Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
Clin Genet ; 105(2): 214-219, 2024 02.
Artigo em Inglês | MEDLINE | ID: mdl-37899549

RESUMO

Critical genes involved in embryonic development are often transcription factors, regulating many downstream genes. LMX1B is a homeobox gene that is involved in formation of the limbs, eyes and kidneys, heterozygous loss-of-function sequence variants and deletions cause Nail-Patella syndrome. Most of the reported variants are localised within the gene's coding sequence, however, approximately 5%-10% of affected individuals do not have a pathogenic variant identified within this region. In this study, we present a family with four affected individuals across two generations with a deletion spanning a conserved upstream LMX1B-binding sequence. This deletion is de novo in the mother of three affected children. Furthermore, in this family, the manifestations appear limited to the nails and limbs, and therefore may reflect an attenuated phenotype of the classic Nail-Patella phenotype that includes ophthalmological and renal manifestations.


Assuntos
Genes Homeobox , Unhas , Criança , Humanos , Proteínas de Homeodomínio/genética , Mutação , Patela , Fenótipo , Fatores de Transcrição/genética
2.
Nucleic Acids Res ; 50(8): 4500-4514, 2022 05 06.
Artigo em Inglês | MEDLINE | ID: mdl-35451487

RESUMO

Histone H3.3 is an H3 variant which differs from the canonical H3.1/2 at four residues, including a serine residue at position 31 which is evolutionarily conserved. The H3.3 S31 residue is phosphorylated (H3.3 S31Ph) at heterochromatin regions including telomeres and pericentric repeats. However, the role of H3.3 S31Ph in these regions remains unknown. In this study, we find that H3.3 S31Ph regulates heterochromatin accessibility at telomeres during replication through regulation of H3K9/K36 histone demethylase KDM4B. In mouse embryonic stem (ES) cells, substitution of S31 with an alanine residue (H3.3 A31 -phosphorylation null mutant) results in increased KDM4B activity that removes H3K9me3 from telomeres. In contrast, substitution with a glutamic acid (H3.3 E31, mimics S31 phosphorylation) inhibits KDM4B, leading to increased H3K9me3 and DNA damage at telomeres. H3.3 E31 expression also increases damage at other heterochromatin regions including the pericentric heterochromatin and Y chromosome-specific satellite DNA repeats. We propose that H3.3 S31Ph regulation of KDM4B is required to control heterochromatin accessibility of repetitive DNA and preserve chromatin integrity.


Assuntos
Heterocromatina , Histonas , Animais , Camundongos , Histonas/genética , Histonas/metabolismo , Heterocromatina/genética , Histona Desmetilases/metabolismo , Fosforilação , Montagem e Desmontagem da Cromatina
3.
Proc Natl Acad Sci U S A ; 117(22): 12131-12142, 2020 06 02.
Artigo em Inglês | MEDLINE | ID: mdl-32414923

RESUMO

Topoisomerase IIα (TOP2A) is a core component of mitotic chromosomes and important for establishing mitotic chromosome condensation. The primary roles of TOP2A in mitosis have been difficult to decipher due to its multiple functions across the cell cycle. To more precisely understand the role of TOP2A in mitosis, we used the auxin-inducible degron (AID) system to rapidly degrade the protein at different stages of the human cell cycle. Removal of TOP2A prior to mitosis does not affect prophase timing or the initiation of chromosome condensation. Instead, it prevents chromatin condensation in prometaphase, extends the length of prometaphase, and ultimately causes cells to exit mitosis without chromosome segregation occurring. Surprisingly, we find that removal of TOP2A from cells arrested in prometaphase or metaphase cause dramatic loss of compacted mitotic chromosome structure and conclude that TOP2A is crucial for maintenance of mitotic chromosomes. Treatments with drugs used to poison/inhibit TOP2A function, such as etoposide and ICRF-193, do not phenocopy the effects on chromosome structure of TOP2A degradation by AID. Our data point to a role for TOP2A as a structural chromosome maintenance enzyme locking in condensation states once sufficient compaction is achieved.


Assuntos
Estruturas Cromossômicas/química , Cromossomos Humanos/química , DNA Topoisomerases Tipo II/metabolismo , Heterocromatina/química , Mitose , Segregação de Cromossomos , Estruturas Cromossômicas/genética , Cromossomos Humanos/genética , Citocinese , DNA Topoisomerases Tipo II/genética , Células HCT116 , Heterocromatina/genética , Humanos , Metáfase
4.
Proc Natl Acad Sci U S A ; 115(18): 4737-4742, 2018 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-29669917

RESUMO

ATRX (alpha thalassemia/mental retardation X-linked) complexes with DAXX to deposit histone variant H3.3 into repetitive heterochromatin. Recent genome sequencing studies in cancers have revealed mutations in ATRX and their association with ALT (alternative lengthening of telomeres) activation. Here we report depletion of ATRX in mouse ES cells leads to selective loss in ribosomal RNA gene (rDNA) copy number. Supporting this, ATRX-mutated human ALT-positive tumors also show a substantially lower rDNA copy than ALT-negative tumors. Further investigation shows that the rDNA copy loss and repeat instability are caused by a disruption in H3.3 deposition and thus a failure in heterochromatin formation at rDNA repeats in the absence of ATRX. We also find that ATRX-depleted cells are reduced in ribosomal RNA transcription output and show increased sensitivity to RNA polymerase I (Pol I) transcription inhibitor CX5461. In addition, human ALT-positive cancer cell lines are also more sensitive to CX5461 treatment. Our study provides insights into the contribution of ATRX loss of function to tumorigenesis through the loss of rDNA stability and suggests the therapeutic potential of targeting Pol I transcription in ALT cancers.


Assuntos
DNA de Neoplasias/metabolismo , DNA Ribossômico/metabolismo , Dosagem de Genes , Mutação , Proteínas de Neoplasias/metabolismo , Neoplasias/metabolismo , Proteína Nuclear Ligada ao X/metabolismo , Benzotiazóis/farmacologia , Linhagem Celular Tumoral , DNA de Neoplasias/genética , DNA Ribossômico/genética , Instabilidade Genômica , Humanos , Naftiridinas/farmacologia , Proteínas de Neoplasias/genética , Neoplasias/genética , Neoplasias/patologia , RNA Polimerase I/antagonistas & inibidores , RNA Polimerase I/genética , RNA Polimerase I/metabolismo , Transcrição Gênica/efeitos dos fármacos , Transcrição Gênica/genética , Proteína Nuclear Ligada ao X/genética
5.
PLoS Genet ; 12(12): e1006483, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27977684

RESUMO

Bloom syndrome is a recessive human genetic disorder with features of genome instability, growth deficiency and predisposition to cancer. The only known causative gene is the BLM helicase that is a member of a protein complex along with topoisomerase III alpha, RMI1 and 2, which maintains replication fork stability and dissolves double Holliday junctions to prevent genome instability. Here we report the identification of a second gene, RMI2, that is deleted in affected siblings with Bloom-like features. Cells from homozygous individuals exhibit elevated rates of sister chromatid exchange, anaphase DNA bridges and micronuclei. Similar genome and chromosome instability phenotypes are observed in independently derived RMI2 knockout cells. In both patient and knockout cell lines reduced localisation of BLM to ultra fine DNA bridges and FANCD2 at foci linking bridges are observed. Overall, loss of RMI2 produces a partially active BLM complex with mild features of Bloom syndrome.


Assuntos
Síndrome de Bloom/genética , Proteínas de Ligação a DNA/genética , Proteína do Grupo de Complementação D2 da Anemia de Fanconi/genética , Neoplasias/genética , Proteínas Nucleares/genética , Síndrome de Bloom/complicações , Síndrome de Bloom/patologia , Instabilidade Cromossômica/genética , DNA Helicases/genética , DNA Cruciforme/genética , Predisposição Genética para Doença , Instabilidade Genômica , Humanos , Complexos Multiproteicos/genética , Neoplasias/complicações , Neoplasias/patologia , Troca de Cromátide Irmã/genética
6.
Am J Respir Cell Mol Biol ; 59(3): 375-382, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29481304

RESUMO

Primary ciliary dyskinesia is an inherited, currently incurable condition. In the respiratory system, primary ciliary dyskinesia causes impaired functioning of the mucociliary escalator, leading to nasal congestion, cough, and recurrent otitis media, and commonly progresses to cause more serious and permanent damage, including hearing deficits, chronic sinusitis, and bronchiectasis. New treatment options for the condition are thus necessary. In characterizing an immortalized human bronchial epithelial cell line (BCi-NS1.1) grown at an air-liquid interface to permit differentiation, we have identified that these cells have dyskinetic motile cilia. The cells had a normal male karyotype, and phenotypic markers of epithelial cell differentiation emerged, as previously shown. Ciliary beat frequency (CBF) as assessed by high-speed videomicroscopy was lower than normal (4.4 Hz). Although changes in CBF induced by known modulators were as expected, the cilia displayed a dyskinetic, circular beat pattern characteristic of central microtubular agenesis with outer doublet transposition. This ultrastructural defect was confirmed by electron microscopy. We propose that the BCi-NS1.1 cell line is a useful model system for examination of modulators of CBF and more specifically could be used to screen for novel drugs with the ability to enhance CBF and perhaps repair a dyskinetic ciliary beat pattern.


Assuntos
Diferenciação Celular/fisiologia , Cílios/patologia , Transtornos da Motilidade Ciliar/patologia , Discinesias/patologia , Células Epiteliais/citologia , Linhagem Celular , Células Cultivadas , Humanos
7.
Chromosome Res ; 25(1): 61-76, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-28181049

RESUMO

A fundamental requirement in nature is for a cell to correctly package and divide its replicated genome. Condensin is a mechanical multisubunit complex critical to this process. Condensin uses ATP to power conformational changes in DNA to enable to correct DNA compaction, organization, and segregation of DNA from the simplest bacteria to humans. The highly conserved nature of the condensin complex and the structural similarities it shares with the related cohesin complex have provided important clues as to how it functions in cells. The fundamental requirement for condensin in mitosis and meiosis is well established, yet the precise mechanism of action is still an open question. Mutation or removal of condensin subunits across a range of species disrupts orderly chromosome condensation leading to errors in chromosome segregation and likely death of the cell. There are divergences in function across species for condensin. Once considered to function solely in mitosis and meiosis, an accumulating body of evidence suggests that condensin has key roles in also regulating the interphase genome. This review will examine how condensin organizes our genomes, explain where and how it binds the genome at a mechanical level, and highlight controversies and future directions as the complex continues to fascinate and baffle biologists.


Assuntos
Adenosina Trifosfatases/fisiologia , Proteínas de Ligação a DNA/fisiologia , Genoma/genética , Complexos Multiproteicos/fisiologia , Adenosina Trifosfatases/ultraestrutura , Animais , Segregação de Cromossomos , Proteínas de Ligação a DNA/ultraestrutura , Humanos , Interfase , Meiose , Mitose , Complexos Multiproteicos/ultraestrutura
8.
PLoS Genet ; 11(2): e1004964, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25675407

RESUMO

Histones package DNA and regulate epigenetic states. For the latter, probably the most important histone is H3. Mammals have three near-identical H3 isoforms: canonical H3.1 and H3.2, and the replication-independent variant H3.3. This variant can accumulate in slowly dividing somatic cells, replacing canonical H3. Some replication-independent histones, through their ability to incorporate outside S-phase, are functionally important in the very slowly dividing mammalian germ line. Much remains to be learned of H3.3 functions in germ cell development. Histone H3.3 presents a unique genetic paradigm in that two conventional intron-containing genes encode the identical protein. Here, we present a comprehensive analysis of the developmental effects of null mutations in each of these genes. H3f3a mutants were viable to adulthood. Females were fertile, while males were subfertile with dysmorphic spermatozoa. H3f3b mutants were growth-deficient, dying at birth. H3f3b heterozygotes were also growth-deficient, with males being sterile because of arrest of round spermatids. This sterility was not accompanied by abnormalities in sex chromosome inactivation in meiosis I. Conditional ablation of H3f3b at the beginning of folliculogenesis resulted in zygote cleavage failure, establishing H3f3b as a maternal-effect gene, and revealing a requirement for H3.3 in the first mitosis. Simultaneous ablation of H3f3a and H3f3b in folliculogenesis resulted in early primary oocyte death, demonstrating a crucial role for H3.3 in oogenesis. These findings reveal a heavy reliance on H3.3 for growth, gametogenesis, and fertilization, identifying developmental processes that are particularly susceptible to H3.3 deficiency. They also reveal partial redundancy in function of H3f3a and H3f3b, with the latter gene being generally the most important.


Assuntos
Sobrevivência Celular/genética , Cromatina/genética , Fertilidade/genética , Histonas/genética , Oogênese , Animais , Replicação do DNA/genética , Feminino , Feto , Masculino , Meiose/genética , Camundongos , Oócitos/crescimento & desenvolvimento , Espermatócitos/crescimento & desenvolvimento , Espermatócitos/patologia , Espermatozoides/crescimento & desenvolvimento , Espermatozoides/patologia , Zigoto
9.
Prog Mol Subcell Biol ; 56: 541-554, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28840252

RESUMO

The accurate segregation of chromosomes to daughter cells is essential for healthy development to occur. Imbalances in chromosome number have long been associated with cancers amongst other medical disorders. Little is known whether abnormal chromosome numbers are an early contributor to the cancer progression pathway. Centromere DNA and protein defects are known to impact on the fidelity of chromosome segregation in cell and model systems. In this chapter we discuss recent developments in understanding the contribution of centromere abnormalities at the protein and DNA level and their role in cancer in human and mouse systems.


Assuntos
Centrômero/genética , Centrômero/patologia , Segregação de Cromossomos , Proteínas de Ligação a DNA/metabolismo , DNA/metabolismo , Neoplasias/genética , Neoplasias/patologia , Animais , Centrômero/metabolismo , Humanos , Neoplasias/metabolismo
10.
Chromosome Res ; 24(2): 243-69, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-27008552

RESUMO

Condensin is an integral component of the mitotic chromosome condensation machinery, which ensures orderly segregation of chromosomes during cell division. In metazoans, condensin exists as two complexes, condensin I and II. It is not yet clear what roles these complexes may play outside mitosis, and so we have examined their behaviour both in normal interphase and in premature chromosome condensation (PCC). We find that a small fraction of condensin I is retained in interphase nuclei, and our data suggests that this interphase nuclear condensin I is active in both gene regulation and chromosome condensation. Furthermore, live cell imaging demonstrates condensin II dramatically increases on G1 nuclei following completion of mitosis. Our PCC studies show condensins I and II and topoisomerase II localise to the chromosome axis in G1-PCC and G2/M-PCC, while KIF4 binding is altered. Individually, condensins I and II are dispensable for PCC. However, when both are knocked out, G1-PCC chromatids are less well structured. Our results define new roles for the condensins during interphase and provide new information about the mechanism of PCC.


Assuntos
Adenosina Trifosfatases/metabolismo , Segregação de Cromossomos/fisiologia , Cromossomos/metabolismo , DNA Topoisomerases Tipo II/metabolismo , Proteínas de Ligação a DNA/metabolismo , Interfase/fisiologia , Complexos Multiproteicos/metabolismo , Animais , Linhagem Celular , Núcleo Celular/metabolismo , Galinhas , Proteínas Cromossômicas não Histona/genética , Regulação da Expressão Gênica/genética , Técnicas de Inativação de Genes , Imageamento Tridimensional/métodos , Hibridização in Situ Fluorescente/métodos , Mitose/fisiologia , Mapeamento Físico do Cromossomo , Regiões Promotoras Genéticas
11.
J Biol Chem ; 290(10): 6156-67, 2015 Mar 06.
Artigo em Inglês | MEDLINE | ID: mdl-25605712

RESUMO

The condensin complex plays a key role in organizing mitotic chromosomes. In vertebrates, there are two condensin complexes that have independent and cooperative roles in folding mitotic chromosomes. In this study, we dissect the role of a putative Cdk1 site on the condensin II subunit CAP-D3 in chicken DT40 cells. This conserved site has been shown to activate condensin II during prophase in human cells, and facilitate further phosphorylation by polo-like kinase I. We examined the functional significance of this phosphorylation mark by mutating the orthologous site of CAP-D3 (CAP-D3(T1403A)) in chicken DT40 cells. We show that this mutation is a gain of function mutant in chicken cells; it disrupts prophase, results in a dramatic shortening of the mitotic chromosome axis, and leads to abnormal INCENP localization. Our results imply phosphorylation of CAP-D3 acts to limit condensin II binding onto mitotic chromosomes. We present the first in vivo example that alters the ratio of condensin I:II on mitotic chromosomes. Our results demonstrate this ratio is a critical determinant in shaping mitotic chromosomes.


Assuntos
Adenosina Trifosfatases/genética , Cromatina/ultraestrutura , Cromossomos/genética , Proteínas de Ligação a DNA/genética , Mitose/genética , Complexos Multiproteicos/genética , Adenosina Trifosfatases/química , Adenosina Trifosfatases/ultraestrutura , Animais , Proteína Quinase CDC2/genética , Galinhas , Cromatina/genética , Cromossomos/ultraestrutura , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/ultraestrutura , Células HeLa , Humanos , Complexos Multiproteicos/química , Complexos Multiproteicos/ultraestrutura , Mutação , Fosforilação , Treonina/química , Treonina/genética
12.
PLoS Genet ; 8(9): e1002919, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22969435

RESUMO

Reduced DNA methylation has been reported in DICER1-deficient mouse ES cells. Reductions seen at pericentric satellite repeats have suggested that siRNAs are required for the proper assembly of heterochromatin. More recent studies have postulated that the reduced methylation is an indirect effect: the loss of Mir290 cluster miRNAs leads to upregulation of the transcriptional repressor RBL2 that targets the downregulation of DNA methyltransferase (Dnmt) genes. However, the observations have been inconsistent. We surmised that the inconsistency could be related to cell line "age," given that DNA methylation is lost progressively with passage in DNMT-deficient ES cells. We therefore subjected Dicer1(-/-) ES cells to two experimental regimes to rigorously test the level of functional DNMT activity. First, we cultured them for a prolonged period. If DNMT activity was reduced, further losses of methylation would occur. Second, we measured their DNMT activity in a rebound DNA methylation assay: DNA methylation was stripped from Cre/loxP conditionally mutant Dicer1 ES cells using a shRNA targeting Dnmt1 mRNA. Cre expression then converted these cells to Dicer1(-/-), allowing for DNMT1 recovery and forcing the cells to remethylate in the absence of RNAi. In both cases, we found functional DNMT activity to be normal. Finally, we also show that the level of RBL2 protein is not at excess levels in Dicer1(-/-) ES cells as has been assumed. These studies reveal that reduced functional DNMT activity is not a salient feature of DICER1-deficient ES cells. We suggest that the reduced DNA methylation sometimes observed in these cells could be due to stochastic alterations in DNA methylation patterns that could offer growth or survival advantages in culture, or to the dysregulation of pathways acting in opposition to the DNMT pathway.


Assuntos
RNA Helicases DEAD-box/genética , Metilação de DNA , Células-Tronco Embrionárias/metabolismo , Ribonuclease III/genética , Animais , RNA Helicases DEAD-box/metabolismo , DNA (Citosina-5-)-Metiltransferase 1 , DNA (Citosina-5-)-Metiltransferases/metabolismo , Camundongos , MicroRNAs/metabolismo , Proteína p130 Retinoblastoma-Like/metabolismo , Ribonuclease III/metabolismo
13.
J Cell Sci ; 125(Pt 6): 1591-604, 2012 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-22344259

RESUMO

In vertebrates, two condensin complexes exist, condensin I and condensin II, which have differing but unresolved roles in organizing mitotic chromosomes. To dissect accurately the role of each complex in mitosis, we have made and studied the first vertebrate conditional knockouts of the genes encoding condensin I subunit CAP-H and condensin II subunit CAP-D3 in chicken DT40 cells. Live-cell imaging reveals highly distinct segregation defects. CAP-D3 (condensin II) knockout results in masses of chromatin-containing anaphase bridges. CAP-H (condensin I)-knockout anaphases have a more subtle defect, with chromatids showing fine chromatin fibres that are associated with failure of cytokinesis and cell death. Super-resolution microscopy reveals that condensin-I-depleted mitotic chromosomes are wider and shorter, with a diffuse chromosome scaffold, whereas condensin-II-depleted chromosomes retain a more defined scaffold, with chromosomes more stretched and seemingly lacking in axial rigidity. We conclude that condensin II is required primarily to provide rigidity by establishing an initial chromosome axis around which condensin I can arrange loops of chromatin.


Assuntos
Adenosina Trifosfatases/fisiologia , Cromossomos/genética , Proteínas de Ligação a DNA/fisiologia , Mitose/genética , Complexos Multiproteicos/fisiologia , Adenosina Trifosfatases/deficiência , Adenosina Trifosfatases/genética , Animais , Linhagem Celular Tumoral , Galinhas , Proteínas de Ligação a DNA/deficiência , Proteínas de Ligação a DNA/genética , Técnicas de Inativação de Genes/métodos , Complexos Multiproteicos/deficiência , Complexos Multiproteicos/genética
14.
Chromosoma ; 121(4): 327-40, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22527114

RESUMO

The centromere is a chromosomal structure that is essential for the accurate segregation of replicated eukaryotic chromosomes to daughter cells. In most centromeres, the underlying DNA is principally made up of repetitive DNA elements, such as tandemly repeated satellite DNA and retrotransposable elements. Paradoxically, for such an essential genomic region, the DNA is rapidly evolving both within and between species. In this review, we show that the centromere locus is a resilient structure that can undergo evolutionary cycles of birth, growth, maturity, death and resurrection. The birth phase is highlighted by examples in humans and other organisms where centromere DNA deletions or chromosome rearrangements can trigger the epigenetic assembly of neocentromeres onto genomic sites without typical features of centromere DNA. In addition, functional centromeres can be generated in the laboratory using various methodologies. Recent mapping of the foundation centromere mark, the histone H3 variant CENP-A, onto near-complete genomes has uncovered examples of new centromeres which have not accumulated centromere repeat DNA. During the growth period of the centromere, repeat DNA begins to appear at some, but not all, loci. The maturity stage is characterised by centromere repeat accumulation, expansions and contractions and the rapid evolution of the centromere DNA between chromosomes of the same species and between species. This stage provides inherent centromere stability, facilitated by repression of gene activity and meiotic recombination at and around the centromeres. Death to a centromere can result from genomic instability precipitating rearrangements, deletions, accumulation of mutations and the loss of essential centromere binding proteins. Surprisingly, ancestral centromeres can undergo resurrection either in the field or in the laboratory, via as yet poorly understood mechanisms. The underlying principle for the preservation of a centromeric evolutionary life cycle is to provide resilience and perpetuity for the all-important structure and function of the centromere.


Assuntos
Evolução Biológica , Centrômero/genética , Sequências Repetitivas de Ácido Nucleico , Animais , Centrômero/química , Histonas/genética , Histonas/metabolismo , Humanos , Cariótipo
15.
Eur J Hum Genet ; 31(5): 521-525, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36446895

RESUMO

We aimed to determine whether SNP-microarray genomic testing of saliva had a greater diagnostic yield than blood for pathogenic copy number variants (CNVs). We selected patients who underwent CMA testing of both blood and saliva from 23,289 blood and 21,857 saliva samples. Our cohort comprised 370 individuals who had testing of both, 224 with syndromic intellectual disability (ID) and 146 with isolated ID. Mosaic pathogenic CNVs or aneuploidy were detected in saliva but not in blood in 20/370 (4.4%). All 20 individuals had syndromic ID, accounting for 9.1% of the syndromic ID sub-cohort. Pathogenic CNVs were large in size (median of 46 Mb), and terminal in nature, with median mosaicism of 27.5% (not exceeding 40%). By contrast, non-mosaic pathogenic CNVs were 100% concordant between blood and saliva, considerably smaller in size (median of 0.65 Mb), and predominantly interstitial in location. Given that salivary microarray testing has increased diagnostic utility over blood in individuals with syndromic ID, we recommend it as a first-tier testing in this group.


Assuntos
Deficiência Intelectual , Criança , Humanos , Deficiência Intelectual/diagnóstico , Deficiência Intelectual/genética , Saliva , Deficiências do Desenvolvimento/genética , Aberrações Cromossômicas , Mosaicismo , Genômica , Variações do Número de Cópias de DNA
16.
Genome Res ; 19(12): 2202-13, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19737860

RESUMO

The Y centromere sequence of house mouse, Mus musculus, remains unknown despite our otherwise significant knowledge of the genome sequence of this important mammalian model organism. Here, we report the complete molecular characterization of the C57BL/6J chromosome Y centromere, which comprises a highly diverged minor satellite-like sequence (designated Ymin) with higher-order repeat (HOR) sequence organization previously undescribed at mouse centromeres. The Ymin array is approximately 90 kb in length and resides within a single BAC clone that provides sequence information spanning an endogenous animal centromere for the first time. By exploiting direct patrilineal inheritance of the Y chromosome, we demonstrate stability of the Y centromere DNA structure spanning at least 175 inbred generations to beyond the time of domestication of the East Asian M.m. molossinus "fancy" mouse through which the Y chromosome was first introduced into the classical inbred laboratory mouse strains. Despite this stability, at least three unequal genetic exchange events have altered Ymin HOR unit length and sequence structure since divergence of the ancestral Mus musculus subspecies around 900,000 yr ago, with major turnover of the HOR arrays driving rapid divergence of sequence and higher-order structure at the mouse Y centromere. A comparative sequence analysis between the human and chimpanzee centromeres indicates a similar rapid divergence of the primate Y centromere. Our data point to a unique DNA sequence and organizational architecture for the mouse Y centromere that has evolved independently of all other mouse centromeres.


Assuntos
Sequência de Bases , Evolução Biológica , Centrômero/genética , DNA Satélite/genética , Cromossomo Y/genética , Animais , Cromossomos Artificiais Bacterianos , Feminino , Biblioteca Gênica , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Pan troglodytes , Sequências Repetitivas de Ácido Nucleico , Análise de Sequência de DNA , Especificidade da Espécie , Fatores de Tempo
17.
BMC Biochem ; 11: 50, 2010 Dec 31.
Artigo em Inglês | MEDLINE | ID: mdl-21194474

RESUMO

BACKGROUND: Cell biologists face the need to rapidly analyse their proteins of interest in order to gain insight into their function. Often protein purification, cellular localisation and Western blot analyses can be multi-step processes, where protein is lost, activity is destroyed or effective antibodies have not yet been generated. AIM: To develop a method that simplifies the critical protein analytical steps of the laboratory researcher, leading to easy, efficient and rapid protein purification, cellular localisation and quantification. RESULTS: We have tagged the SMC2 subunit of the condensin complex with the Streptavidin-Binding Peptide (SBP), optimising and demonstrating the efficacious use of this tag for performing these protein analytical steps. Based on silver staining, and Western analysis, SBP delivered an outstanding specificity and purity of the condensin complex. We also developed a rapid and highly specific procedure to localise SBP-tagged proteins in cells in a single step procedure thus bypassing the need for using antibodies. Furthermore we have shown that the SBP tag can be used for isolating tagged proteins from chemically cross-linked cell populations for capturing DNA-protein interactions. CONCLUSIONS: The small 38-amino acid synthetic SBP offers the potential to successfully perform all four critical analytical procedures as a single step and should have a general utility for the study of many proteins and protein complexes.


Assuntos
Adenosina Trifosfatases/metabolismo , Proteínas de Transporte/metabolismo , Proteínas de Ligação a DNA/metabolismo , Complexos Multiproteicos/metabolismo , Proteínas Nucleares/metabolismo , Adenosina Trifosfatases/química , Adenosina Trifosfatases/isolamento & purificação , Animais , Western Blotting , Proteínas de Transporte/química , Proteínas de Transporte/genética , Proteínas de Ciclo Celular , Linhagem Celular Tumoral , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/isolamento & purificação , Fluorescência , Humanos , Microscopia de Fluorescência , Complexos Multiproteicos/química , Complexos Multiproteicos/isolamento & purificação , Mutação , Proteínas Nucleares/química , Proteínas Nucleares/genética , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Reprodutibilidade dos Testes , Estreptavidina/química , Estreptavidina/metabolismo
18.
BMC Genomics ; 10: 498, 2009 Oct 27.
Artigo em Inglês | MEDLINE | ID: mdl-19860919

RESUMO

BACKGROUND: Many mammalian genes are arranged in a bidirectional manner, sharing a common promoter and regulatory elements. This is especially true for promoters containing a CpG island, usually unmethylated and associated with an 'open' or accessible chromatin structure. In evolutionary terms, a primary function of genomic methylation is postulated to entail protection of the host genome from the disruption associated with activity of parasitic or transposable elements. These are usually epigenetically silenced following insertion into mammalian genomes, becoming sequence degenerate over time. Despite this, it is clear that many transposable element-derived DNAs have evaded host-mediated epigenetic silencing to remain expressed (domesticated) in mammalian genomes, several of which have demonstrated essential roles during mammalian development. RESULTS: The current study provides evidence that many CpG island-associated promoters associated with single genes exhibit inherent bidirectionality, facilitating "hijack" by transposable elements to create novel antisense 'head-to-head' bidirectional gene pairs in the genome that facilitates escape from host-mediated epigenetic silencing. This is often associated with an increase in CpG island length and transcriptional activity in the antisense direction. From a list of over 60 predicted protein-coding genes derived from transposable elements in the human genome and 40 in the mouse, we have found that a significant proportion are orientated in a bidirectional manner with CpG associated regulatory regions. CONCLUSION: These data strongly suggest that the selective force that shields endogenous CpG-containing promoter from epigenetic silencing can extend to exogenous foreign DNA elements inserted in close proximity in the antisense orientation, with resulting transcription and maintenance of sequence integrity of such elements in the host genome. Over time, this may result in "domestication" of such elements to provide novel cellular and developmental functions.


Assuntos
Ilhas de CpG/genética , Genoma , Regiões Promotoras Genéticas/genética , Sequências Repetitivas de Ácido Nucleico/genética , Transcrição Gênica , Animais , Cromatina/genética , Elementos de DNA Transponíveis/genética , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Humanos , Camundongos , Ratos , Retroviridae/genética
19.
Trends Cell Biol ; 14(7): 359-68, 2004 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15246429

RESUMO

At each mitosis, accurate segregation of every chromosome is ensured by the assembly of a kinetochore at each centromeric locus. Six foundation kinetochore proteins that assemble hierarchically and co-dependently have been identified in vertebrates. CENP-A, Mis12, CENP-C, CENP-H and CENP-I localize to a core domain of centromeric chromatin. The sixth protein, CENP-B, although not essential in higher eukaryotes, has homologues in fission yeast that bind pericentric DNA and are essential for heterochromatin formation. Foundation kinetochore proteins have various roles and mutual interactions, and their associations with centromeric DNA and heterochromatin create structural domains that support the different functions of the centromere. Advances in molecular and microscopic techniques, coupled with rare centromere variants, have enabled us to gain fresh insights into the linear and 3D organization of centromeric chromatin.


Assuntos
Centrômero/fisiologia , Proteínas/fisiologia , Animais , Centrômero/metabolismo , Humanos , Cinetocoros/fisiologia , Modelos Biológicos , Proteínas/metabolismo , Leveduras , Zea mays
20.
Open Biol ; 9(12): 190222, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31795919

RESUMO

Topoisomerase III beta (TOP3B) is one of the least understood members of the topoisomerase family of proteins and remains enigmatic. Our recent data shed light on the function and relevance of TOP3B to disease. A homozygous deletion for the TOP3B gene was identified in a patient with bilateral renal cancer. Analyses in both patient and modelled human cells show the disruption of TOP3B causes genome instability with a rise in DNA damage and chromosome bridging (mis-segregation). The primary molecular defect underlying this pathology is a significant increase in R-loop formation. Our data show that TOP3B is necessary to prevent the accumulation of excessive R-loops and identify TOP3B as a putative cancer gene, and support recent data showing that R-loops are involved in cancer aetiology.


Assuntos
DNA Topoisomerases Tipo I/deficiência , Instabilidade Genômica , Estruturas R-Loop , Linhagem Celular Tumoral , Dano ao DNA , Homozigoto , Humanos , Deleção de Sequência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA