Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Brain ; 144(9): 2625-2634, 2021 10 22.
Article in English | MEDLINE | ID: mdl-33848319

ABSTRACT

Although CSF analysis routinely enables the diagnosis of neurological diseases, it is mainly used for the gross distinction between infectious, autoimmune inflammatory, and degenerative disorders of the CNS. To investigate, whether a multi-dimensional cellular blood and CSF characterization can support the diagnosis of clinically similar neurological diseases, we analysed 546 patients with autoimmune neuroinflammatory, degenerative, or vascular conditions in a cross-sectional retrospective study. By combining feature selection with dimensionality reduction and machine learning approaches we identified pan-disease parameters that were altered across all autoimmune neuroinflammatory CNS diseases and differentiated them from other neurological conditions and inter-autoimmunity classifiers that subdifferentiate variants of CNS-directed autoimmunity. Pan-disease as well as diseases-specific changes formed a continuum, reflecting clinical disease evolution. A validation cohort of 231 independent patients confirmed that combining multiple parameters into composite scores can assist the classification of neurological patients. Overall, we showed that the integrated analysis of blood and CSF parameters improves the differential diagnosis of neurological diseases, thereby facilitating early treatment decisions.


Subject(s)
Inflammation Mediators/cerebrospinal fluid , Nervous System Diseases/cerebrospinal fluid , Nervous System Diseases/classification , Biomarkers/cerebrospinal fluid , Cohort Studies , Diagnosis, Differential , Female , Humans , Male , Nervous System Diseases/diagnosis , Retrospective Studies
2.
Pharmacol Res Perspect ; 9(3): e00770, 2021 05.
Article in English | MEDLINE | ID: mdl-33929099

ABSTRACT

We conducted a systematic evaluation of lung inflammation indued by repeated intranasal exposure (for 10 consecutive days) to a human aeroallergen, house dust mite (HDM) in BALB/c mice. Peak influx of neutrophils, monocytes/lymphocytes, and eosinophils was observed in bronchoalveolar lavage (BAL) on days 1, 7 and 11, respectively, and normalized to baseline by day 21. Peak elevations of Th2, myeloid-derived cytokines/chemokines and serum IgE were seen both in BAL and lung tissue homogenates between days 7 and 11, and declined thereafter; however, IL-33 levels remained elevated from day 7 to day 21. Airway hyperreactivity to inhaled methacholine was significantly increased by day 11 and decreased to baseline by day 21. The lung tissue showed perivascular and peribronchial cuffing, epithelial hypertrophy and hyperplasia and goblet cell formation in airways by day 11, and resolution by day 21. Levels of soluble collagen and tissue inhibitors of metalloproteinases (TIMP) also increased reflecting tissue remodeling in the lung. Microarray analysis demonstrated a significant time-dependent up-regulation of several genes including IL-33, CLCA3, CCL17, CD4, CD10, CD27, IL-13, Foxa3, IL-4, IL-10, and CD19, in BAL cells as well as the lung. Pre-treatment of HDM challenged mice with CCL17 and IL-13 antibodies reduced BAL cellularity, airway hyper-responsiveness (AHR), and histopathological changes. Notably, anti-IL-13, but not anti-CCL17 monoclonal antibodies (mAbs) reduced BAL neutrophilia while both mAbs attenuated eosinophilia. These results suggest that CCL17 has an overlapping, yet distinct profile versus IL-13 in the HDM model of pulmonary inflammation and potential for CCL17-based therapeutics in treating Th2 inflammation.


Subject(s)
Allergens/immunology , Asthma/immunology , Cytokines/immunology , Pyroglyphidae/immunology , Animals , Antibodies, Monoclonal/therapeutic use , Antigens, CD/genetics , Antigens, CD/immunology , Asthma/blood , Asthma/drug therapy , Asthma/genetics , Bronchoalveolar Lavage Fluid/cytology , Cytokines/antagonists & inhibitors , Cytokines/genetics , Eosinophilia/blood , Eosinophilia/drug therapy , Eosinophilia/genetics , Eosinophilia/immunology , Female , Gene Expression , Immunoglobulin E/blood , Inflammation/blood , Inflammation/drug therapy , Inflammation/genetics , Inflammation/immunology , Lung/immunology , Lung/pathology , Mice, Inbred BALB C , Neutrophils/drug effects
3.
Brain ; 144(2): 450-461, 2021 03 03.
Article in English | MEDLINE | ID: mdl-33374005

ABSTRACT

Multiple sclerosis is an autoimmune disease of the CNS in which both genetic and environmental factors are involved. Genome-wide association studies revealed more than 200 risk loci, most of which harbour genes primarily expressed in immune cells. However, whether genetic differences are translated into cell-specific gene expression profiles and to what extent these are altered in patients with multiple sclerosis are still open questions in the field. To assess cell type-specific gene expression in a large cohort of patients with multiple sclerosis, we sequenced the whole transcriptome of fluorescence-activated cell sorted T cells (CD4+ and CD8+) and CD14+ monocytes from treatment-naive patients with multiple sclerosis (n = 106) and healthy subjects (n = 22). We identified 479 differentially expressed genes in CD4+ T cells, 435 in monocytes, and 54 in CD8+ T cells. Importantly, in CD4+ T cells, we discovered upregulated transcripts from the NAE1 gene, a critical subunit of the NEDD8 activating enzyme, which activates the neddylation pathway, a post-translational modification analogous to ubiquitination. Finally, we demonstrated that inhibition of NEDD8 activating enzyme using the specific inhibitor pevonedistat (MLN4924) significantly ameliorated disease severity in murine experimental autoimmune encephalomyelitis. Our findings provide novel insights into multiple sclerosis-associated gene regulation unravelling neddylation as a crucial pathway in multiple sclerosis pathogenesis with implications for the development of tailored disease-modifying agents.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Monocytes/metabolism , Multiple Sclerosis/metabolism , Protein Processing, Post-Translational , Adult , Female , Gene Expression Profiling , Humans , Lipopolysaccharide Receptors , Male , Middle Aged , Monocytes/immunology , Multiple Sclerosis/immunology , Young Adult
4.
F1000Res ; 9: 992, 2020.
Article in English | MEDLINE | ID: mdl-33456761

ABSTRACT

Background: Severe coronavirus disease 2019 (COVID-19) is associated with multiple comorbidities and is characterized by an auto-aggressive inflammatory state leading to massive collateral damage. To identify preventive and therapeutic strategies against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), it is important to ascertain the molecular interactions between virus and host, and how they translate into disease pathophysiology. Methods: We matched virus-human protein interactions of human coronaviruses and other respiratory viruses with lists of genes associated with autoimmune diseases and comorbidities associated to worse COVID-19 course. We then selected the genes included in the statistically significant intersection between SARS-CoV-2 network and disease associated gene sets, identifying a meta-interactome. We analyzed the meta-interactome genes expression in samples derived from lungs of infected humans, and their regulation by IFN-ß. Finally, we performed a drug repurposing screening to target the network's most critical nodes. Results: We found a significant enrichment of SARS-CoV-2 interactors in immunological pathways and a strong association with autoimmunity and three prognostically relevant conditions (type 2 diabetes, coronary artery diseases, asthma), that present more independent physiopathological subnetworks. We observed a reduced expression of meta-interactome genes in human lungs after SARS-CoV-2 infection, and a regulatory potential of type I interferons. We also underscored multiple repurposable drugs to tailor the therapeutic strategies. Conclusions: Our data underscored a plausible genetic background that may contribute to the distinct observed pathophysiologies of severe COVID-19. Also, these results may help identify the most promising therapeutic targets and treatments for this condition.


Subject(s)
Autoimmunity , COVID-19/genetics , COVID-19/immunology , Asthma , Comorbidity , Coronary Artery Disease , Diabetes Mellitus, Type 2 , Drug Repositioning , Host-Pathogen Interactions , Humans , SARS-CoV-2
5.
Ann Neurol ; 84(1): 51-63, 2018 07.
Article in English | MEDLINE | ID: mdl-29908077

ABSTRACT

OBJECTIVE: Primary progressive multiple sclerosis (PPMS) causes accumulation of neurological disability from disease onset without clinical attacks typical of relapsing multiple sclerosis (RMS). However, whether genetic variation influences the disease course remains unclear. We aimed to determine whether mutations causative of neurological disorders that share features with multiple sclerosis (MS) contribute to risk for developing PPMS. METHODS: We examined whole-genome sequencing (WGS) data from 38 PPMS and 81 healthy subjects of European ancestry. We selected pathogenic variants exclusively found in PPMS patients that cause monogenic neurological disorders and performed two rounds of replication genotyping in 746 PPMS, 3,049 RMS, and 1,000 healthy subjects. To refine our findings, we examined the burden of rare, potentially pathogenic mutations in 41 genes that cause hereditary spastic paraplegias (HSPs) in PPMS (n = 314), secondary progressive multiple sclerosis (SPMS; n = 587), RMS (n = 2,248), and healthy subjects (n = 987) genotyped using the MS replication chip. RESULTS: WGS and replication studies identified three pathogenic variants in PPMS patients that cause neurological disorders sharing features with MS: KIF5A p.Ala361Val in spastic paraplegia 10; MLC1 p.Pro92Ser in megalencephalic leukodystrophy with subcortical cysts, and REEP1 c.606 + 43G>T in Spastic Paraplegia 31. Moreover, we detected a significant enrichment of HSP-related mutations in PPMS patients compared to controls (risk ratio [RR] = 1.95; 95% confidence interval [CI], 1.27-2.98; p = 0.002), as well as in SPMS patients compared to controls (RR = 1.57; 95% CI, 1.18-2.10; p = 0.002). Importantly, this enrichment was not detected in RMS. INTERPRETATION: This study provides evidence to support the hypothesis that rare Mendelian genetic variants contribute to the risk for developing progressive forms of MS. Ann Neurol 2018;83:51-63.


Subject(s)
Genome-Wide Association Study , Multiple Sclerosis, Chronic Progressive/genetics , Multiple Sclerosis, Chronic Progressive/physiopathology , Mutation/genetics , Adolescent , Adult , Aged , Child , Child, Preschool , Cohort Studies , Cysts/genetics , Female , Genotype , Hereditary Central Nervous System Demyelinating Diseases/genetics , Humans , Kinesins , Male , Membrane Proteins/genetics , Membrane Transport Proteins/genetics , Meta-Analysis as Topic , Middle Aged , Paraplegia/genetics , Phenotype , Young Adult
6.
Int J Epidemiol ; 45(3): 728-40, 2016 06.
Article in English | MEDLINE | ID: mdl-26971321

ABSTRACT

BACKGROUND: Based on epidemiological commonalities, multiple sclerosis (MS) and Hodgkin lymphoma (HL), two clinically distinct conditions, have long been suspected to be aetiologically related. MS and HL occur in roughly the same age groups, both are associated with Epstein-Barr virus infection and ultraviolet (UV) light exposure, and they cluster mutually in families (though not in individuals). We speculated if in addition to sharing environmental risk factors, MS and HL were also genetically related. Using data from genome-wide association studies (GWAS) of 1816 HL patients, 9772 MS patients and 25 255 controls, we therefore investigated the genetic overlap between the two diseases. METHODS: From among a common denominator of 404 K single nucleotide polymorphisms (SNPs) studied, we identified SNPs and human leukocyte antigen (HLA) alleles independently associated with both diseases. Next, we assessed the cumulative genome-wide effect of MS-associated SNPs on HL and of HL-associated SNPs on MS. To provide an interpretational frame of reference, we used data from published GWAS to create a genetic network of diseases within which we analysed proximity of HL and MS to autoimmune diseases and haematological and non-haematological malignancies. RESULTS: SNP analyses revealed genome-wide overlap between HL and MS, most prominently in the HLA region. Polygenic HL risk scores explained 4.44% of HL risk (Nagelkerke R(2)), but also 2.36% of MS risk. Conversely, polygenic MS risk scores explained 8.08% of MS risk and 1.94% of HL risk. In the genetic disease network, HL was closer to autoimmune diseases than to solid cancers. CONCLUSIONS: HL displays considerable genetic overlap with MS and other autoimmune diseases.


Subject(s)
Genome-Wide Association Study , Hodgkin Disease/genetics , Multiple Sclerosis/genetics , Polymorphism, Single Nucleotide , Female , Gene Regulatory Networks , Genetic Predisposition to Disease , Humans , Linear Models , Male
7.
BMC Med Genet ; 16: 55, 2015 Jul 28.
Article in English | MEDLINE | ID: mdl-26212423

ABSTRACT

BACKGROUND: Multiple sclerosis (MS) is an autoimmune disease of the central nervous system, with a strong genetic component. Over 100 genetic loci have been implicated in susceptibility to MS in European populations, the most prominent being the 15:01 allele of the HLA-DRB1 gene. The prevalence of MS is high in European populations including those of Ashkenazi origin, and low in African and Asian populations including those of Jewish origin. METHODS: Here we identified and extracted a total of 213 Ashkenazi MS cases and 546 ethnically matched healthy control individuals from two previous genome-wide case-control association analyses, and 72 trios (affected proband and two unaffected parents) from a previous genome-wide transmission disequilibrium association study, using genetic data to define Ashkenazi. We compared the pattern of genetic risk between Ashkenazi and non-Ashkenazi Europeans. We also sought to identify novel Ashkenazi-specific risk loci by performing association tests on the subset of Ashkenazi cases, controls, probands, and parents from each study. RESULTS: The HLA-DRB1*15:01 allele and the non-HLA risk alleles were present at relatively low frequencies among Ashkenazi and explained a smaller fraction of the population-level risk when compared to non-Ashkenazi Europeans. Alternative HLA susceptibility alleles were identified in an Ashkenazi-only association study, including HLA-A*68:02 and one or both genes in the HLA-B*38:01-HLA-C*12:03 haplotype. The genome-wide screen in Ashkenazi did not reveal any loci associated with MS risk. CONCLUSION: These results suggest that genetic susceptibility to MS in Ashkenazi Jews has not been as well established as that of non-Ashkenazi Europeans. This implies value in studying large well-characterized Ashkenazi populations to accelerate gene discovery in complex genetic diseases.


Subject(s)
Jews/genetics , Multiple Sclerosis/ethnology , Multiple Sclerosis/genetics , Alleles , Case-Control Studies , Family , Female , Gene Frequency , Genetic Predisposition to Disease/ethnology , Genome-Wide Association Study , HLA-A Antigens/genetics , HLA-B38 Antigen/genetics , HLA-C Antigens/genetics , Haplotypes , Humans , Jews/statistics & numerical data , Male , Polymorphism, Single Nucleotide , Risk Factors
8.
Brain ; 138(Pt 6): 1518-30, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25818868

ABSTRACT

The aims of this study were: (i) to determine to what degree multiple sclerosis-associated loci discovered in European populations also influence susceptibility in African Americans; (ii) to assess the extent to which the unique linkage disequilibrium patterns in African Americans can contribute to localizing the functionally relevant regions or genes; and (iii) to search for novel African American multiple sclerosis-associated loci. Using the ImmunoChip custom array we genotyped 803 African American cases with multiple sclerosis and 1516 African American control subjects at 130 135 autosomal single nucleotide polymorphisms. We conducted association analysis with rigorous adjustments for population stratification and admixture. Of the 110 non-major histocompatibility complex multiple sclerosis-associated variants identified in Europeans, 96 passed stringent quality control in our African American data set and of these, >70% (69) showed over-representation of the same allele amongst cases, including 21 with nominally significant evidence for association (one-tailed test P < 0.05). At a further eight loci we found nominally significant association with an alternate correlated risk-tagging single nucleotide polymorphism from the same region. Outside the regions known to be associated in Europeans, we found seven potentially associated novel candidate multiple sclerosis variants (P < 10(-4)), one of which (rs2702180) also showed nominally significant evidence for association (one-tailed test P = 0.034) in an independent second cohort of 620 African American cases and 1565 control subjects. However, none of these novel associations reached genome-wide significance (combined P = 6.3 × 10(-5)). Our data demonstrate substantial overlap between African American and European multiple sclerosis variants, indicating common genetic contributions to multiple sclerosis risk.


Subject(s)
Black or African American/genetics , Genetic Predisposition to Disease/genetics , Multiple Sclerosis/genetics , Oligonucleotide Array Sequence Analysis , Alleles , Case-Control Studies , Genome-Wide Association Study , Genotype , Humans , Linkage Disequilibrium/genetics , Polymorphism, Single Nucleotide/genetics
9.
Neurobiol Aging ; 36(3): 1435-8, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25618617

ABSTRACT

Although numerous genetic variants affecting aging and mortality have been identified, for example, apolipoprotein E ε4, the genetic component influencing cognitive aging has not been fully defined yet. A better knowledge of the genetics of aging will prove helpful in understanding the underlying biological processes. Here, we describe the whole genome sequences of 2 female octogenarians. We provide the repertoire of genomic variants that the 2 octogenarians have in common. We also describe the overlap with the previously reported genomes of 2 supercentenarians­individuals aged ≥110 years. We assessed the genetic disease propensities of the octogenarians and non-aged control genomes and could not find support for the hypothesis that long-lived healthy individuals might exhibit greater genetic fitness than the general population. Furthermore, there is no evidence for an accumulation of previously described variants promoting longevity in the 2 octogenarians. These findings suggest that genetic fitness, as currently defined, is not the sole factor enabling an increased life span. We identified a number of healthy-cognitive-aging candidate genetic loci awaiting confirmation in larger studies.


Subject(s)
Aging/genetics , Aging/psychology , Cognition/physiology , Genome, Human/genetics , Genomics , Aged, 80 and over , Apolipoprotein E4/genetics , Female , Genes, Overlapping , Genetic Predisposition to Disease/genetics , Genetic Variation , Humans , Longevity/genetics , Precision Medicine
10.
Bioinformatics ; 31(2): 262-4, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25260698

ABSTRACT

UNLABELLED: Protein interaction network-based pathway analysis (PINBPA) for genome-wide association studies (GWAS) has been developed as a Cytoscape app, to enable analysis of GWAS data in a network fashion. Users can easily import GWAS summary-level data, draw Manhattan plots, define blocks, prioritize genes with random walk with restart, detect enriched subnetworks and test the significance of subnetworks via a user-friendly interface. AVAILABILITY AND IMPLEMENTATION: PINBPA app is freely available in Cytoscape app store. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Subject(s)
Data Mining/methods , Genome-Wide Association Study , Protein Interaction Maps , Software , Algorithms , Humans , Proteomics
11.
Mult Scler ; 21(7): 894-904, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25392319

ABSTRACT

BACKGROUND: Interferon beta (IFNb) reduces relapse frequency and disability progression in patients with multiple sclerosis (MS). OBJECTIVES: Early identification of prognostic biomarkers of IFNb-treated patients will allow more effective management of MS. METHODS: The IMPROVE study evaluated subcutaneous IFNb versus placebo in 180 patients with relapsing-remitting MS. Magnetic resonance imaging scans, clinical assessments, and blood samples were obtained at baseline and every 4 weeks from every participant. Thirty-nine biomarkers (32 transcripts; seven proteins) were studied in 155 patients from IMPROVE. Therapeutic response was defined by absence of new combined unique lesions, relapses, and sustained increase in Expanded Disability Status Scale over 1 year. A machine learning approach was used to examine the association between biomarker expression and treatment response. RESULTS: While baseline levels of individual genes were relatively poor predictors, combinations of three genes were able to identify subjects with sub-optimal therapeutic responses. The triplet CASP2/IRF4/IRF6, previously identified in an independent dataset, was tested among other combinations. This triplet showed acceptable predictive accuracy (0.68) and specificity (0.88), but had relatively low sensitivity (0.22) resulting in an area under the curve (AUC) of 0.63. Other combinations of biomarkers resulted in AUC of up to 0.80 (e.g. CASP2/IL10/IL12Rb1). CONCLUSIONS: Baseline expression, or induction ratios, of specific gene combinations correlate with future therapeutic response to IFNb, and have the potential to be prognostically useful.


Subject(s)
Biomarkers/analysis , Immunologic Factors/therapeutic use , Interferon-beta/therapeutic use , Multiple Sclerosis, Relapsing-Remitting/drug therapy , Adult , Area Under Curve , Caspase 2/genetics , Cysteine Endopeptidases/genetics , Enzyme-Linked Immunosorbent Assay , Female , Humans , Interferon Regulatory Factors/genetics , Magnetic Resonance Imaging , Male , Multiple Sclerosis, Relapsing-Remitting/genetics , Polymerase Chain Reaction , Prognosis , ROC Curve , Sensitivity and Specificity , Treatment Outcome
12.
Nature ; 509(7499): 189-94, 2014 May 08.
Article in English | MEDLINE | ID: mdl-24776795

ABSTRACT

Astrocytes, the most abundant cells in the central nervous system, promote synapse formation and help to refine neural connectivity. Although they are allocated to spatially distinct regional domains during development, it is unknown whether region-restricted astrocytes are functionally heterogeneous. Here we show that postnatal spinal cord astrocytes express several region-specific genes, and that ventral astrocyte-encoded semaphorin 3a (Sema3a) is required for proper motor neuron and sensory neuron circuit organization. Loss of astrocyte-encoded Sema3a leads to dysregulated α-motor neuron axon initial segment orientation, markedly abnormal synaptic inputs, and selective death of α- but not of adjacent γ-motor neurons. In addition, a subset of TrkA(+) sensory afferents projects to ectopic ventral positions. These findings demonstrate that stable maintenance of a positional cue by developing astrocytes influences multiple aspects of sensorimotor circuit formation. More generally, they suggest that regional astrocyte heterogeneity may help to coordinate postnatal neural circuit refinement.


Subject(s)
Astrocytes/physiology , Motor Neurons/physiology , Neural Pathways/physiology , Sensory Receptor Cells/physiology , Animals , Astrocytes/cytology , Axons/physiology , Cell Polarity , Cell Survival/drug effects , Humans , Mice , Motor Neurons/cytology , Motor Neurons/drug effects , Semaphorin-3A/deficiency , Semaphorin-3A/genetics , Semaphorin-3A/metabolism , Semaphorin-3A/pharmacology , Sensory Receptor Cells/cytology , Spinal Cord/cytology , Synapses/metabolism
13.
BMC Med Genet ; 14: 126, 2013 Dec 07.
Article in English | MEDLINE | ID: mdl-24314052

ABSTRACT

BACKGROUND: Cerebral palsy (CP) is a group of nonprogressive disorders of movement and posture caused by abnormal development of, or damage to, motor control centers of the brain. A single nucleotide polymorphism (SNP), rs1800795, in the promoter region of the interleukin-6 (IL6) gene has been implicated in the pathogenesis of CP by mediating IL-6 protein levels in amniotic fluid and cord plasma and within brain lesions. This SNP has been associated with other neurological, vascular, and malignant processes as well, often as part of a haplotype block. METHODS: To refine the regional genetic association with CP, we sequenced (Sanger) the IL6 gene and part of the promoter region in 250 infants with CP and 305 controls. RESULTS: We identified a haplotype of 7 SNPs that includes rs1800795. In a recessive model of inheritance, the variant haplotype conferred greater risk (OR = 4.3, CI = [2.0-10.1], p = 0.00007) than did the lone variant at rs1800795 (OR = 2.5, CI = [1.4-4.6], p = 0.002). The risk haplotype contains one SNP (rs2069845, CI = [1.2-4.3], OR = 2.3, p = 0.009) that disrupts a methylation site. CONCLUSIONS: The risk haplotype identified in this study overlaps with previously identified haplotypes that include additional promoter SNPs. A risk haplotype at the IL6 gene likely confers risk to CP, and perhaps other diseases, via a multi-factorial mechanism.


Subject(s)
Cerebral Palsy/genetics , Interleukin-6/genetics , Polymorphism, Single Nucleotide , Case-Control Studies , Frameshift Mutation , Genetic Predisposition to Disease , Haplotypes , Humans , Linkage Disequilibrium , Promoter Regions, Genetic
14.
Glia ; 61(9): 1518-32, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23840004

ABSTRACT

Developmental regulation of gliogenesis in the mammalian CNS is incompletely understood, in part due to a limited repertoire of lineage-specific genes. We used Aldh1l1-GFP as a marker for gliogenic radial glia and later-stage precursors of developing astrocytes and performed gene expression profiling of these cells. We then used this dataset to identify candidate transcription factors that may serve as glial markers or regulators of glial fate. Our analysis generated a database of developmental stage-related markers of Aldh1l1+ cells between murine embryonic day 13.5-18.5. Using these data we identify the bZIP transcription factor Nfe2l1 and demonstrate that it promotes glial fate under direct Sox9 regulatory control. Thus, this dataset represents a resource for identifying novel regulators of glial development.


Subject(s)
Gene Expression Regulation, Developmental/physiology , Isoenzymes/metabolism , NF-E2-Related Factor 1/metabolism , Neuroglia/metabolism , Retinal Dehydrogenase/metabolism , SOX9 Transcription Factor/metabolism , Spinal Cord/cytology , Age Factors , Aldehyde Dehydrogenase 1 Family , Animals , Cell Differentiation , Cells, Cultured , Chickens , Computational Biology , Electroporation , Embryo, Mammalian , Flow Cytometry , Gene Expression Profiling , Glial Fibrillary Acidic Protein , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Isoenzymes/genetics , Mice , Mice, Transgenic , NF-E2-Related Factor 1/genetics , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurons/classification , Neurons/metabolism , Oligonucleotide Array Sequence Analysis , Retinal Dehydrogenase/genetics , SOX9 Transcription Factor/genetics , Spinal Cord/embryology , Transcription Factors/genetics , Transcription Factors/metabolism
15.
Hum Mol Genet ; 22(20): 4194-205, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-23748426

ABSTRACT

Multiple sclerosis (MS) is the most common autoimmune disease of the central nervous system (CNS). It is characterized by the infiltration of autoreactive immune cells into the CNS, which target the myelin sheath, leading to the loss of neuronal function. Although it is accepted that MS is a multifactorial disorder with both genetic and environmental factors influencing its development and course, the molecular pathogenesis of MS has not yet been fully elucidated. Here, we studied the longitudinal gene expression profiles of whole-blood RNA from a cohort of 195 MS patients and 66 healthy controls. We analyzed these transcriptomes at both the individual transcript and the biological pathway level. We found 62 transcripts to be significantly up-regulated in MS patients; the expression of 11 of these genes was counter-regulated by interferon treatment, suggesting partial restoration of a 'healthy' gene expression profile. Global pathway analyses linked the proteasome and Wnt signaling to MS disease processes. Since genotypes from a subset of individuals were available, we were able to identify expression quantitative trait loci (eQTL), a number of which involved two genes of the MS gene signature. However, all these eQTL were also present in healthy controls. This study highlights the challenge posed by analyzing transcripts from whole blood and how these can be mitigated by using large, well-characterized cohorts of patients with longitudinal follow-up and multi-modality measurements.


Subject(s)
Gene Expression Profiling , Multiple Sclerosis/genetics , RNA/blood , RNA/genetics , Adult , Aged , Case-Control Studies , Female , Gene Expression Regulation , Humans , Interferons/therapeutic use , Longitudinal Studies , Male , Middle Aged , Multiple Sclerosis/drug therapy , Multiple Sclerosis/immunology , Multiple Sclerosis/metabolism , Proteasome Endopeptidase Complex/genetics , Quantitative Trait Loci , Transcriptome , Wnt Signaling Pathway/genetics , Young Adult
16.
BMC Genomics ; 13: 477, 2012 Sep 14.
Article in English | MEDLINE | ID: mdl-22974163

ABSTRACT

BACKGROUND: A detailed analysis of whole genomes can be now achieved with next generation sequencing. Epstein Barr Virus (EBV) transformation is a widely used strategy in clinical research to obtain an unlimited source of a subject's DNA. Although the mechanism of transformation and immortalization by EBV is relatively well known at the transcriptional and proteomic level, the genetic consequences of EBV transformation are less well understood. A detailed analysis of the genetic alterations introduced by EBV transformation is highly relevant, as it will inform on the usefulness and limitations of this approach. RESULTS: We used whole genome sequencing to assess the genomic signature of a low-passage lymphoblastoid cell line (LCL). Specifically, we sequenced the full genome (40X) of an individual using DNA purified from fresh whole blood as well as DNA from his LCL. A total of 217.33 Gb of sequence were generated from the cell line and 238.95 Gb from the normal genomic DNA. We determined with high confidence that 99.2% of the genomes were identical, with no reproducible changes in structural variation (chromosomal rearrangements and copy number variations) or insertion/deletion polymorphisms (indels). CONCLUSIONS: Our results suggest that, at this level of resolution, the LCL is genetically indistinguishable from its genomic counterpart and therefore their use in clinical research is not likely to introduce a significant bias.


Subject(s)
DNA/genetics , Genome, Viral/genetics , Herpesvirus 4, Human/genetics , Cell Line , Cell Transformation, Viral/genetics , Humans
17.
J Med Genet ; 48(6): 375-82, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21507892

ABSTRACT

BACKGROUND: Manitoba-oculo-tricho-anal (MOTA) syndrome is a rare condition defined by eyelid colobomas, cryptophthalmos and anophthalmia/microphthalmia, an aberrant hairline, a bifid or broad nasal tip, and gastrointestinal anomalies such as omphalocele and anal stenosis. Autosomal recessive inheritance had been assumed because of consanguinity in the Oji-Cre population of Manitoba and reports of affected siblings, but no locus or cytogenetic aberration had previously been described. METHODS AND RESULTS: This study shows that MOTA syndrome is caused by mutations in FREM1, a gene previously mutated in bifid nose, renal agenesis, and anorectal malformations (BNAR) syndrome. MOTA syndrome and BNAR syndrome can therefore be considered as part of a phenotypic spectrum that is similar to, but distinct from and less severe than, Fraser syndrome. Re-examination of Frem1(bat/bat) mutant mice found new evidence that Frem1 is involved in anal and craniofacial development, with anal prolapse, eyelid colobomas, telecanthus, a shortened snout and reduced philtral height present in the mutant mice, similar to the human phenotype in MOTA syndrome. CONCLUSIONS: The milder phenotypes associated with FREM1 deficiency in humans (MOTA syndrome and BNAR syndrome) compared to that resulting from FRAS1 and FREM2 loss of function (Fraser syndrome) are also consistent with the less severe phenotypes resulting from Frem1 loss of function in mice. Together, Fraser, BNAR and MOTA syndromes constitute a clinically overlapping group of FRAS-FREM complex diseases.


Subject(s)
Abnormalities, Multiple/genetics , Anus, Imperforate/genetics , Coloboma/genetics , Extracellular Matrix Proteins/genetics , Fraser Syndrome/genetics , Hypertelorism/genetics , Receptors, Interleukin/genetics , Abnormalities, Multiple/pathology , Adolescent , Adult , Anal Canal/abnormalities , Anal Canal/pathology , Animals , Anorectal Malformations , Anus, Imperforate/pathology , Base Sequence , Child , Child, Preschool , Coloboma/pathology , Eyelids/abnormalities , Female , Fraser Syndrome/pathology , Gene Dosage , Hernia, Umbilical/genetics , Hernia, Umbilical/pathology , Humans , Hypertelorism/pathology , Male , Mice , Molecular Sequence Data , Mutation , Nose/abnormalities , Nose Diseases/genetics , Oligonucleotide Array Sequence Analysis , Pedigree , Phenotype , Reverse Transcriptase Polymerase Chain Reaction , Syndrome
18.
J Proteome Res ; 8(11): 4954-65, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19764811

ABSTRACT

Protein phosphorylation plays an essential role in signal transduction pathways that regulate substrate and energy metabolism, contractile function, and muscle mass in human skeletal muscle. Abnormal phosphorylation of signaling enzymes has been identified in insulin-resistant muscle using phosphoepitope-specific antibodies, but its role in other skeletal muscle disorders remains largely unknown. This may be in part due to insufficient knowledge of relevant targets. Here, we therefore present the first large-scale in vivo phosphoproteomic study of human skeletal muscle from 3 lean, healthy volunteers. Trypsin digestion of 3-5 mg human skeletal muscle protein was followed by phosphopeptide enrichment using SCX and TiO(2). The resulting phosphopeptides were analyzed by HPLC-ESI-MS/MS. Using this unbiased approach, we identified 306 distinct in vivo phosphorylation sites in 127 proteins, including 240 phosphoserines, 53 phosphothreonines, and 13 phosphotyrosines in at least 2 out of 3 subjects. In addition, 61 ambiguous phosphorylation sites were identified in at least 2 out of 3 subjects. The majority of phosphoproteins detected are involved in sarcomeric function, excitation-contraction coupling (the Ca(2+)-cycle), glycolysis, and glycogen metabolism. Of particular interest, we identified multiple novel phosphorylation sites on several sarcomeric Z-disk proteins known to be involved in signaling and muscle disorders. These results provide numerous new targets for the investigation of human skeletal muscle phosphoproteins in health and disease and demonstrate feasibility of phosphoproteomics research of human skeletal muscle in vivo.


Subject(s)
Chromatography, High Pressure Liquid/methods , Muscle Proteins/analysis , Muscle, Skeletal/chemistry , Phosphopeptides/chemistry , Spectrometry, Mass, Electrospray Ionization/methods , Tandem Mass Spectrometry/methods , Adult , Amino Acid Sequence , Computational Biology , Humans , Male , Middle Aged , Molecular Sequence Data , Phosphorylation , Proteome/analysis , Proteomics/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...