Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Cell ; 149(3): 525-37, 2012 Apr 27.
Article in English | MEDLINE | ID: mdl-22521361

ABSTRACT

Balanced chromosomal abnormalities (BCAs) represent a relatively untapped reservoir of single-gene disruptions in neurodevelopmental disorders (NDDs). We sequenced BCAs in patients with autism or related NDDs, revealing disruption of 33 loci in four general categories: (1) genes previously associated with abnormal neurodevelopment (e.g., AUTS2, FOXP1, and CDKL5), (2) single-gene contributors to microdeletion syndromes (MBD5, SATB2, EHMT1, and SNURF-SNRPN), (3) novel risk loci (e.g., CHD8, KIRREL3, and ZNF507), and (4) genes associated with later-onset psychiatric disorders (e.g., TCF4, ZNF804A, PDE10A, GRIN2B, and ANK3). We also discovered among neurodevelopmental cases a profoundly increased burden of copy-number variants from these 33 loci and a significant enrichment of polygenic risk alleles from genome-wide association studies of autism and schizophrenia. Our findings suggest a polygenic risk model of autism and reveal that some neurodevelopmental genes are sensitive to perturbation by multiple mutational mechanisms, leading to variable phenotypic outcomes that manifest at different life stages.


Subject(s)
Child Development Disorders, Pervasive/genetics , Chromosome Aberrations , Autistic Disorder/diagnosis , Autistic Disorder/genetics , Child , Child Development Disorders, Pervasive/diagnosis , Chromosome Breakage , Chromosome Deletion , DNA Copy Number Variations , Genetic Predisposition to Disease , Genome-Wide Association Study , Humans , Nervous System/growth & development , Schizophrenia/genetics , Sequence Analysis, DNA , Signal Transduction
2.
Am J Hum Genet ; 99(5): 1015-1033, 2016 Nov 03.
Article in English | MEDLINE | ID: mdl-27745839

ABSTRACT

In this exciting era of "next-gen cytogenetics," integrating genomic sequencing into the prenatal diagnostic setting is possible within an actionable time frame and can provide precise delineation of balanced chromosomal rearrangements at the nucleotide level. Given the increased risk of congenital abnormalities in newborns with de novo balanced chromosomal rearrangements, comprehensive interpretation of breakpoints could substantially improve prediction of phenotypic outcomes and support perinatal medical care. Herein, we present and evaluate sequencing results of balanced chromosomal rearrangements in ten prenatal subjects with respect to the location of regulatory chromatin domains (topologically associated domains [TADs]). The genomic material from all subjects was interpreted to be "normal" by microarray analyses, and their rearrangements would not have been detected by cell-free DNA (cfDNA) screening. The findings of our systematic approach correlate with phenotypes of both pregnancies with untoward outcomes (5/10) and with healthy newborns (3/10). Two pregnancies, one with a chromosomal aberration predicted to be of unknown clinical significance and another one predicted to be likely benign, were terminated prior to phenotype-genotype correlation (2/10). We demonstrate that the clinical interpretation of structural rearrangements should not be limited to interruption, deletion, or duplication of specific genes and should also incorporate regulatory domains of the human genome with critical ramifications for the control of gene expression. As detailed in this study, our molecular approach to both detecting and interpreting the breakpoints of structural rearrangements yields unparalleled information in comparison to other commonly used first-tier diagnostic methods, such as non-invasive cfDNA screening and microarray analysis, to provide improved genetic counseling for phenotypic outcome in the prenatal setting.


Subject(s)
Chromosome Aberrations , Congenital Abnormalities/genetics , Gene Rearrangement , Nucleotides/genetics , Prenatal Diagnosis/methods , Alleles , Chromosome Mapping , Congenital Abnormalities/diagnosis , Female , Gene Expression Regulation , Genetic Testing , Genome, Human , Genomics , High-Throughput Nucleotide Sequencing , Humans , Karyotyping , Male , Pregnancy , SOX9 Transcription Factor/genetics , SOX9 Transcription Factor/metabolism , Sequence Analysis, DNA , Translocation, Genetic
3.
Am J Hum Genet ; 96(5): 784-96, 2015 May 07.
Article in English | MEDLINE | ID: mdl-25937446

ABSTRACT

The 16p11.2 600 kb copy-number variants (CNVs) are associated with mirror phenotypes on BMI, head circumference, and brain volume and represent frequent genetic lesions in autism spectrum disorders (ASDs) and schizophrenia. Here we interrogated the transcriptome of individuals carrying reciprocal 16p11.2 CNVs. Transcript perturbations correlated with clinical endophenotypes and were enriched for genes associated with ASDs, abnormalities of head size, and ciliopathies. Ciliary gene expression was also perturbed in orthologous mouse models, raising the possibility that ciliary dysfunction contributes to 16p11.2 pathologies. In support of this hypothesis, we found structural ciliary defects in the CA1 hippocampal region of 16p11.2 duplication mice. Moreover, by using an established zebrafish model, we show genetic interaction between KCTD13, a key driver of the mirrored neuroanatomical phenotypes of the 16p11.2 CNV, and ciliopathy-associated genes. Overexpression of BBS7 rescues head size and neuroanatomical defects of kctd13 morphants, whereas suppression or overexpression of CEP290 rescues phenotypes induced by KCTD13 under- or overexpression, respectively. Our data suggest that dysregulation of ciliopathy genes contributes to the clinical phenotypes of these CNVs.


Subject(s)
Child Development Disorders, Pervasive/genetics , Chromosomes, Human, Pair 16/genetics , DNA Copy Number Variations/genetics , Schizophrenia/genetics , Animals , Brain , Child , Child Development Disorders, Pervasive/pathology , Chromosome Deletion , Ciliary Body/metabolism , Ciliary Body/pathology , Gene Expression Regulation , Humans , Mice , Potassium Channels, Voltage-Gated/genetics , Schizophrenia/pathology , Transcriptome , Zebrafish , Zebrafish Proteins/genetics
4.
Genome Res ; 25(2): 155-66, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25561519

ABSTRACT

RNA polymerase III (Pol III) synthesizes tRNAs and other small noncoding RNAs to regulate protein synthesis. Dysregulation of Pol III transcription has been linked to cancer, and germline mutations in genes encoding Pol III subunits or tRNA processing factors cause neurogenetic disorders in humans, such as hypomyelinating leukodystrophies and pontocerebellar hypoplasia. Here we describe an autosomal recessive disorder characterized by cerebellar hypoplasia and intellectual disability, as well as facial dysmorphic features, short stature, microcephaly, and dental anomalies. Whole-exome sequencing revealed biallelic missense alterations of BRF1 in three families. In support of the pathogenic potential of the discovered alleles, suppression or CRISPR-mediated deletion of brf1 in zebrafish embryos recapitulated key neurodevelopmental phenotypes; in vivo complementation showed all four candidate mutations to be pathogenic in an apparent isoform-specific context. BRF1 associates with BDP1 and TBP to form the transcription factor IIIB (TFIIIB), which recruits Pol III to target genes. We show that disease-causing mutations reduce Brf1 occupancy at tRNA target genes in Saccharomyces cerevisiae and impair cell growth. Moreover, BRF1 mutations reduce Pol III-related transcription activity in vitro. Taken together, our data show that BRF1 mutations that reduce protein activity cause neurodevelopmental anomalies, suggesting that BRF1-mediated Pol III transcription is required for normal cerebellar and cognitive development.


Subject(s)
Abnormalities, Multiple/genetics , Intellectual Disability/genetics , Mutation , RNA Polymerase III/metabolism , TATA-Binding Protein Associated Factors/genetics , Transcription, Genetic , Abnormalities, Multiple/diagnosis , Adolescent , Amino Acid Sequence , Amino Acid Substitution , Animals , Brain/pathology , Cell Proliferation , Child , Child, Preschool , Exome , Facies , Female , High-Throughput Nucleotide Sequencing , Humans , Infant , Intellectual Disability/diagnosis , Magnetic Resonance Imaging , Male , Models, Molecular , Molecular Sequence Data , Pedigree , Phenotype , Protein Conformation , Protein Isoforms , Siblings , Syndrome , TATA-Binding Protein Associated Factors/chemistry , TATA-Binding Protein Associated Factors/metabolism , Zebrafish
5.
Am J Hum Genet ; 94(6): 870-83, 2014 Jun 05.
Article in English | MEDLINE | ID: mdl-24906019

ABSTRACT

Reciprocal copy-number variation (CNV) of a 593 kb region of 16p11.2 is a common genetic cause of autism spectrum disorder (ASD), yet it is not completely penetrant and can manifest in a wide array of phenotypes. To explore its molecular consequences, we performed RNA sequencing of cerebral cortex from mouse models with CNV of the syntenic 7qF3 region and lymphoblast lines from 34 members of 7 multiplex ASD-affected families harboring the 16p11.2 CNV. Expression of all genes in the CNV region correlated well with their DNA copy number, with no evidence of dosage compensation. We observed effects on gene expression outside the CNV region, including apparent positional effects in cis and in trans at genomic segments with evidence of physical interaction in Hi-C chromosome conformation data. One of the most significant positional effects was telomeric to the 16p11.2 CNV and includes the previously described "distal" 16p11.2 microdeletion. Overall, 16p11.2 CNV was associated with altered expression of genes and networks that converge on multiple hypotheses of ASD pathogenesis, including synaptic function (e.g., NRXN1, NRXN3), chromatin modification (e.g., CHD8, EHMT1, MECP2), transcriptional regulation (e.g., TCF4, SATB2), and intellectual disability (e.g., FMR1, CEP290). However, there were differences between tissues and species, with the strongest effects being consistently within the CNV region itself. Our analyses suggest that through a combination of indirect regulatory effects and direct effects on nuclear architecture, alteration of 16p11.2 genes disrupts expression networks that involve other genes and pathways known to contribute to ASD, suggesting an overlap in mechanisms of pathogenesis.


Subject(s)
Autistic Disorder/genetics , Chromosome Deletion , Chromosome Duplication , Chromosomes, Human, Pair 16/genetics , Animals , Cerebral Cortex/pathology , Child , DNA Copy Number Variations , Female , Genome-Wide Association Study , Genotype , Humans , Intellectual Disability/genetics , Male , Mice , Pedigree , Phenotype , Sequence Analysis, RNA , Transcription, Genetic
6.
Am J Med Genet A ; 173(2): 395-406, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27759917

ABSTRACT

We performed whole-genome sequencing on an individual from a family with variable psychiatric phenotypes that had a sensory processing disorder, apraxia, and autism. The proband harbored a maternally inherited balanced translocation (46,XY,t(11;14)(p12;p12)mat) that disrupted LRRC4C, a member of the highly specialized netrin G family of axon guidance molecules. The proband also inherited a paternally derived chromosomal inversion that disrupted DPP6, a potassium channel interacting protein. Copy Number (CN) analysis in 14,077 cases with neurodevelopmental disorders and 8,960 control subjects revealed that 60% of cases with exonic deletions in LRRC4C had a second clinically recognizable syndrome associated with variable clinical phenotypes, including 16p11.2, 1q44, and 2q33.1 CN syndromes, suggesting LRRC4C deletion variants may be modifiers of neurodevelopmental disorders. In vitro, functional assessments modeling patient deletions in LRRC4C suggest a negative regulatory role of these exons found in the untranslated region of LRRC4C, which has a single, terminal coding exon. These data suggest that the proband's autism may be due to the inheritance of disruptions in both DPP6 and LRRC4C, and may highlight the importance of the netrin G family and potassium channel interacting molecules in neurodevelopmental disorders. © 2016 Wiley Periodicals, Inc.


Subject(s)
Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/genetics , Genetic Association Studies , Nerve Tissue Proteins/genetics , Neurodevelopmental Disorders/diagnosis , Neurodevelopmental Disorders/genetics , Phenotype , Potassium Channels/genetics , Receptors, Cell Surface/genetics , 5' Untranslated Regions , Adolescent , Adult , Apraxias/diagnosis , Apraxias/genetics , Autistic Disorder/diagnosis , Autistic Disorder/genetics , Child , Child, Preschool , Chromosome Breakpoints , Chromosome Inversion , Comparative Genomic Hybridization , DNA Copy Number Variations , Female , Gene Expression , Gene Expression Profiling , High-Throughput Nucleotide Sequencing , Humans , Karyotype , Male , Middle Aged , Multigene Family , Pedigree , Translocation, Genetic , Young Adult
7.
Proc Natl Acad Sci U S A ; 111(42): E4468-77, 2014 Oct 21.
Article in English | MEDLINE | ID: mdl-25294932

ABSTRACT

Truncating mutations of chromodomain helicase DNA-binding protein 8 (CHD8), and of many other genes with diverse functions, are strong-effect risk factors for autism spectrum disorder (ASD), suggesting multiple mechanisms of pathogenesis. We explored the transcriptional networks that CHD8 regulates in neural progenitor cells (NPCs) by reducing its expression and then integrating transcriptome sequencing (RNA sequencing) with genome-wide CHD8 binding (ChIP sequencing). Suppressing CHD8 to levels comparable with the loss of a single allele caused altered expression of 1,756 genes, 64.9% of which were up-regulated. CHD8 showed widespread binding to chromatin, with 7,324 replicated sites that marked 5,658 genes. Integration of these data suggests that a limited array of direct regulatory effects of CHD8 produced a much larger network of secondary expression changes. Genes indirectly down-regulated (i.e., without CHD8-binding sites) reflect pathways involved in brain development, including synapse formation, neuron differentiation, cell adhesion, and axon guidance, whereas CHD8-bound genes are strongly associated with chromatin modification and transcriptional regulation. Genes associated with ASD were strongly enriched among indirectly down-regulated loci (P < 10(-8)) and CHD8-bound genes (P = 0.0043), which align with previously identified coexpression modules during fetal development. We also find an intriguing enrichment of cancer-related gene sets among CHD8-bound genes (P < 10(-10)). In vivo suppression of chd8 in zebrafish produced macrocephaly comparable to that of humans with inactivating mutations. These data indicate that heterozygous disruption of CHD8 precipitates a network of gene-expression changes involved in neurodevelopmental pathways in which many ASD-associated genes may converge on shared mechanisms of pathogenesis.


Subject(s)
Child Development Disorders, Pervasive/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/physiology , Gene Expression Regulation, Developmental , Neural Stem Cells/physiology , Transcription Factors/genetics , Transcription Factors/physiology , Zebrafish Proteins/physiology , Animals , Axons/metabolism , Binding Sites , Child Development Disorders, Pervasive/metabolism , Chromatin/metabolism , DNA Helicases/metabolism , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , Genome , Heterozygote , Humans , Megalencephaly/metabolism , Mutation , Neoplasms/metabolism , Neurons/metabolism , Protein Binding , Risk Factors , Sequence Analysis, RNA , Software , Zebrafish , Zebrafish Proteins/genetics
8.
Am J Hum Genet ; 92(3): 375-86, 2013 Mar 07.
Article in English | MEDLINE | ID: mdl-23472757

ABSTRACT

NRXN1 microdeletions occur at a relatively high frequency and confer increased risk for neurodevelopmental and neurobehavioral abnormalities. The mechanism that makes NRXN1 a deletion hotspot is unknown. Here, we identified deletions of the NRXN1 region in affected cohorts, confirming a strong association with the autism spectrum and other neurodevelopmental disorders. Interestingly, deletions in both affected and control individuals were clustered in the 5' portion of NRXN1 and its immediate upstream region. To explore the mechanism of deletion, we mapped and analyzed the breakpoints of 32 deletions. At the deletion breakpoints, frequent microhomology (68.8%, 2-19 bp) suggested predominant mechanisms of DNA replication error and/or microhomology-mediated end-joining. Long terminal repeat (LTR) elements, unique non-B-DNA structures, and MEME-defined sequence motifs were significantly enriched, but Alu and LINE sequences were not. Importantly, small-size inverted repeats (minus self chains, minus sequence motifs, and partial complementary sequences) were significantly overrepresented in the vicinity of NRXN1 region deletion breakpoints, suggesting that, although they are not interrupted by the deletion process, such inverted repeats can predispose a region to genomic instability by mediating single-strand DNA looping via the annealing of partially reverse complementary strands and the promoting of DNA replication fork stalling and DNA replication error. Our observations highlight the potential importance of inverted repeats of variable sizes in generating a rearrangement hotspot in which individual breakpoints are not recurrent. Mechanisms that involve short inverted repeats in initiating deletion may also apply to other deletion hotspots in the human genome.


Subject(s)
Cell Adhesion Molecules, Neuronal/genetics , DNA Copy Number Variations , Inverted Repeat Sequences , Mental Disorders/genetics , Nerve Tissue Proteins/genetics , Sequence Deletion , Calcium-Binding Proteins , Cohort Studies , DNA Replication/genetics , DNA, B-Form/genetics , DNA, Single-Stranded/genetics , Exons , Genetic Predisposition to Disease , Genomic Instability , Humans , Neural Cell Adhesion Molecules , Terminal Repeat Sequences
9.
Neurodegener Dis ; 16(3-4): 245-59, 2016.
Article in English | MEDLINE | ID: mdl-26959244

ABSTRACT

BACKGROUND: Huntington's disease is induced by CAG expansion in a single gene coding the huntingtin protein. The mutated huntingtin (mtHtt) primarily causes degeneration of neurons in the brain, but it also affects peripheral tissues, including testes. OBJECTIVE: We studied sperm and testes of transgenic boars expressing the N-terminal region of human mtHtt. METHODS: In this study, measures of reproductive parameters and electron microscopy (EM) images of spermatozoa and testes of transgenic (TgHD) and wild-type (WT) boars of F1 (24-48 months old) and F2 (12-36 months old) generations were compared. In addition, immunofluorescence, immunohistochemistry, Western blot, hormonal analysis and whole-genome sequencing were done in order to elucidate the effects of mtHtt. RESULTS: Evidence for fertility failure of both TgHD generations was observed at the age of 13 months. Reproductive parameters declined and progressively worsened with age. EM revealed numerous pathological features in sperm tails and in testicular epithelium from 24- and 36-month-old TgHD boars. Moreover, immunohistochemistry confirmed significantly lower proliferation activity of spermatogonia in transgenic testes. mtHtt was highly expressed in spermatozoa and testes of TgHD boars and localized in all cells of seminiferous tubules. Levels of fertility-related hormones did not differ in TgHD and WT siblings. Genome analysis confirmed that insertion of the lentiviral construct did not interrupt any coding sequence in the pig genome. CONCLUSIONS: The sperm and testicular degeneration of TgHD boars is caused by gain-of-function of the highly expressed mtHtt.


Subject(s)
Huntingtin Protein/metabolism , Mutation , Spermatozoa/metabolism , Spermatozoa/pathology , Testis/metabolism , Testis/pathology , Aging/metabolism , Aging/pathology , Animals , Animals, Genetically Modified , Cell Proliferation/physiology , Disease Models, Animal , Genetic Vectors , Humans , Huntingtin Protein/genetics , Huntington Disease/metabolism , Huntington Disease/pathology , Lentivirus/genetics , Male , Sperm Count , Swine , Swine, Miniature
10.
Am J Hum Genet ; 91(6): 1128-34, 2012 Dec 07.
Article in English | MEDLINE | ID: mdl-23217328

ABSTRACT

Large intergenic noncoding (linc) RNAs represent a newly described class of ribonucleic acid whose importance in human disease remains undefined. We identified a severely developmentally delayed 16-year-old female with karyotype 46,XX,t(2;11)(p25.1;p15.1)dn in the absence of clinically significant copy number variants (CNVs). DNA capture followed by next-generation sequencing of the translocation breakpoints revealed disruption of a single noncoding gene on chromosome 2, LINC00299, whose RNA product is expressed in all tissues measured, but most abundantly in brain. Among a series of additional, unrelated subjects referred for clinical diagnostic testing who showed CNV affecting this locus, we identified four with exon-crossing deletions in association with neurodevelopmental abnormalities. No disruption of the LINC00299 coding sequence was seen in almost 14,000 control subjects. Together, these subjects with disruption of LINC00299 implicate this particular noncoding RNA in brain development and raise the possibility that, as a class, abnormalities of lincRNAs may play a significant role in human developmental disorders.


Subject(s)
Developmental Disabilities/genetics , Mutation , RNA, Long Noncoding/genetics , Adolescent , Alternative Splicing , Base Sequence , Chromosome Breakpoints , Chromosomes, Human, Pair 11 , Chromosomes, Human, Pair 2 , Female , Gene Order , Humans , Lymphocytes/metabolism , Molecular Sequence Data , Neural Stem Cells/metabolism , Translocation, Genetic
11.
N Engl J Med ; 367(23): 2226-32, 2012 Dec 06.
Article in English | MEDLINE | ID: mdl-23215558

ABSTRACT

Conventional cytogenetic testing offers low-resolution detection of balanced karyotypic abnormalities but cannot provide the precise, gene-level knowledge required to predict outcomes. The use of high-resolution whole-genome deep sequencing is currently impractical for the purpose of routine clinical care. We show here that whole-genome "jumping libraries" can offer an immediately applicable, nucleotide-level complement to conventional genetic diagnostics within a time frame that allows for clinical action. We performed large-insert sequencing of DNA extracted from amniotic-fluid cells with a balanced de novo translocation. The amniotic-fluid sample was from a patient in the third trimester of pregnancy who underwent amniocentesis because of severe polyhydramnios after multiple fetal anomalies had been detected on ultrasonography. Using a 13-day sequence and analysis pipeline, we discovered direct disruption of CHD7, a causal locus in the CHARGE syndrome (coloboma of the eye, heart anomaly, atresia of the choanae, retardation, and genital and ear anomalies). Clinical findings at birth were consistent with the CHARGE syndrome, a diagnosis that could not have been reliably inferred from the cytogenetic breakpoint. This case study illustrates the potential power of customized whole-genome jumping libraries when used to augment prenatal karyotyping.


Subject(s)
CHARGE Syndrome/genetics , Chromosome Disorders/diagnosis , Genetic Testing/methods , Genomic Library , Heart Defects, Congenital/genetics , Oligonucleotide Array Sequence Analysis , Prenatal Diagnosis/methods , Adult , CHARGE Syndrome/diagnosis , Chromosome Aberrations , Female , Fetal Diseases/diagnosis , Genome, Human , Heart Defects, Congenital/diagnostic imaging , Humans , Karyotype , Mutation , Pregnancy , Translocation, Genetic , Ultrasonography, Prenatal
12.
Hum Genet ; 132(5): 537-52, 2013 May.
Article in English | MEDLINE | ID: mdl-23354975

ABSTRACT

We describe a female subject (DGAP100) with a 46,X,t(X;5)(p11.3;q35.3)inv(5)(q35.3q35.1)dn, severe psychomotor retardation with hypotonia, global postnatal growth restriction, microcephaly, globally reduced cerebral volume, seizures, facial dysmorphia and cleft palate. Fluorescence in situ hybridization and whole-genome sequencing demonstrated that the X chromosome breakpoint disrupts KDM6A in the second intron. No genes were directly disrupted on chromosome 5. KDM6A is a histone 3 lysine 27 demethylase and a histone 3 lysine 4 methyltransferase. Expression of KDM6A is significantly reduced in DGAP100 lymphoblastoid cells compared to control samples. We identified nine additional cases with neurodevelopmental delay and various other features consistent with the DGAP100 phenotype with copy number variation encompassing KDM6A from microarray databases. We evaluated haploinsufficiency of kdm6a in a zebrafish model. kdm6a is expressed in the pharyngeal arches and ethmoid plate of the developing zebrafish, while a kdm6a morpholino knockdown exhibited craniofacial defects. We conclude KDM6A dosage regulation is associated with severe and diverse structural defects and developmental abnormalities.


Subject(s)
Abnormalities, Multiple/genetics , Chromosomes, Human, Pair 5 , Haploinsufficiency/genetics , Histone Demethylases/genetics , Nuclear Proteins/genetics , X Chromosome , Animals , Branchial Region/enzymology , Cell Line , Chromosomes, Human, Pair 5/genetics , Cleft Palate/genetics , Comparative Genomic Hybridization , DNA Copy Number Variations , Female , Gene Knockdown Techniques , Histone Demethylases/metabolism , Humans , Intellectual Disability/genetics , Karyotyping , Microcephaly/genetics , Muscle Hypotonia/genetics , Nuclear Proteins/metabolism , Phenotype , Psychomotor Disorders/genetics , Seizures/genetics , Translocation, Genetic , X Chromosome/genetics , Young Adult , Zebrafish/embryology , Zebrafish/genetics , Zebrafish/growth & development , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
14.
Hum Mutat ; 33(4): 728-40, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22290657

ABSTRACT

SOX5 encodes a transcription factor involved in the regulation of chondrogenesis and the development of the nervous system. Despite its important developmental roles, SOX5 disruption has yet to be associated with human disease. We report one individual with a reciprocal translocation breakpoint within SOX5, eight individuals with intragenic SOX5 deletions (four are apparently de novo and one inherited from an affected parent), and seven individuals with larger 12p12 deletions encompassing SOX5. Common features in these subjects include prominent speech delay, intellectual disability, behavior abnormalities, and dysmorphic features. The phenotypic impact of the deletions may depend on the location of the deletion and, consequently, which of the three major SOX5 protein isoforms are affected. One intragenic deletion, involving only untranslated exons, was present in a more mildly affected subject, was inherited from a healthy parent and grandparent, and is similar to a deletion found in a control cohort. Therefore, some intragenic SOX5 deletions may have minimal phenotypic effect. Based on the location of the deletions in the subjects compared to the controls, the de novo nature of most of these deletions, and the phenotypic similarities among cases, SOX5 appears to be a dosage-sensitive, developmentally important gene.


Subject(s)
Body Dysmorphic Disorders/genetics , Developmental Disabilities/genetics , Haploinsufficiency , Language Development Disorders/genetics , Mental Disorders/genetics , SOXD Transcription Factors/genetics , Adolescent , Adult , Case-Control Studies , Child , Child, Preschool , Chromosomes, Human, Pair 12 , Female , Humans , Male
15.
Adv Healthc Mater ; 11(9): e2101944, 2022 05.
Article in English | MEDLINE | ID: mdl-34889072

ABSTRACT

Engineered immune cells are an exciting therapeutic modality, which survey and attack tumors. Backpacking strategies exploit cell targeting capabilities for delivery of drugs to combat tumors and their immune-suppressive environments. Here, a new platform for arming cell therapeutics through dual receptor and polymeric prodrug engineering is developed. Macrophage and T cell therapeutics are engineered to express a bioorthogonal single chain variable fragment receptor. The receptor binds a fluorescein ligand that directs cell loading with ligand-tagged polymeric prodrugs, termed "drugamers." The fluorescein ligand facilitates stable binding of drugamer to engineered macrophages over 10 days with 80% surface retention. Drugamers also incorporate prodrug monomers of the phosphoinositide-3-kinase inhibitor, PI-103. The extended release of PI-103 from the drugamer sustains antiproliferative activity against a glioblastoma cell line compared to the parent drug. The versatility and modularity of this cell arming system is demonstrated by loading T cells with a second fluorescein-drugamer. This drugamer incorporates a small molecule estrogen analog, CMP8, which stabilizes a degron-tagged transgene to provide temporal regulation of protein activity in engineered T cells. These results demonstrate that this bioorthogonal receptor and drugamer system can be used to arm multiple immune cell classes with both antitumor and transgene-activating small molecule prodrugs.


Subject(s)
Neoplasms , Prodrugs , Fluoresceins , Humans , Ligands , Polymers/chemistry , Prodrugs/chemistry , Prodrugs/pharmacology
16.
Hepatology ; 52(4): 1350-9, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20803559

ABSTRACT

UNLABELLED: Immune-mediated liver injury in hepatitis is due to activated T cells producing interferon-γ (IFN-γ). It is important to identify negative feedback immune mechanisms that can regulate T cell activity. In this study, we demonstrate that liver inflammation mediated by type 1 T helper (Th1) cells can induce the accumulation of myeloid-derived suppressor cells (MDSCs), pleiomorphic cells capable of modulating T cell-mediated immunity, that heretofore have been studied almost exclusively in the context of tumor-associated inflammation. Mice deficient in the gene encoding transforming growth factor-ß1 (Tgfb1(-/-) mice) acutely develop liver necroinflammation caused by IFN-γ-producing clusters of differentiation 4-positive (CD4(+)) T cells. Liver Th1 cell accumulation was accompanied by myeloid cells expressing CD11b and Gr1, phenotypic hallmarks of MDSCs. Isolated Tgfb1(-/-) liver CD11b(+)Gr1(+) cells were functional MDSCs, readily suppressing T cell proliferation in vitro. Pharmacologic inhibitors of inducible nitric oxide (NO) synthase completely eliminated suppressor function. Suppressor function and the production of NO were dependent on cell-cell contact between MDSCs and T cells, and upon IFN-γ, and were specifically associated with the "monocytic" CD11b(+)Ly6G(-) Ly6C(hi) subset of liver Tgfb1(-/-) CD11b(+) cells. The rapid accumulation of CD11b(+)Gr1(+) cells in Tgfb1(-/-) liver was abrogated when mice were either depleted of CD4(+) T cells or rendered unable to produce IFN-γ, showing that Th1 activity induces MDSC accumulation. CONCLUSION: Th1 liver inflammation mobilizes an MDSC response that, through the production of NO, can inhibit T cell proliferation. We propose that MDSCs serve an important negative feedback function in liver immune homeostasis, and that insufficient or inappropriate activity of this cell population may contribute to inflammatory liver pathology.


Subject(s)
CD11b Antigen/physiology , Hepatitis/immunology , Interferon-gamma/physiology , Transforming Growth Factor beta1/physiology , Animals , CD4-Positive T-Lymphocytes/immunology , Cell Communication , Cell Proliferation/drug effects , Liver/immunology , Mice , Mice, Knockout , Myeloid Cells/immunology , Myeloid Progenitor Cells/physiology , Nitric Oxide/physiology , Receptors, Chemokine/biosynthesis , Th1 Cells/immunology
17.
FEBS Open Bio ; 11(3): 705-713, 2021 03.
Article in English | MEDLINE | ID: mdl-33480189

ABSTRACT

Combinations of human lysozyme (hLYS) and antimicrobial peptides (AMPs) are known to exhibit either additive or synergistic activity, and as a result, they have therapeutic potential for persistent and antibiotic-resistant infections. We examined hLYS activity against Pseudomonas aeruginosa when combined with six different AMPs. In contrast to prior reports, we discovered that some therapeutically relevant AMPs manifest striking antagonistic interactions with hLYS across particular concentration ranges. We further found that the synthetic AMP Tet009 can inhibit hLYS-mediated bacterial lysis. To the best of our knowledge, these results represent the first observations of antagonism between hLYS and AMPs, and they advise that future development of lytic enzyme and AMP combination therapies considers the potential for antagonistic interactions.


Subject(s)
Antimicrobial Peptides/pharmacology , Muramidase/adverse effects , Pseudomonas aeruginosa/growth & development , Antimicrobial Peptides/chemistry , Bacteriolysis/drug effects , Drug Antagonism , Humans , Pseudomonas aeruginosa/drug effects
18.
Sci Rep ; 7: 41120, 2017 01 25.
Article in English | MEDLINE | ID: mdl-28120936

ABSTRACT

Integration of exogenous DNA into a host genome represents an important route to generate animal and cellular models for exploration into human disease and therapeutic development. In most models, little is known concerning structural integrity of the transgene, precise site of integration, or its impact on the host genome. We previously used whole-genome and targeted sequencing approaches to reconstruct transgene structure and integration sites in models of Huntington's disease, revealing complex structural rearrangements that can result from transgenesis. Here, we demonstrate in the R6/2 mouse, a widely used Huntington's disease model, that integration of a rearranged transgene with coincident deletion of 5,444 bp of host genome within the gene Gm12695 has striking molecular consequences. Gm12695, the function of which is unknown, is normally expressed at negligible levels in mouse brain, but transgene integration has resulted in cortical expression of a partial fragment (exons 8-11) 3' to the transgene integration site in R6/2. This transcript shows significant expression among the extensive network of differentially expressed genes associated with this model, including synaptic transmission, cell signalling and transcription. These data illustrate the value of sequence-level resolution of transgene insertions and transcription analysis to inform phenotypic characterization of transgenic models utilized in therapeutic research.


Subject(s)
Disease Models, Animal , Huntington Disease/pathology , Mice, Transgenic , Animals , Gene Expression Profiling , Gene Rearrangement , Humans , Mice , Sequence Analysis, DNA , Sequence Deletion
19.
Nat Genet ; 49(1): 36-45, 2017 01.
Article in English | MEDLINE | ID: mdl-27841880

ABSTRACT

Despite the clinical significance of balanced chromosomal abnormalities (BCAs), their characterization has largely been restricted to cytogenetic resolution. We explored the landscape of BCAs at nucleotide resolution in 273 subjects with a spectrum of congenital anomalies. Whole-genome sequencing revised 93% of karyotypes and demonstrated complexity that was cryptic to karyotyping in 21% of BCAs, highlighting the limitations of conventional cytogenetic approaches. At least 33.9% of BCAs resulted in gene disruption that likely contributed to the developmental phenotype, 5.2% were associated with pathogenic genomic imbalances, and 7.3% disrupted topologically associated domains (TADs) encompassing known syndromic loci. Remarkably, BCA breakpoints in eight subjects altered a single TAD encompassing MEF2C, a known driver of 5q14.3 microdeletion syndrome, resulting in decreased MEF2C expression. We propose that sequence-level resolution dramatically improves prediction of clinical outcomes for balanced rearrangements and provides insight into new pathogenic mechanisms, such as altered regulation due to changes in chromosome topology.


Subject(s)
Chromosome Aberrations , Congenital Abnormalities/genetics , Gene Rearrangement , Genetic Markers/genetics , Genetic Predisposition to Disease , Genome-Wide Association Study , Female , Humans , Male
20.
Nat Neurosci ; 19(3): 517-22, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26829649

ABSTRACT

Recurrent, reciprocal genomic disorders resulting from non-allelic homologous recombination (NAHR) between near-identical segmental duplications (SDs) are a major cause of human disease, often producing phenotypically distinct syndromes. The genomic architecture of flanking SDs presents a challenge for modeling these syndromes; however, the capability to efficiently generate reciprocal copy number variants (CNVs) that mimic NAHR would represent a valuable modeling tool. We describe here a CRISPR/Cas9 genome engineering method, single-guide CRISPR/Cas targeting of repetitive elements (SCORE), to model reciprocal genomic disorders and demonstrate its capabilities by generating reciprocal CNVs of 16p11.2 and 15q13.3, including alteration of one copy-equivalent of the SDs that mediate NAHR in vivo. The method is reproducible, and RNA sequencing reliably clusters transcriptional signatures from human subjects with in vivo CNVs and their corresponding in vitro models. This new approach will provide broad applicability for the study of genomic disorders and, with further development, may also permit efficient correction of these defects.


Subject(s)
Autistic Disorder/genetics , CRISPR-Cas Systems/genetics , Chromosome Disorders/genetics , DNA Copy Number Variations/genetics , Genetic Engineering/methods , Intellectual Disability/genetics , Segmental Duplications, Genomic/genetics , Seizures/genetics , Sequence Deletion/genetics , Chromosome Deletion , Chromosomes, Human, Pair 15/genetics , Chromosomes, Human, Pair 16/genetics , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Humans
SELECTION OF CITATIONS
SEARCH DETAIL