A, and BTRC. Our results suggest that the splice site mutation in BTRC reduces translational efficiency, likely affecting Wnt signaling through impaired degradation of ß-catenin. We propose that functional effects of mutations should be investigated at the isoform- rather than gene-level resolution.


Subject(s)
Alternative Splicing , Autistic Disorder/genetics , Brain/growth & development , Gene Expression Profiling , Mutation , Transcriptome , Autistic Disorder/physiopathology , Autistic Disorder/psychology , Case-Control Studies , Gene Expression Regulation, Developmental , Genetic Predisposition to Disease , HeLa Cells , Humans , NAV1.2 Voltage-Gated Sodium Channel/genetics , Protein Serine-Threonine Kinases/genetics , Protein-Tyrosine Kinases/genetics , beta-Transducin Repeat-Containing Proteins/genetics , Dyrk Kinases
4.
Mol Psychiatry ; 26(12): 7560-7580, 2021 12.
Article in English | MEDLINE | ID: mdl-34433918

ABSTRACT

Reciprocal deletion and duplication of the 16p11.2 region is the most common copy number variation (CNV) associated with autism spectrum disorders. We generated cortical organoids from skin fibroblasts of patients with 16p11.2 CNV to investigate impacted neurodevelopmental processes. We show that organoid size recapitulates macrocephaly and microcephaly phenotypes observed in the patients with 16p11.2 deletions and duplications. The CNV dosage affects neuronal maturation, proliferation, and synapse number, in addition to its effect on organoid size. We demonstrate that 16p11.2 CNV alters the ratio of neurons to neural progenitors in organoids during early neurogenesis, with a significant excess of neurons and depletion of neural progenitors observed in deletions. Transcriptomic and proteomic profiling revealed multiple pathways dysregulated by the 16p11.2 CNV, including neuron migration, actin cytoskeleton, ion channel activity, synaptic-related functions, and Wnt signaling. The level of the active form of small GTPase RhoA was increased in both, deletions and duplications. Inhibition of RhoA activity rescued migration deficits, but not neurite outgrowth. This study provides insights into potential neurobiological mechanisms behind the 16p11.2 CNV during neocortical development.


Subject(s)
Autism Spectrum Disorder , Autistic Disorder , Autism Spectrum Disorder/genetics , Autistic Disorder/genetics , Brain , Chromosome Deletion , Chromosomes, Human, Pair 16/genetics , DNA Copy Number Variations/genetics , Humans , Neurogenesis/genetics , Organoids , Proteomics
5.
Mol Psychiatry ; 26(7): 3586-3613, 2021 07.
Article in English | MEDLINE | ID: mdl-33727673

ABSTRACT

E3-ubiquitin ligase Cullin3 (Cul3) is a high confidence risk gene for autism spectrum disorder (ASD) and developmental delay (DD). To investigate how Cul3 mutations impact brain development, we generated a haploinsufficient Cul3 mouse model using CRISPR/Cas9 genome engineering. Cul3 mutant mice exhibited social and cognitive deficits and hyperactive behavior. Brain MRI found decreased volume of cortical regions and changes in many other brain regions of Cul3 mutant mice starting from early postnatal development. Spatiotemporal transcriptomic and proteomic profiling of embryonic, early postnatal and adult brain implicated neurogenesis and cytoskeletal defects as key drivers of Cul3 functional impact. Specifically, dendritic growth, filamentous actin puncta, and spontaneous network activity were reduced in Cul3 mutant mice. Inhibition of small GTPase RhoA, a molecular substrate of Cul3 ligase, rescued dendrite length and network activity phenotypes. Our study identified defects in neuronal cytoskeleton and Rho signaling as the primary targets of Cul3 mutation during brain development.


Subject(s)
Autism Spectrum Disorder , Autistic Disorder , Animals , Autism Spectrum Disorder/genetics , Cullin Proteins/genetics , Cytoskeleton , Germ Cells , Haploinsufficiency/genetics , Mice , Neurogenesis/genetics , Proteomics
6.
Nat Commun ; 11(1): 5918, 2020 11 20.
Article in English | MEDLINE | ID: mdl-33219223

ABSTRACT

Identifying pathogenic variants and underlying functional alterations is challenging. To this end, we introduce MutPred2, a tool that improves the prioritization of pathogenic amino acid substitutions over existing methods, generates molecular mechanisms potentially causative of disease, and returns interpretable pathogenicity score distributions on individual genomes. Whilst its prioritization performance is state-of-the-art, a distinguishing feature of MutPred2 is the probabilistic modeling of variant impact on specific aspects of protein structure and function that can serve to guide experimental studies of phenotype-altering variants. We demonstrate the utility of MutPred2 in the identification of the structural and functional mutational signatures relevant to Mendelian disorders and the prioritization of de novo mutations associated with complex neurodevelopmental disorders. We then experimentally validate the functional impact of several variants identified in patients with such disorders. We argue that mechanism-driven studies of human inherited disease have the potential to significantly accelerate the discovery of clinically actionable variants.


Subject(s)
Amino Acid Substitution/genetics , Computational Biology/methods , Genetic Predisposition to Disease , Software , Genome, Human , Humans , Models, Statistical , Mutation , Phenotype , Proteins/genetics
7.
Methods Mol Biol ; 1613: 371-402, 2017.
Article in English | MEDLINE | ID: mdl-28849569

ABSTRACT

Recent advances in genome sequencing and "omics" technologies are opening new opportunities for improving diagnosis and treatment of human diseases. The precision medicine initiative in particular aims at developing individualized treatment options that take into account individual variability in genes and environment of each person. Systems biology approaches that group genes, transcripts and proteins into functionally meaningful networks will play crucial role in the future of personalized medicine. They will allow comparison of healthy and disease-affected tissues and organs from the same individual, as well as between healthy and disease-afflicted individuals. However, the field faces a multitude of challenges ranging from data integration to statistical and combinatorial issues in data analyses. This chapter describes computational approaches developed by us and the others to tackle challenges in tissue-specific network analyses, with the main focus on psychiatric diseases.


Subject(s)
Gene Regulatory Networks , Mental Disorders/metabolism , Systems Biology/methods , Humans , Mental Disorders/genetics , Mutation , Organ Specificity , Precision Medicine , Protein Interaction Mapping
8.
J Neurochem ; 139(1): 11-21, 2016 10.
Article in English | MEDLINE | ID: mdl-27385439

ABSTRACT

The importance of death receptor (DR) signaling in embryonic development and physiological homeostasis is well established, as is the existence of several molecules that modulate DRs function, among them Fas Apoptotis Inhibitory Molecules. Although FAIM1, FAIM2, and FAIM3 inhibit Fas-induced cell death, they are not structurally related, nor do they share expression patterns. Moreover, they inhibit apoptosis through completely different mechanisms. FAIM1 and FAIM2 protect neurons from DR-induced apoptosis and are involved in neurite outgrowth and neuronal plasticity. FAIM1 inhibits Fas ligand- and tumor necrosis factor alpha-induced apoptosis by direct interaction with Fas receptor and through the stabilization of levels of X-linked inhibitor of apoptosis protein, a potent anti-apoptotic protein that inhibits caspases. Low FAIM1 levels are found in Alzheimer's disease, thus sensitizing neurons to tumor necrosis factor alpha and prompting neuronal loss. FAIM2 protects from Fas by direct interaction with Fas receptor, as well as by modulating calcium release at the endoplasmic reticulum through interaction with Bcl-xL. Several studies prove the role of FAIM2 in diseases of the nervous system, such as ischemia, bacterial meningitis, and neuroblastoma. The less characterized member of the FAIM family is FAIM3, which is expressed in tissues of the digestive and urinary tracts, bone marrow and testes, and restricted to the cerebellum in the nervous system. FAIM3 protects against DR-induced apoptosis by inducing the expression of other DR-antagonists such as CFLAR or through the interaction with the DR-adaptor protein Fas-associated via death domain. FAIM3 null mouse models reveal this protein as an important mediator of inflammatory autoimmune responses such as those triggered in autoimmune encephalomyelitis. Given the differences between FAIMs and the variety of processes in which they are involved, here we sought to provide a concise review about these molecules and their roles in the physiology and pathology of the nervous system. Even though they share name and inhibit Fas-induced cell death, Fas apoptotic inhibitory molecules (FAIMs) are not structurally related and inhibit apoptosis through completely different mechanisms. In this review, we describe FAIM1, FAIM2, and FAIM3 functions in the nervous system, and their implication in diverse pathologies such as neurodegenerative disease, cancer, or autoimmune diseases.


Subject(s)
Apoptosis Regulatory Proteins/genetics , Cell Death/genetics , Nervous System , fas Receptor/antagonists & inhibitors , fas Receptor/genetics , Animals , Apoptosis/drug effects , Cell Death/drug effects , Humans , Mice
9.
J Biol Chem ; 291(3): 1221-34, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26582200

ABSTRACT

Death receptors are members of the tumor necrosis factor receptor superfamily involved in the extrinsic apoptotic pathway. Lifeguard (LFG) is a death receptor antagonist mainly expressed in the nervous system that specifically blocks Fas ligand (FasL)-induced apoptosis. To investigate its mechanism of action, we studied its subcellular localization and its interaction with members of the Bcl-2 family proteins. We performed an analysis of LFG subcellular localization in murine cortical neurons and found that LFG localizes mainly to the ER and Golgi. We confirmed these results with subcellular fractionation experiments. Moreover, we show by co-immunoprecipitation experiments that LFG interacts with Bcl-XL and Bcl-2, but not with Bax or Bak, and this interaction likely occurs in the endoplasmic reticulum. We further investigated the relationship between LFG and Bcl-XL in the inhibition of apoptosis and found that LFG protects only type II apoptotic cells from FasL-induced death in a Bcl-XL dependent manner. The observation that LFG itself is not located in mitochondria raises the question as to whether LFG in the ER participates in FasL-induced death. Indeed, we investigated the degree of calcium mobilization after FasL stimulation and found that LFG inhibits calcium release from the ER, a process that correlates with LFG blockage of cytochrome c release to the cytosol and caspase activation. On the basis of our observations, we propose that there is a required step in the induction of type II apoptotic cell death that involves calcium mobilization from the ER and that this step is modulated by LFG.


Subject(s)
Apoptosis , Calcium Signaling , Endoplasmic Reticulum/metabolism , Fas Ligand Protein/antagonists & inhibitors , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Animals , Cell Line , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/metabolism , Fas Ligand Protein/genetics , Fas Ligand Protein/metabolism , Female , Golgi Apparatus/metabolism , HEK293 Cells , Humans , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/chemistry , Membrane Proteins/genetics , Mice, Inbred C57BL , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/genetics , Neurons/cytology , Peptide Fragments/antagonists & inhibitors , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Interaction Domains and Motifs , Protein Transport , RNA Interference , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism
10.
Mol Cancer ; 14: 62, 2015 Mar 19.
Article in English | MEDLINE | ID: mdl-25890358

ABSTRACT

BACKGROUND: Patients with high-risk neuroblastoma (NBL) tumors have a high mortality rate. Consequently, there is an urgent need for the development of new treatments for this condition. Targeting death receptor signaling has been proposed as an alternative to standard chemo- and radio-therapies in various tumors. In NBL, this therapeutic strategy has been largely disregarded, possibly because ~50-70% of all human NBLs are characterized by caspase-8 silencing. However, the expression of caspase-8 is detected in a significant group of NBL patients, and they could therefore benefit from treatments that induce cell death through death receptor activation. Given that cytokines, such as TNFα, are able to upregulate Fas expression, we sought to address the therapeutic relevance of co-treatment with TNFα and FasL in NBL. METHODS: For the purpose of the study we used a set of eight NBL cell lines. Here we explore the cell death induced by TNFα, FasL, cisplatin, and etoposide, or a combination thereof by Hoechst staining and calcein viability assay. Further assessment of the signaling pathways involved was performed by caspase activity assays and Western blot experiments. Characterization of Fas expression levels was achieved by qRT-PCR, cell surface biotinylation assays, and cytometry. RESULTS: We have found that TNFα is able to increase FasL-induced cell death by a mechanism that involves the NF-κB-mediated induction of the Fas receptor. Moreover, TNFα sensitized NBL cells to DNA-damaging agents (i.e. cisplatin and etoposide) that induce the expression of FasL. Priming to FasL-, cisplatin-, and etoposide-induced cell death could only be achieved in NBLs that display TNFα-induced upregulation of Fas. Further analysis denotes that the high degree of heterogeneity between NBLs is also manifested in Fas expression and modulation thereof by TNFα. CONCLUSIONS: In summary, our findings reveal that TNFα sensitizes NBL cells to FasL-induced cell death by NF-κB-mediated upregulation of Fas and unveil a new mechanism through which TNFα enhances the efficacy of currently used NBL treatments, cisplatin and etoposide.


Subject(s)
Drug Resistance, Neoplasm , Fas Ligand Protein/pharmacology , Gene Expression Regulation, Neoplastic , NF-kappa B/metabolism , Neuroblastoma/genetics , Neuroblastoma/metabolism , Tumor Necrosis Factor-alpha/pharmacology , fas Receptor/genetics , Caspase 8/metabolism , Cell Death/drug effects , Cell Line, Tumor , Cisplatin/pharmacology , Death Domain Receptor Signaling Adaptor Proteins/metabolism , Enzyme Activation/drug effects , Etoposide/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Humans , Interferon-gamma/pharmacology , Transcription, Genetic
11.
J Neurosci ; 33(49): 19262-75, 2013 Dec 04.
Article in English | MEDLINE | ID: mdl-24305822

ABSTRACT

The neuronal long isoform of Fas Apoptotic Inhibitory Molecule (FAIM-L) protects from death receptor (DR)-induced apoptosis, yet its mechanism of protection remains unknown. Here, we show that FAIM-L protects rat neuronal Type II cells from Fas-induced apoptosis. XIAP has previously emerged as a molecular discriminator that is upregulated in Type II and downregulated in Type I apoptotic signaling. We demonstrate that FAIM-L requires sustained endogenous levels of XIAP to protect Type II cells as well as murine cortical neurons from Fas-induced apoptosis. FAIM-L interacts with the BIR2 domain of XIAP through an IAP-binding motif, the mutation of which impairs the antiapoptotic function of FAIM-L. Finally, we report that FAIM-L inhibits XIAP auto-ubiquitinylation and maintains its stability, thus conferring protection from apoptosis. Our results bring new understanding of the regulation of endogenous XIAP by a DR antagonist, pointing out at FAIM-L as a promising therapeutic tool for protection from apoptosis in pathological situations where XIAP levels are decreased.


Subject(s)
Apoptosis Regulatory Proteins/pharmacology , Apoptosis/genetics , Inhibitor of Apoptosis Proteins/metabolism , Neuroprotective Agents , Ubiquitination/drug effects , X-Linked Inhibitor of Apoptosis Protein/physiology , fas Receptor/physiology , Animals , Apoptosis Regulatory Proteins/genetics , Carrier Proteins/metabolism , Caspases/metabolism , Cytochromes c/metabolism , Female , Immunoprecipitation , Inhibitor of Apoptosis Proteins/genetics , Lentivirus/genetics , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Mutagenesis, Site-Directed , PC12 Cells , Plasmids/genetics , Protein Binding , Protein Conformation , Rats , Real-Time Polymerase Chain Reaction , X-Linked Inhibitor of Apoptosis Protein/genetics , bcl-2-Associated X Protein/metabolism , fas Receptor/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...