Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters

Publication year range
1.
Am J Hum Genet ; 109(5): 909-927, 2022 05 05.
Article in English | MEDLINE | ID: mdl-35390279

ABSTRACT

Pontocerebellar hypoplasias (PCHs) are congenital disorders characterized by hypoplasia or early atrophy of the cerebellum and brainstem, leading to a very limited motor and cognitive development. Although over 20 genes have been shown to be mutated in PCHs, a large proportion of affected individuals remains undiagnosed. We describe four families with children presenting with severe neonatal brainstem dysfunction and pronounced deficits in cognitive and motor development associated with four different bi-allelic mutations in PRDM13, including homozygous truncating variants in the most severely affected individuals. Brain MRI and fetopathological examination revealed a PCH-like phenotype, associated with major hypoplasia of inferior olive nuclei and dysplasia of the dentate nucleus. Notably, histopathological examinations highlighted a sparse and disorganized Purkinje cell layer in the cerebellum. PRDM13 encodes a transcriptional repressor known to be critical for neuronal subtypes specification in the mouse retina and spinal cord but had not been implicated, so far, in hindbrain development. snRNA-seq data mining and in situ hybridization in humans show that PRDM13 is expressed at early stages in the progenitors of the cerebellar ventricular zone, which gives rise to cerebellar GABAergic neurons, including Purkinje cells. We also show that loss of function of prdm13 in zebrafish leads to a reduction in Purkinje cells numbers and a complete absence of the inferior olive nuclei. Altogether our data identified bi-allelic mutations in PRDM13 as causing a olivopontocerebellar hypoplasia syndrome and suggest that early deregulations of the transcriptional control of neuronal fate specification could contribute to a significant number of cases.


Subject(s)
Brain Diseases , Zebrafish , Animals , Brain Diseases/pathology , Brain Stem , Cerebellum/abnormalities , Cerebellum/pathology , Developmental Disabilities , Histone-Lysine N-Methyltransferase/genetics , Humans , Mice , Mutation/genetics , Nervous System Malformations , Neurogenesis/genetics , Purkinje Cells/metabolism , Transcription Factors/genetics , Zebrafish/metabolism
2.
Clin Genet ; 104(4): 466-471, 2023 10.
Article in English | MEDLINE | ID: mdl-37243350

ABSTRACT

CHARGE syndrome, due to CHD7 pathogenic variations, is an autosomal dominant disorder characterized by a large spectrum of severity. Despite the great number of variations reported, no clear genotype-to-phenotype correlation has been reported. Unsupervised machine learning and clustering was undertaken using a retrospective cohort of 42 patients, after deep radiologic and clinical phenotyping, to establish genotype-phenotype correlation for CHD7-related CHARGE syndrome. It resulted in three clusters showing phenotypes of different severities. While no clear genotype-phenotype correlation appeared within the first two clusters, a single patient was outlying the cohort data (cluster 3) with the most atypical phenotype and the most distal frameshift variant in the gene. We added two other patients with similar distal pathogenic variants and observed a tendency toward mild and/or atypical phenotypes. We hypothesized that this finding could potentially be related to escaping nonsense mediated RNA decay, but found no evidence of such decay in vivo for any of the CHD7 pathogenic variation tested. This indicates that this milder phenotype may rather result from the production of a protein retaining all functional domains.


Subject(s)
CHARGE Syndrome , Humans , CHARGE Syndrome/genetics , Retrospective Studies , Phenotype , Genetic Association Studies , Genotype , Mutation/genetics
3.
Nucleic Acids Res ; 49(W1): W93-W103, 2021 07 02.
Article in English | MEDLINE | ID: mdl-34019647

ABSTRACT

Copy Number Variants (CNVs) are an important cause of rare diseases. Array-based Comparative Genomic Hybridization tests yield a ∼12% diagnostic rate, with ∼8% of patients presenting CNVs of unknown significance. CNVs interpretation is particularly challenging on genomic regions outside of those overlapping with previously reported structural variants or disease-associated genes. Recent studies showed that a more comprehensive evaluation of CNV features, leveraging both coding and non-coding impacts, can significantly improve diagnostic rates. However, currently available CNV interpretation tools are mostly gene-centric or provide only non-interactive annotations difficult to assess in the clinical practice. Here, we present CNVxplorer, a web server suited for the functional assessment of CNVs in a clinical diagnostic setting. CNVxplorer mines a comprehensive set of clinical, genomic, and epigenomic features associated with CNVs. It provides sequence constraint metrics, impact on regulatory elements and topologically associating domains, as well as expression patterns. Analyses offered cover (a) agreement with patient phenotypes; (b) visualizations of associations among genes, regulatory elements and transcription factors; (c) enrichment on functional and pathway annotations and (d) co-occurrence of terms across PubMed publications related to the query CNVs. A flexible evaluation workflow allows dynamic re-interrogation in clinical sessions. CNVxplorer is publicly available at http://cnvxplorer.com.


Subject(s)
DNA Copy Number Variations , Rare Diseases/genetics , Software , Animals , Gene Expression , Genome, Human , Humans , Internet , Mice, Knockout , Phenotype , Protein Interaction Mapping , Rare Diseases/diagnosis , Regulatory Sequences, Nucleic Acid
4.
Proc Natl Acad Sci U S A ; 117(24): 13626-13636, 2020 06 16.
Article in English | MEDLINE | ID: mdl-32487729

ABSTRACT

Humans homozygous or hemizygous for variants predicted to cause a loss of function (LoF) of the corresponding protein do not necessarily present with overt clinical phenotypes. We report here 190 autosomal genes with 207 predicted LoF variants, for which the frequency of homozygous individuals exceeds 1% in at least one human population from five major ancestry groups. No such genes were identified on the X and Y chromosomes. Manual curation revealed that 28 variants (15%) had been misannotated as LoF. Of the 179 remaining variants in 166 genes, only 11 alleles in 11 genes had previously been confirmed experimentally to be LoF. The set of 166 dispensable genes was enriched in olfactory receptor genes (41 genes). The 41 dispensable olfactory receptor genes displayed a relaxation of selective constraints similar to that observed for other olfactory receptor genes. The 125 dispensable nonolfactory receptor genes also displayed a relaxation of selective constraints consistent with greater redundancy. Sixty-two of these 125 genes were found to be dispensable in at least three human populations, suggesting possible evolution toward pseudogenes. Of the 179 LoF variants, 68 could be tested for two neutrality statistics, and 8 displayed robust signals of positive selection. These latter variants included a known FUT2 variant that confers resistance to intestinal viruses, and an APOL3 variant involved in resistance to parasitic infections. Overall, the identification of 166 genes for which a sizeable proportion of humans are homozygous for predicted LoF alleles reveals both redundancies and advantages of such deficiencies for human survival.


Subject(s)
Human Genetics , Loss of Function Mutation , Alleles , Apolipoproteins L/genetics , Fucosyltransferases/genetics , Genetic Variation , Homozygote , Humans , Proteins/genetics , Sex Chromosomes/genetics , Galactoside 2-alpha-L-fucosyltransferase
5.
Semin Immunol ; 36: 13-16, 2018 04.
Article in English | MEDLINE | ID: mdl-29229549

ABSTRACT

Advances in genomics and medicine have enabled the identification of (currently) 346 primary immunodeficiencies (PIDs) caused by mutations in 336 different genes. Most of these PIDs are monogenic conditions with Mendelian inheritance. Given this large number, it is possible to analyze the distribution of PIDs associated with infections and/or immunopathology according to the nature of the defect - even though this exercise can be challenging and arguable because of the pleiotropic nature of some gene products. The results of this analysis nevertheless strongly suggests that innate immune responses (mediated by pattern recognition receptor (PRR) engagement) are largely redundant, whereas adaptive immune responses are essential. Conversely, gain of function is more frequent in PRR-mediated immune responses than in adaptive immune responses - suggesting that robust innate immune pathways are less stringently regulated than energetically costly and potentially harmful adaptive immune responses.


Subject(s)
Adaptive Immunity/genetics , Immunity, Innate/genetics , Immunologic Deficiency Syndromes/immunology , Infections/genetics , Mutation/genetics , Receptors, Pattern Recognition/genetics , Animals , Energy Metabolism , Humans
6.
PLoS Pathog ; 13(10): e1006678, 2017 Oct.
Article in English | MEDLINE | ID: mdl-29073251

ABSTRACT

Cellular permissiveness to HIV infection is highly heterogeneous across individuals. Heterogeneity is also found across CD4+ T cells from the same individual, where only a fraction of cells gets infected. To explore the basis of permissiveness, we performed single-cell RNA-seq analysis of non-infected CD4+ T cells from high and low permissive individuals. Transcriptional heterogeneity translated in a continuum of cell states, driven by T-cell receptor-mediated cell activation and was strongly linked to permissiveness. Proteins expressed at the cell surface and displaying the highest correlation with T cell activation were tested as biomarkers of cellular permissiveness to HIV. FACS sorting using antibodies against several biomarkers of permissiveness led to an increase of HIV cellular infection rates. Top candidate biomarkers included CD25, a canonical activation marker. The combination of CD25 high expression with other candidate biomarkers led to the identification of CD298, CD63 and CD317 as the best biomarkers for permissiveness. CD25highCD298highCD63highCD317high cell population showed an enrichment of HIV-infection of up to 28 fold as compared to the unsorted cell population. The purified hyper-permissive cell subpopulation was characterized by a downregulation of interferon-induced genes and several known restriction factors. Single-cell RNA-seq analysis coupled with functional characterization of cell biomarkers provides signatures of the "HIV-permissive cell".


Subject(s)
CD4-Positive T-Lymphocytes/immunology , HIV Infections/immunology , HIV-1/immunology , Lymphocyte Activation/immunology , Cells, Cultured , Humans , Single-Cell Analysis/methods , Virus Replication/physiology
7.
Retrovirology ; 13(1): 43, 2016 06 27.
Article in English | MEDLINE | ID: mdl-27350062

ABSTRACT

BACKGROUND: Primary CD4+ T cells and cell lines differ in their permissiveness to HIV infection. Impaired innate immunity may contribute to this different phenotype. FINDINGS: We used transcriptome profiling of 1503 innate immunity genes in primary CD4+ T cells and permissive cell lines. Two clusters of differentially expressed genes were identified: a set of 249 genes that were highly expressed in primary cells and minimally expressed in cell lines and a set of 110 genes with the opposite pattern. Specific to HIV, HEK293T, Jurkat, SupT1 and CEM cell lines displayed unique patterns of downregulation of genes involved in viral sensing and restriction. Activation of primary CD4+ T cells resulted in reversal of the pattern of expression of those sets of innate immunity genes. Functional analysis of prototypical innate immunity pathways of permissive cell lines confirmed impaired responses identified in transcriptome analyses. CONCLUSION: Integrity of innate immunity genes and pathways needs to be considered in designing gain/loss functional genomic screens of viral infection.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/virology , HIV-1/physiology , Immunity, Innate , Virus Replication , Cell Line , Cells, Cultured , Gene Expression Profiling , HEK293 Cells , HIV-1/immunology , Humans , Immunity, Innate/genetics , Lymphocyte Activation , Phenotype , Receptors, Antigen, T-Cell/immunology , Virus Cultivation
8.
Bioinformatics ; 31(20): 3380-2, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-26099264

ABSTRACT

UNLABELLED: Cell differentiation processes are achieved through a continuum of hierarchical intermediate cell states that might be captured by single-cell RNA seq. Existing computational approaches for the assessment of cell-state hierarchies from single-cell data can be formalized under a general framework composed of (i) a metric to assess cell-to-cell similarities (with or without a dimensionality reduction step) and (ii) a graph-building algorithm (optionally making use of a cell clustering step). The Sincell R package implements a methodological toolbox allowing flexible workflows under such a framework. Furthermore, Sincell contributes new algorithms to provide cell-state hierarchies with statistical support while accounting for stochastic factors in single-cell RNA seq. Graphical representations and functional association tests are provided to interpret hierarchies. The functionalities of Sincell are illustrated in a real case study, which demonstrates its ability to discriminate noisy from stable cell-state hierarchies. AVAILABILITY AND IMPLEMENTATION: Sincell is an open-source R/Bioconductor package available at http://bioconductor.org/packages/sincell. A detailed manual and a vignette are provided with the package. CONTACT: antonio.rausell@isb-sib.ch SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Subject(s)
Cell Differentiation/genetics , Gene Expression Profiling , Sequence Analysis, RNA , Software , Algorithms , Data Interpretation, Statistical , Single-Cell Analysis
9.
PLoS Comput Biol ; 11(12): e1004647, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26642228

ABSTRACT

Sequencing projects have identified large numbers of rare stop-gain and frameshift variants in the human genome. As most of these are observed in the heterozygous state, they test a gene's tolerance to haploinsufficiency and dominant loss of function. We analyzed the distribution of truncating variants across 16,260 autosomal protein coding genes in 11,546 individuals. We observed 39,893 truncating variants affecting 12,062 genes, which significantly differed from an expectation of 12,916 genes under a model of neutral de novo mutation (p<10-4). Extrapolating this to increasing numbers of sequenced individuals, we estimate that 10.8% of human genes do not tolerate heterozygous truncating variants. An additional 10 to 15% of truncated genes may be rescued by incomplete penetrance or compensatory mutations, or because the truncating variants are of limited functional impact. The study of protein truncating variants delineates the essential genome and, more generally, identifies rare heterozygous variants as an unexplored source of diversity of phenotypic traits and diseases.


Subject(s)
Chromosome Mapping/methods , Codon, Nonsense/genetics , Genetic Variation/genetics , Genome, Human/genetics , Proteins/genetics , Base Sequence , Humans , Molecular Sequence Data
10.
Mol Cell Proteomics ; 13(11): 2975-85, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25044018

ABSTRACT

Age-related cognitive decline is a serious health concern in our aging society. Decreased cognitive function observed during healthy brain aging is most likely caused by changes in brain connectivity and synaptic dysfunction in particular brain regions. Here we show that aged C57BL/6J wild-type mice have hippocampus-dependent spatial memory impairments. To identify the molecular mechanisms that are relevant to these memory deficits, we investigated the temporal profile of mouse hippocampal synaptic proteome changes at 20, 40, 50, 60, 70, 80, 90, and 100 weeks of age. Extracellular matrix proteins were the only group of proteins that showed robust and progressive up-regulation over time. This was confirmed by immunoblotting and histochemical analysis, which indicated that the increased levels of hippocampal extracellular matrix might limit synaptic plasticity as a potential cause of age-related cognitive decline. In addition, we observed that stochasticity in synaptic protein expression increased with age, in particular for proteins that were previously linked with various neurodegenerative diseases, whereas low variance in expression was observed for proteins that play a basal role in neuronal function and synaptic neurotransmission. Together, our findings show that both specific changes and increased variance in synaptic protein expression are associated with aging and may underlie reduced synaptic plasticity and impaired cognitive performance in old age.


Subject(s)
Cognitive Dysfunction/physiopathology , Extracellular Matrix Proteins/metabolism , Hippocampus/physiology , Maze Learning/physiology , Memory Disorders/physiopathology , Aging/physiology , Animals , Cognition/physiology , Extracellular Matrix/metabolism , Extracellular Matrix Proteins/biosynthesis , Hippocampus/cytology , Male , Mice , Mice, Inbred C57BL , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/physiopathology , Neuronal Plasticity/physiology , Proteome/analysis , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Stochastic Processes , Tandem Mass Spectrometry
11.
Proc Natl Acad Sci U S A ; 110(19): 7820-5, 2013 May 07.
Article in English | MEDLINE | ID: mdl-23610393

ABSTRACT

Immunoresponsive gene 1 (Irg1) is highly expressed in mammalian macrophages during inflammation, but its biological function has not yet been elucidated. Here, we identify Irg1 as the gene coding for an enzyme producing itaconic acid (also known as methylenesuccinic acid) through the decarboxylation of cis-aconitate, a tricarboxylic acid cycle intermediate. Using a gain-and-loss-of-function approach in both mouse and human immune cells, we found Irg1 expression levels correlating with the amounts of itaconic acid, a metabolite previously proposed to have an antimicrobial effect. We purified IRG1 protein and identified its cis-aconitate decarboxylating activity in an enzymatic assay. Itaconic acid is an organic compound that inhibits isocitrate lyase, the key enzyme of the glyoxylate shunt, a pathway essential for bacterial growth under specific conditions. Here we show that itaconic acid inhibits the growth of bacteria expressing isocitrate lyase, such as Salmonella enterica and Mycobacterium tuberculosis. Furthermore, Irg1 gene silencing in macrophages resulted in significantly decreased intracellular itaconic acid levels as well as significantly reduced antimicrobial activity during bacterial infections. Taken together, our results demonstrate that IRG1 links cellular metabolism with immune defense by catalyzing itaconic acid production.


Subject(s)
Gene Expression Regulation , Hydro-Lyases/metabolism , Macrophages/metabolism , Proteins/metabolism , Succinates/metabolism , Animals , Carboxy-Lyases , Catalysis , Cell Line , Green Fluorescent Proteins/metabolism , HEK293 Cells , Humans , Inflammation , Lipopolysaccharide Receptors/metabolism , Macrophages/immunology , Mice , Mice, Inbred C57BL , Monocytes/cytology , Mycobacterium tuberculosis/metabolism , RNA, Small Interfering/metabolism
12.
Retrovirology ; 12: 37, 2015 May 07.
Article in English | MEDLINE | ID: mdl-25947229

ABSTRACT

BACKGROUND: Myeloid cells are key players in the recognition and response of the host against invading viruses. Paradoxically, upon HIV-1 infection, myeloid cells might also promote viral pathogenesis through trans-infection, a mechanism that promotes HIV-1 transmission to target cells via viral capture and storage. The receptor Siglec-1 (CD169) potently enhances HIV-1 trans-infection and is regulated by immune activating signals present throughout the course of HIV-1 infection, such as interferon α (IFNα). RESULTS: Here we show that IFNα-activated dendritic cells, monocytes and macrophages have an enhanced ability to capture and trans-infect HIV-1 via Siglec-1 recognition of viral membrane gangliosides. Monocytes from untreated HIV-1-infected individuals trans-infect HIV-1 via Siglec-1, but this capacity diminishes after effective antiretroviral treatment. Furthermore, Siglec-1 is expressed on myeloid cells residing in lymphoid tissues, where it can mediate viral trans-infection. CONCLUSIONS: Siglec-1 on myeloid cells could fuel novel CD4(+) T-cell infections and contribute to HIV-1 dissemination in vivo.


Subject(s)
HIV-1/immunology , HIV-1/physiology , Interferon-alpha/metabolism , Myeloid Cells/virology , Sialic Acid Binding Ig-like Lectin 1/biosynthesis , Up-Regulation , Adult , CD4-Positive T-Lymphocytes/virology , Cells, Cultured , Humans , Male
13.
Retrovirology ; 12: 41, 2015 May 16.
Article in English | MEDLINE | ID: mdl-25980612

ABSTRACT

BACKGROUND: Known antiretroviral restriction factors are encoded by genes that are under positive selection pressure, induced during HIV-1 infection, up-regulated by interferons, and/or interact with viral proteins. To identify potential novel restriction factors, we performed genome-wide scans for human genes sharing molecular and evolutionary signatures of known restriction factors and tested the anti-HIV-1 activity of the most promising candidates. RESULTS: Our analyses identified 30 human genes that share characteristics of known restriction factors. Functional analyses of 27 of these candidates showed that over-expression of a strikingly high proportion of them significantly inhibited HIV-1 without causing cytotoxic effects. Five factors (APOL1, APOL6, CD164, TNFRSF10A, TNFRSF10D) suppressed infectious HIV-1 production in transfected 293T cells by >90% and six additional candidates (FCGR3A, CD3E, OAS1, GBP5, SPN, IFI16) achieved this when the virus was lacking intact accessory vpr, vpu and nef genes. Unexpectedly, over-expression of two factors (IL1A, SP110) significantly increased infectious HIV-1 production. Mechanistic studies suggest that the newly identified potential restriction factors act at different steps of the viral replication cycle, including proviral transcription and production of viral proteins. Finally, we confirmed that mRNA expression of most of these candidate restriction factors in primary CD4+ T cells is significantly increased by type I interferons. CONCLUSIONS: A limited number of human genes share multiple characteristics of genes encoding for known restriction factors. Most of them display anti-retroviral activity in transient transfection assays and are expressed in primary CD4+ T cells.


Subject(s)
HIV-1/immunology , HIV-1/physiology , Host-Pathogen Interactions , Immunity, Innate , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/virology , Cell Line , Gene Expression Profiling , Genetic Testing , Humans
14.
PLoS Comput Biol ; 10(7): e1003757, 2014 Jul.
Article in English | MEDLINE | ID: mdl-25058640

ABSTRACT

Loss-of-function variants in innate immunity genes are associated with Mendelian disorders in the form of primary immunodeficiencies. Recent resequencing projects report that stop-gains and frameshifts are collectively prevalent in humans and could be responsible for some of the inter-individual variability in innate immune response. Current computational approaches evaluating loss-of-function in genes carrying these variants rely on gene-level characteristics such as evolutionary conservation and functional redundancy across the genome. However, innate immunity genes represent a particular case because they are more likely to be under positive selection and duplicated. To create a ranking of severity that would be applicable to innate immunity genes we evaluated 17,764 stop-gain and 13,915 frameshift variants from the NHLBI Exome Sequencing Project and 1,000 Genomes Project. Sequence-based features such as loss of functional domains, isoform-specific truncation and nonsense-mediated decay were found to correlate with variant allele frequency and validated with gene expression data. We integrated these features in a Bayesian classification scheme and benchmarked its use in predicting pathogenic variants against Online Mendelian Inheritance in Man (OMIM) disease stop-gains and frameshifts. The classification scheme was applied in the assessment of 335 stop-gains and 236 frameshifts affecting 227 interferon-stimulated genes. The sequence-based score ranks variants in innate immunity genes according to their potential to cause disease, and complements existing gene-based pathogenicity scores. Specifically, the sequence-based score improves measurement of functional gene impairment, discriminates across different variants in a given gene and appears particularly useful for analysis of less conserved genes.


Subject(s)
Computational Biology/methods , Databases, Genetic , Immunity, Innate/genetics , Mutation/genetics , Bayes Theorem , Humans , Interferons/genetics , Interferons/metabolism , Virus Diseases/immunology
16.
Eur J Hum Genet ; 32(7): 864-870, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38760421

ABSTRACT

Carpenter syndrome (CRPTS) is a rare autosomal recessive condition caused by biallelic variants in genes that encode negative regulators of hedgehog signalling (RAB23 [CRPT1] or, more rarely, MEGF8 [CRPT2]), and is characterised by craniosynostosis, polysyndactyly, and other congenital abnormalities. We describe a further six families comprising eight individuals with MEGF8-associated CRPT2, increasing the total number of reported cases to fifteen, and refine the phenotype of CRPT2 compared to CRPT1. The core features of craniosynostosis, polysyndactyly and (in males) cryptorchidism are almost universal in both CRPT1 and CRPT2. However, laterality defects are present in nearly half of those with MEGF8-associated CRPT2, but are rare in RAB23-associated CRPT1. Craniosynostosis in CRPT2 commonly involves a single midline suture in comparison to the multi-suture craniosynostosis characteristic of CRPT1. No patient to date has carried two MEGF8 gene alterations that are both predicted to lead to complete loss-of-function, suggesting that a variable degree of residual MEGF8 activity may be essential for viability and potentially contributing to variable phenotypic severity. These data refine the phenotypic spectrum of CRPT2 in comparison to CRPT1 and more than double the number of likely pathogenic MEGF8 variants in this rare disorder.


Subject(s)
Acrocephalosyndactylia , Phenotype , Humans , Male , Female , Acrocephalosyndactylia/genetics , Acrocephalosyndactylia/pathology , Child , Child, Preschool , Infant , rab GTP-Binding Proteins/genetics , Pedigree , Membrane Proteins/genetics , Adolescent , Mutation
17.
Bioinformatics ; 28(4): 584-6, 2012 Feb 15.
Article in English | MEDLINE | ID: mdl-22171333

ABSTRACT

UNLABELLED: We have implemented in a single package all the features required for extracting, visualizing and manipulating fully conserved positions as well as those with a family-dependent conservation pattern in multiple sequence alignments. The program allows, among other things, to run different methods for extracting these positions, combine the results and visualize them in protein 3D structures and sequence spaces. AVAILABILITY AND IMPLEMENTATION: JDet is a multiplatform application written in Java. It is freely available, including the source code, at http://csbg.cnb.csic.es/JDet. The package includes two of our recently developed programs for detecting functional positions in protein alignments (Xdet and S3Det), and support for other methods can be added as plug-ins. A help file and a guided tutorial for JDet are also available.


Subject(s)
Proteins/chemistry , Sequence Alignment , Software , Amino Acid Sequence , Models, Molecular , Molecular Sequence Data , Proteins/genetics
18.
Proc Natl Acad Sci U S A ; 107(5): 1995-2000, 2010 Feb 02.
Article in English | MEDLINE | ID: mdl-20133844

ABSTRACT

The divergence accumulated during the evolution of protein families translates into their internal organization as subfamilies, and it is directly reflected in the characteristic patterns of differentially conserved residues. These specifically conserved positions in protein subfamilies are known as "specificity determining positions" (SDPs). Previous studies have limited their analysis to the study of the relationship between these positions and ligand-binding specificity, demonstrating significant yet limited predictive capacity. We have systematically extended this observation to include the role of differential protein interactions in the segregation of protein subfamilies and explored in detail the structural distribution of SDPs at protein interfaces. Our results show the extensive influence of protein interactions in the evolution of protein families and the widespread association of SDPs with protein interfaces. The combined analysis of SDPs in interfaces and ligand-binding sites provides a more complete picture of the organization of protein families, constituting the necessary framework for a large scale analysis of the evolution of protein function.


Subject(s)
Proteins/chemistry , Proteins/metabolism , Amino Acid Sequence , Animals , Binding Sites , Biophysical Phenomena , Conserved Sequence , Databases, Protein , Evolution, Molecular , Humans , Ligands , Models, Molecular , Multiprotein Complexes , Protein Conformation , Protein Interaction Domains and Motifs , Proteins/classification , Proteins/genetics , Sequence Homology, Amino Acid
19.
Sci Data ; 10(1): 361, 2023 06 06.
Article in English | MEDLINE | ID: mdl-37280226

ABSTRACT

While the amount of studies involving single-cell or single-nucleus RNA-sequencing technologies grows exponentially within the biomedical research area, the kidney field requires reference transcriptomic signatures to allocate each cluster its matching cell type. The present meta-analysis of 39 previously published datasets, from 7 independent studies, involving healthy human adult kidney samples, offers a set of 24 distinct consensus kidney cell type signatures. The use of these signatures may help to assure the reliability of cell type identification in future studies involving single-cell and single-nucleus transcriptomics while improving the reproducibility in cell type allocation.


Subject(s)
Kidney , Transcriptome , Adult , Humans , Gene Expression Profiling , Reproducibility of Results , Single-Cell Gene Expression Analysis , Datasets as Topic
20.
BMC Med Genomics ; 16(1): 143, 2023 06 21.
Article in English | MEDLINE | ID: mdl-37344844

ABSTRACT

Bi-allelic variants in the mitochondrial arginyl-transfer RNA synthetase (RARS2) gene have been involved in early-onset encephalopathies classified as pontocerebellar hypoplasia (PCH) type 6 and in epileptic encephalopathy. A variant (NM_020320.3:c.-2A > G) in the promoter and 5'UTR of the RARS2 gene has been previously identified in a family with PCH. Only a mild impact of this variant on the mRNA level has been detected. As RARS2 is non-dosage-sensitive, this observation is not conclusive in regard of the pathogenicity of the variant.We report and describe here a new patient with the same variant in the RARS2 gene, at the homozygous state. This patient presents with a clinical phenotype consistent with PCH6 although in the absence of lactic acidosis. In agreement with the previous study, we measured RARS2 mRNA levels in patient's fibroblasts and detected a partially preserved gene expression compared to control. Importantly, this variant is located in the Kozak sequence that controls translation initiation. Therefore, we investigated the impact on protein translation using a bioinformatic approach and western blotting. We show here that this variant, additionally to its effect on the transcription, also disrupts the consensus Kozak sequence, and has a major impact on RARS2 protein translation. Through the identification of this additional case and the characterization of the molecular consequences, we clarified the involvement of this Kozak variant in PCH and on protein synthesis. This work also points to the current limitation in the pathogenicity prediction of variants located in the translation initiation region.


Subject(s)
Arginine-tRNA Ligase , Cerebellar Diseases , Olivopontocerebellar Atrophies , Humans , Olivopontocerebellar Atrophies/genetics , RNA, Messenger/genetics
SELECTION OF CITATIONS
SEARCH DETAIL