Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Cell ; 166(4): 950-962, 2016 Aug 11.
Artigo em Inglês | MEDLINE | ID: mdl-27518565

RESUMO

Posttranslational modifications (PTMs) of tubulin specify microtubules for specialized cellular functions and comprise what is termed a "tubulin code." PTMs of histones comprise an analogous "histone code," although the "readers, writers, and erasers" of the cytoskeleton and epigenome have heretofore been distinct. We show that methylation is a PTM of dynamic microtubules and that the histone methyltransferase SET-domain-containing 2 (SETD2), which is responsible for H3 lysine 36 trimethylation (H3K36me3) of histones, also methylates α-tubulin at lysine 40, the same lysine that is marked by acetylation on microtubules. Methylation of microtubules occurs during mitosis and cytokinesis and can be ablated by SETD2 deletion, which causes mitotic spindle and cytokinesis defects, micronuclei, and polyploidy. These data now identify SETD2 as a dual-function methyltransferase for both chromatin and the cytoskeleton and show a requirement for methylation in maintenance of genomic stability and the integrity of both the tubulin and histone codes.


Assuntos
Montagem e Desmontagem da Cromatina , Citoesqueleto/metabolismo , Código das Histonas , Histona-Lisina N-Metiltransferase/metabolismo , Linhagem Celular Tumoral , Citocinese , Instabilidade Genômica , Histona-Lisina N-Metiltransferase/genética , Histonas/metabolismo , Humanos , Lisina/metabolismo , Metilação , Microtúbulos/metabolismo , Mitose , Processamento de Proteína Pós-Traducional , Tubulina (Proteína)/metabolismo
2.
Proc Natl Acad Sci U S A ; 120(39): e2303752120, 2023 09 26.
Artigo em Inglês | MEDLINE | ID: mdl-37722039

RESUMO

Isochromosomes are mirror-imaged chromosomes with simultaneous duplication and deletion of genetic material which may contain two centromeres to create isodicentric chromosomes. Although isochromosomes commonly occur in cancer and developmental disorders and promote genome instability, mechanisms that prevent isochromosomes are not well understood. We show here that the tumor suppressor and methyltransferase SETD2 is essential to prevent these errors. Using cellular and cytogenetic approaches, we demonstrate that loss of SETD2 or its epigenetic mark, histone H3 lysine 36 trimethylation (H3K36me3), results in the formation of isochromosomes as well as isodicentric and acentric chromosomes. These defects arise during DNA replication and are likely due to faulty homologous recombination by RAD52. These data provide a mechanism for isochromosome generation and demonstrate that SETD2 and H3K36me3 are essential to prevent the formation of this common mutable chromatin structure known to initiate a cascade of genomic instability in cancer.


Assuntos
Isocromossomos , Humanos , Centrômero , Aberrações Cromossômicas , Citogenética , Replicação do DNA , Instabilidade Genômica
3.
Brain ; 144(8): 2527-2540, 2021 09 04.
Artigo em Inglês | MEDLINE | ID: mdl-34014281

RESUMO

Gene discovery efforts in autism spectrum disorder have identified heterozygous defects in chromatin remodeller genes, the 'readers, writers and erasers' of methyl marks on chromatin, as major contributors to this disease. Despite this advance, a convergent aetiology between these defects and aberrant chromatin architecture or gene expression has remained elusive. Recently, data have begun to emerge that chromatin remodellers also function directly on the cytoskeleton. Strongly associated with autism spectrum disorder, the SETD2 histone methyltransferase for example, has now been shown to directly methylate microtubules of the mitotic spindle. However, whether microtubule methylation occurs in post-mitotic cells, for example on the neuronal cytoskeleton, is not known. We found the SETD2 α-tubulin lysine 40 trimethyl mark occurs on microtubules in the brain and in primary neurons in culture, and that the SETD2 C-terminal SRI domain is required for binding and methylation of α-tubulin. A CRISPR knock-in of a pathogenic SRI domain mutation (Setd2SRI) that disables microtubule methylation revealed at least one wild-type allele was required in mice for survival, and while viable, heterozygous Setd2SRI/wtmice exhibited an anxiety-like phenotype. Finally, whereas RNA-sequencing (RNA-seq) and chromatin immunoprecipitation-sequencing (ChIP-seq) showed no concomitant changes in chromatin methylation or gene expression in Setd2SRI/wtmice, primary neurons exhibited structural deficits in axon length and dendritic arborization. These data provide the first demonstration that microtubules of neurons are methylated, and reveals a heterozygous chromatin remodeller defect that specifically disables microtubule methylation is sufficient to drive an autism-associated phenotype.


Assuntos
Ansiedade/metabolismo , Histona-Lisina N-Metiltransferase/metabolismo , Microtúbulos/metabolismo , Neurônios/metabolismo , Animais , Encéfalo/metabolismo , Histonas/metabolismo , Metilação , Camundongos , Fenótipo
4.
Biochem Soc Trans ; 49(1): 93-105, 2021 02 26.
Artigo em Inglês | MEDLINE | ID: mdl-33492339

RESUMO

Alterations in global epigenetic signatures on chromatin are well established to contribute to tumor initiation and progression. Chromatin methylation status modulates several key cellular processes that maintain the integrity of the genome. KDM4A, a demethylase that belongs to the Fe-II dependent dioxygenase family that uses α-ketoglutarate and molecular oxygen as cofactors, is overexpressed in several cancers and is associated with an overall poor prognosis. KDM4A demethylates lysine 9 (H3K9me2/3) and lysine 36 (H3K36me3) methyl marks on histone H3. Given the complexity that exists with these marks on chromatin and their effects on transcription and proliferation, it naturally follows that demethylation serves an equally important role in these cellular processes. In this review, we highlight the role of KDM4A in transcriptional modulation, either dependent or independent of its enzymatic activity, arising from the amplification of this demethylase in cancer. KDM4A modulates re-replication of distinct genomic loci, activates cell cycle inducers, and represses proteins involved in checkpoint control giving rise to proliferative damage, mitotic disturbances and chromosomal breaks, ultimately resulting in genomic instability. In parallel, emerging evidence of non-nuclear substrates of epigenetic modulators emphasize the need to investigate the role of KDM4A in regulating non-nuclear substrates and evaluate their contribution to genomic instability in this context. The existence of promising KDM-specific inhibitors makes these demethylases an attractive target for therapeutic intervention in cancers.


Assuntos
Instabilidade Genômica/genética , Histona Desmetilases com o Domínio Jumonji/fisiologia , Animais , Transformação Celular Neoplásica/genética , Histonas/metabolismo , Humanos , Metilação , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patologia , Processamento de Proteína Pós-Traducional/genética , Transdução de Sinais/genética
5.
J Cell Sci ; 131(24)2018 12 14.
Artigo em Inglês | MEDLINE | ID: mdl-30518623

RESUMO

Loss of the gene von Hippel-Lindau (VHL) is associated with loss of primary cilia and is causally linked to elevated levels of Aurora kinase A (AURKA). We developed an image-based high-throughput screening (HTS) assay using a dual-labeling image analysis strategy that identifies both the cilium and the basal body. By using this strategy, we screened small-molecule compounds for the targeted rescue of cilia defects associated with VHL deficiency with high accuracy and reproducibility. Bexarotene was identified and validated as a positive regulator of the primary cilium. Importantly, the inability of an alternative retinoid X receptor (RXR) agonist to rescue ciliogenesis, in contrast to bexarotene, suggested that multiple bexarotene-driven mechanisms were responsible for the rescue. We found that bexarotene decreased AURKA expression in VHL-deficient cells, thereby restoring the ability of these cells to ciliate in the absence of VHL Finally, bexarotene treatment reduced the propensity of subcutaneous lesions to develop into tumors in a mouse xenograft model of renal cell carcinoma (RCC), with a concomitant decrease in activated AURKA, highlighting the potential of bexarotene treatment as an intervention strategy in the clinic to manage renal cystogenesis associated with VHL deficiency and elevated AURKA expression.


Assuntos
Aurora Quinase A/metabolismo , Bexaroteno/farmacologia , Carcinoma de Células Renais/tratamento farmacológico , Aurora Quinase A/genética , Linhagem Celular Tumoral , Cílios/efeitos dos fármacos , Cílios/metabolismo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Mutação/efeitos dos fármacos , Mutação/genética , Proteína Supressora de Tumor Von Hippel-Lindau/efeitos dos fármacos , Proteína Supressora de Tumor Von Hippel-Lindau/metabolismo
6.
J Am Soc Nephrol ; 26(3): 553-64, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25313256

RESUMO

von Hippel-Lindau (VHL) gene mutations are associated with clear cell renal cell carcinoma (ccRCC). A hallmark of ccRCC is loss of the primary cilium. Loss of this key organelle in ccRCC is caused by loss of VHL and associated with increased Aurora kinase A (AURKA) and histone deacetylase 6 (HDAC6) activities, which drive disassembly of the primary cilium. However, the underlying mechanism by which VHL loss increases AURKA levels has not been clearly elucidated, although it has been suggested that hypoxia-inducible factor-1α (HIF-1α) mediates increased AURKA expression in VHL-null cells. By contrast, we found that elevated AURKA expression is not increased by HIF-1α, suggesting an alternate mechanism for AURKA dysregulation in VHL-null cells. We report here that AURKA expression is driven by ß-catenin transcription in VHL-null cells. In a panel of RCC cell lines, we observed nuclear accumulation of ß-catenin and increased AURKA signaling to HDAC6. Moreover, HIF-1α inhibited AURKA expression by inhibiting ß-catenin transcription. VHL knockdown activated ß-catenin and elevated AURKA expression, decreased primary cilia formation, and caused significant shortening of cilia length in cells that did form cilia. The ß-catenin responsive transcription inhibitor iCRT14 reduced AURKA levels and rescued ciliary defects, inducing a significant increase in primary cilia formation in VHL-deficient cells. These data define a role for ß-catenin in regulating AURKA and formation of primary cilia in the setting of VHL deficiency, opening new avenues for treatment with ß-catenin inhibitors to rescue ciliogenesis in ccRCC.


Assuntos
Aurora Quinase A/metabolismo , Carcinoma de Células Renais/metabolismo , Neoplasias Renais/metabolismo , Proteína Supressora de Tumor Von Hippel-Lindau/metabolismo , beta Catenina/metabolismo , Carcinoma de Células Renais/patologia , Linhagem Celular Tumoral , Cílios/patologia , Células Epiteliais/metabolismo , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Neoplasias Renais/patologia
7.
Proc Natl Acad Sci U S A ; 109(43): 17466-71, 2012 Oct 23.
Artigo em Inglês | MEDLINE | ID: mdl-23045645

RESUMO

Two Sentrin/small ubiquitin-like modifier (SUMO)-specific protease 7 (SENP7) variants are naturally expressed in breast epithelia. Breast cancer (BCa) onset down-regulates the short SENP7 splice variant (SENP7S) and enhances the long transcript (SENP7L). Here, we show that SENP7L induction promotes gene expression profiles that favor aberrant proliferation and initiate epithelial-mesenchymal transition (EMT). SENP7L exhibits an interaction domain for the epigenetic remodeler heterochromatin protein 1 α (HP1α) and isopeptidase activity against SUMO-modified HP1α. Loss of this interaction domain, as observed with SENP7S, favors HP1α SUMOylation. SUMOylated HP1α is enriched at E2F-responsive and mesenchymal gene promoters, silences transcription of these genes, and promotes cellular senescence. Elevated SENP7L renders HP1α hypo-SUMOylated, which relieves transcriptional repression of the same genes and concurrently decreases transcription of epithelial-promoting genes via an HP1α-independent mechanism. Consequently, SENP7L levels correlate with EMT, motility, and invasiveness of BCa cells. Stable knockdown of elevated SENP7L levels lessens the dissemination of highly metastatic BCa cells to the lungs from primary implantation sites in in vivo studies. Thus, differential splicing of the SENP7 regulates either tumor suppression or progression.


Assuntos
Endopeptidases/metabolismo , Transição Epitelial-Mesenquimal/fisiologia , Sequência de Aminoácidos , Animais , Linhagem Celular , Homólogo 5 da Proteína Cromobox , Endopeptidases/química , Endopeptidases/genética , Endopeptidases/fisiologia , Perfilação da Expressão Gênica , Humanos , Dados de Sequência Molecular , RNA Mensageiro/genética , Homologia de Sequência de Aminoácidos
8.
bioRxiv ; 2024 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-38464252

RESUMO

Centrosomes play a fundamental role in nucleating and organizing microtubules in the cell and are vital for faithful chromosome segregation and maintenance of genomic stability. Loss of structural or functional integrity of centrosomes causes genomic instability and is a driver of oncogenesis. The lysine demethylase 4A (KDM4A) is an epigenetic 'eraser' of chromatin methyl marks, which we show also localizes to the centrosome with single molecule resolution. We additionally discovered KDM4A demethylase enzymatic activity is required to maintain centrosome homeostasis, and is required for centrosome integrity, a new functionality unlinked to altered expression of genes regulating centrosome number. We find rather, that KDM4A interacts with both mother and daughter centriolar proteins to localize to the centrosome in all stages of mitosis. Loss of KDM4A results in supernumerary centrosomes and accrual of chromosome segregation errors including chromatin bridges and micronuclei, markers of genomic instability. In summary, these data highlight a novel role for an epigenetic 'eraser' regulating centrosome integrity, mitotic fidelity, and genomic stability at the centrosome.

9.
J Chromatogr Sci ; 60(1): 26-34, 2022 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-33667297

RESUMO

Promising preliminary clinical data have stimulated research on the use of the mammalian target of rapamycin (mTOR) inhibitors in lung cancer. AC1LPSZG is an mTOR inhibitor that can significantly reduce the viability in lung adenosquamous carcinoma cell line HTB-178 cells, showing potential benefits in effective control of non-small cell lung carcinomas. In this study, a sensitive LC-MS/MS analytical method for quantification of AC1LPSZG has been developed and optimized to a running time of 3 min per sample. A linear dose-response for quantification was observed over the range of 10-5000 ng/mL in rat plasma with required precision and accuracy. High extraction recovery was achieved in the ranges of 86.87-102.51% at QC levels from rat plasma without significant matrix effect. Stability profile of AC1LPSZG in rat plasma and in extract after protein precipitation suggested that samples should be processed within 6 h after collection and stored at -80 °C until analysis within 30 days. The method was successfully applied to plasma pharmacokinetics (PK) study of AC1LPSZG in rat, showing the plasma drug concentration followed a two-compartment model.


Assuntos
Plasma , Espectrometria de Massas em Tandem , Animais , Cromatografia Líquida , Ratos , Reprodutibilidade dos Testes
10.
Sci Rep ; 11(1): 10461, 2021 05 17.
Artigo em Inglês | MEDLINE | ID: mdl-34002003

RESUMO

Loss of primary cilia in cells deficient for the tumor suppressor von Hippel Lindau (VHL) arise from elevated Aurora Kinase A (AURKA) levels. VHL in its role as an E3 ubiquitin ligase targets AURKA for degradation and in the absence of VHL, high levels of AURKA result in destabilization of the primary cilium. We identified NVP-BEZ235, a dual PI3K/AKT and mTOR inhibitor, in an image-based high throughput screen, as a small molecule that restored primary cilia in VHL-deficient cells. We identified the ability of AKT to modulate AURKA expression at the transcript and protein level. Independent modulation of AKT and mTOR signaling decreased AURKA expression in cells confirming AURKA as a new signaling node downstream of the PI3K cascade. Corroborating these data, a genetic knockdown of AKT in cells deficient for VHL rescued the ability of these cells to ciliate. Finally, inhibition of AKT/mTOR using NVP-BEZ235 was efficacious in reducing tumor burden in a 786-0 xenograft model of renal cell carcinoma. These data highlight a previously unappreciated signaling node downstream of the AKT/mTOR pathway via AURKA that can be targeted in VHL-null cells to restore ciliogenesis.


Assuntos
Aurora Quinase A/metabolismo , Carcinoma de Células Renais/tratamento farmacológico , Cílios/efeitos dos fármacos , Imidazóis/farmacologia , Neoplasias Renais/tratamento farmacológico , Quinolinas/farmacologia , Doença de von Hippel-Lindau/tratamento farmacológico , Carcinoma de Células Renais/genética , Carcinoma de Células Renais/patologia , Linhagem Celular Tumoral , Cílios/patologia , Técnicas de Silenciamento de Genes , Humanos , Imidazóis/uso terapêutico , Neoplasias Renais/genética , Neoplasias Renais/patologia , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Quinolinas/uso terapêutico , Transdução de Sinais/efeitos dos fármacos , Serina-Treonina Quinases TOR/antagonistas & inibidores , Serina-Treonina Quinases TOR/metabolismo , Carga Tumoral/efeitos dos fármacos , Proteína Supressora de Tumor Von Hippel-Lindau/genética , Proteína Supressora de Tumor Von Hippel-Lindau/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto , Doença de von Hippel-Lindau/complicações , Doença de von Hippel-Lindau/genética , Doença de von Hippel-Lindau/patologia
11.
Sci Adv ; 7(14)2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33811077

RESUMO

Epigenetic effectors "read" marks "written" on chromatin to regulate function and fidelity of the genome. Here, we show that this coordinated read-write activity of the epigenetic machinery extends to the cytoskeleton, with PBRM1 in the PBAF chromatin remodeling complex reading microtubule methyl marks written by the SETD2 histone methyltransferase. PBRM1 binds SETD2 methyl marks via BAH domains, recruiting PBAF components to the mitotic spindle. This read-write activity was required for normal mitosis: Loss of SETD2 methylation or pathogenic BAH domain mutations disrupt PBRM1 microtubule binding and PBAF recruitment and cause genomic instability. These data reveal PBRM1 functions beyond chromatin remodeling with domains that allow it to integrate chromatin and cytoskeletal activity via its acetyl-binding BD and methyl-binding BAH domains, respectively. Conserved coordinated activity of the epigenetic machinery on the cytoskeleton opens a previously unknown window into how chromatin remodeler defects can drive disease via both epigenetic and cytoskeletal dysfunction.


Assuntos
Microtúbulos , Leitura , Cromatina/metabolismo , Montagem e Desmontagem da Cromatina , Citoesqueleto/metabolismo , Microtúbulos/metabolismo
12.
Mol Carcinog ; 49(5): 429-39, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20146253

RESUMO

The tuberous sclerosis complex 2 (Tsc2) gene product, tuberin, acts as a negative regulator of mTOR signaling, and loss of tuberin function leads to tumors of the brain, skin, kidney, heart, and lungs. Previous studies have shown that loss of tuberin function affects the stability and subcellular localization of the cyclin-dependent kinase inhibitor (CKI) p27, although the mechanism(s) by which tuberin modulates p27 stability has/have not been elucidated. Previous studies have also shown that AMP-activated protein kinase (AMPK), which functions in an energy-sensing pathway in the cell, becomes activated in the absence of tuberin. Here we show that in Tsc2-null tumors and cell lines, AMPK activation correlates with an increase in p27 levels, and inhibition of AMPK signaling decreases p27 levels in these cells. In addition, activation of AMPK led to phosphorylation of p27 at the conserved terminal threonine residue of murine p27 (T197) in both in vitro kinase assays and in cells. Phosphorylation of p27 at T197 led to increased interaction between p27 and 14-3-3 proteins and increased the protein stability of p27. Furthermore, activation of AMPK signaling promoted the interaction between p27 and 14-3-3 proteins and increased the stability of the p27 protein in a manner that was dependent on T197. These data identify a conserved mechanism for the regulation of p27 stability via phosphorylation at the terminal threonine (mT197/hT198) and binding of 14-3-3 proteins, which when AMPK is activated results in stabilization of the p27 protein.


Assuntos
Proteínas 14-3-3/metabolismo , Adenilato Quinase/metabolismo , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Animais , Linhagem Celular , Humanos , Camundongos , Fosforilação , Ligação Proteica , Transdução de Sinais
13.
Sci Adv ; 6(40)2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-33008892

RESUMO

The methyltransferase SET domain-containing 2 (SETD2) was originally identified as Huntingtin (HTT) yeast partner B. However, a SETD2 function associated with the HTT scaffolding protein has not been elucidated, and no linkage between HTT and methylation has yet been uncovered. Here, we show that SETD2 is an actin methyltransferase that trimethylates lysine-68 (ActK68me3) in cells via its interaction with HTT and the actin-binding adapter HIP1R. ActK68me3 localizes primarily to the insoluble F-actin cytoskeleton in cells and regulates actin polymerization/depolymerization dynamics. Disruption of the SETD2-HTT-HIP1R axis inhibits actin methylation, causes defects in actin polymerization, and impairs cell migration. Together, these data identify SETD2 as a previously unknown HTT effector regulating methylation and polymerization of actin filaments and provide new avenues for understanding how defects in SETD2 and HTT drive disease via aberrant cytoskeletal methylation.


Assuntos
Actinas , Proteínas de Ligação ao GTP/metabolismo , Histona-Lisina N-Metiltransferase/metabolismo , Lisina , Actinas/metabolismo , Citoesqueleto/metabolismo , Lisina/metabolismo , Metilação , Processamento de Proteína Pós-Traducional
14.
Cancer Res ; 78(12): 3135-3146, 2018 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-29724720

RESUMO

Loss of the short arm of chromosome 3 (3p) occurs early in >95% of clear cell renal cell carcinoma (ccRCC). Nearly ubiquitous 3p loss in ccRCC suggests haploinsufficiency for 3p tumor suppressors as early drivers of tumorigenesis. We previously reported methyltransferase SETD2, which trimethylates H3 histones on lysine 36 (H3K36me3) and is located in the 3p deletion, to also trimethylate microtubules on lysine 40 (αTubK40me3) during mitosis, with αTubK40me3 required for genomic stability. We now show that monoallelic, Setd2-deficient cells retaining H3K36me3, but not αTubK40me3, exhibit a dramatic increase in mitotic defects and micronuclei count, with increased viability compared with biallelic loss. In SETD2-inactivated human kidney cells, rescue with a pathogenic SETD2 mutant deficient for microtubule (αTubK40me3), but not histone (H3K36me3) methylation, replicated this phenotype. Genomic instability (micronuclei) was also a hallmark of patient-derived cells from ccRCC. These data show that the SETD2 tumor suppressor displays a haploinsufficiency phenotype disproportionately impacting microtubule methylation and serves as an early driver of genomic instability.Significance: Loss of a single allele of a chromatin modifier plays a role in promoting oncogenesis, underscoring the growing relevance of tumor suppressor haploinsufficiency in tumorigenesis. Cancer Res; 78(12); 3135-46. ©2018 AACR.


Assuntos
Carcinoma de Células Renais/genética , Cromossomos Humanos Par 3/genética , Histona-Lisina N-Metiltransferase/genética , Neoplasias Renais/genética , Microtúbulos/metabolismo , Animais , Carcinogênese/genética , Carcinoma de Células Renais/patologia , Linhagem Celular Tumoral , Fibroblastos , Técnicas de Silenciamento de Genes , Instabilidade Genômica , Haploinsuficiência , Histona-Lisina N-Metiltransferase/metabolismo , Histonas/metabolismo , Humanos , Neoplasias Renais/patologia , Túbulos Renais Proximais/citologia , Túbulos Renais Proximais/patologia , Lisina/metabolismo , Metilação , Camundongos , Micronúcleos com Defeito Cromossômico
15.
J Mol Biol ; 360(1): 21-36, 2006 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-16753177

RESUMO

Myotonic dystrophy type 2 (DM2) is caused by the extreme expansion of the repeating tetranucleotide CCTG*CAGG sequence from <30 repeats in normal individuals to approximately 11,000 for the full mutation in certain patients. This repeat is in intron 1 of the zinc finger protein 9 gene on chromosome 3q21. Since prior work demonstrated that CTG*CAG and GAA*TTC triplet repeats (responsible for DM1 and Friedreich's ataxia, respectively) can expand by genetic recombination, we investigated the capacity of the DM2 tetranucleotide repeats to also expand during this process. Both gene conversion and unequal crossing over are attractive mechanisms to effect these very large expansions. (CCTG*CAGG)n (where n=30, 75, 114 or 160) repeats showed high recombination crossover frequencies (up to 27-fold higher than the non-repeating control) in an intramolecular plasmid system in Escherichia coli. Furthermore, a distinct orientation effect was observed where orientation II (CAGG on the leading strand template) was more prone to recombine. Expansions of up to double the length of the tetranucleotide repeats were found. Also, the repeating tetranucleotide sequence was more prone to expansions (to give lengths longer than a single repeating tract) than deletions as observed for the CTG*CAG and GAA*TTC repeats. We determined that the DM2 tetranucleotide repeats showed a lower thermodynamic stability when compared to the DM1 trinucleotide repeats, which could make them better targets for DNA repair events, thus explaining their expansion-prone behavior. Genetic studies in SOS-repair mutants revealed high frequencies of recombination crossovers although the SOS-response itself was not induced. Thus, the genetic instabilities of the CCTG*CAGG repeats may be mediated by a recombination-repair mechanism that is influenced by DNA structure.


Assuntos
Escherichia coli/genética , Distrofia Miotônica/genética , Proteínas Serina-Treonina Quinases/genética , Mapeamento Cromossômico , Cromossomos Humanos Par 3 , Reparo do DNA , Escherichia coli/metabolismo , Humanos , Mutação , Miotonina Proteína Quinase , Plasmídeos/metabolismo , Proteínas Serina-Treonina Quinases/química , Recombinação Genética , Especificidade da Espécie , Termodinâmica , Dedos de Zinco
16.
Nucleic Acids Res ; 33(12): 3785-98, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16006624

RESUMO

Substantial progress has been realized in the past several years in our understanding of the molecular mechanisms responsible for the expansions and deletions (genetic instabilities) of repeating tri-, tetra- and pentanucleotide repeating sequences associated with a number of hereditary neurological diseases. These instabilities occur by replication, recombination and repair processes, probably acting in concert, due to slippage of the DNA complementary strands relative to each other. The biophysical properties of the folded-back repeating sequence strands play a critical role in these instabilities. Non-B DNA structural elements (hairpins and slipped structures, DNA unwinding elements, tetraplexes, triplexes and sticky DNA) are described. The replication mechanisms are influenced by pausing of the replication fork, orientation of the repeat strands, location of the repeat sequences relative to replication origins and the flap endonuclease. Methyl-directed mismatch repair, nucleotide excision repair, and repair of damage caused by mutagens are discussed. Genetic recombination and double-strand break repair advances in Escherichia coli, yeast and mammalian models are reviewed. Furthermore, the newly discovered capacities of certain triplet repeat sequences to cause gross chromosomal rearrangements are discussed.


Assuntos
Expansão das Repetições de DNA , Doenças do Sistema Nervoso/genética , Deleção de Sequência , Animais , Aberrações Cromossômicas , DNA/química , Reparo do DNA , Replicação do DNA , Instabilidade Genômica , Humanos , Recombinação Genética , Sequências Repetitivas de Ácido Nucleico
17.
Autophagy ; 12(4): 711-2, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27050462

RESUMO

Peroxisomes are autonomously replicating and highly metabolic organelles necessary for ß-oxidation of fatty acids, a process that generates large amounts of reactive oxygen species (ROS). Maintaining a balance between biogenesis and degradation of peroxisomes is essential to maintain cellular redox balance, but how cells do this has remained somewhat of a mystery. While it is known that peroxisomes can be degraded via selective autophagy (pexophagy), little is known about how mammalian cells regulate pexophagy to maintain peroxisome homeostasis. We have uncovered a mechanism for regulating pexophagy in mammalian cells that defines a new role for ATM (ATM serine/threonine kinase) kinase as a "first responder" to peroxisomal ROS. ATM is delivered to the peroxisome by the PEX5 import receptor, which recognizes an SRL sequence located at the C terminus of ATM to localize this kinase to peroxisomes. In response to ROS, the ATM kinase is activated and performs 2 functions: i) it signals to AMPK, which activates TSC2 to suppresses MTORC1 and phosphorylates ULK1 to induce autophagy, and ii) targets specific peroxisomes for pexophagy by phosphorylating PEX5 at Ser141, which triggers ubiquitination of PEX5 at Lys209 and binding of the autophagy receptor protein SQSTM1/p62 to induce pexophagy.


Assuntos
Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , Autofagia , Peroxissomos/metabolismo , Animais , Humanos , Mamíferos/metabolismo , Modelos Biológicos , Espécies Reativas de Oxigênio/metabolismo
19.
MAbs ; 8(8): 1590-1597, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27594515

RESUMO

Posttranslational modifications (PTMs) on microtubules differentiate these cytoskeletal elements for a variety of cellular functions. We recently identified SETD2 as a dual-function histone and microtubule methyltransferase, and methylation as a new microtubule PTM that occurs on lysine 40 of α-tubulin, which is trimethylated (α-TubK40me3) by SETD2. In the course of these studies, we generated polyclonal (α-TubK40me3 pAb) and monoclonal (α-TubK40me3 mAb) antibodies to a methylated α-tubulin peptide (GQMPSD-Kme3-TIGGGDC). Here, we characterize these antibodies, and the specific mono-, di- or tri-methylated lysine residues they recognize. While both the pAb and mAb antibodies recognized lysines methylated by SETD2 on microtubules and histones, the clone 18 mAb was more specific for methylated microtubules, with little cross-reactivity for methylated histones. The clone 18 mAb recognized specific subsets of microtubules during mitosis and cytokinesis, and lacked the chromatin staining seen by immunocytochemistry with the pAb. Western blot analysis using these antibodies revealed that methylated α-tubulin migrated faster than unmethylated α-tubulin, suggesting methylation may be a signal for additional processing of α-tubulin and/or microtubules. As the first reagents that specifically recognize methylated α-tubulin, these antibodies are a valuable tool for studying this new modification of the cytoskeleton, and the function of methylated microtubules.


Assuntos
Anticorpos Monoclonais/imunologia , Lisina/imunologia , Tubulina (Proteína)/química , Tubulina (Proteína)/imunologia , Anticorpos/imunologia , Humanos , Lisina/química , Lisina/metabolismo , Metilação , Microtúbulos/química , Microtúbulos/imunologia , Microtúbulos/metabolismo , Mitose/fisiologia , Processamento de Proteína Pós-Traducional , Tubulina (Proteína)/metabolismo
20.
Nat Cell Biol ; 17(10): 1259-1269, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26344566

RESUMO

Peroxisomes are highly metabolic, autonomously replicating organelles that generate reactive oxygen species (ROS) as a by-product of fatty acid ß-oxidation. Consequently, cells must maintain peroxisome homeostasis, or risk pathologies associated with too few peroxisomes, such as peroxisome biogenesis disorders, or too many peroxisomes, inducing oxidative damage and promoting diseases such as cancer. We report that the PEX5 peroxisome import receptor binds ataxia-telangiectasia mutated (ATM) and localizes this kinase to the peroxisome. In response to ROS, ATM signalling activates ULK1 and inhibits mTORC1 to induce autophagy. Specificity for autophagy of peroxisomes (pexophagy) is provided by ATM phosphorylation of PEX5 at Ser 141, which promotes PEX5 monoubiquitylation at Lys 209, and recognition of ubiquitylated PEX5 by the autophagy adaptor protein p62, directing the autophagosome to peroxisomes to induce pexophagy. These data reveal an important new role for ATM in metabolism as a sensor of ROS that regulates pexophagy.


Assuntos
Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , Autofagia , Peroxissomos/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Proteínas Mutadas de Ataxia Telangiectasia/genética , Western Blotting , Células Cultivadas , Células HEK293 , Células Hep G2 , Humanos , Peróxido de Hidrogênio/farmacologia , Células MCF-7 , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos Knockout , Microscopia Eletrônica , Microscopia de Fluorescência , Complexos Multiproteicos/metabolismo , Mutação , Receptor 1 de Sinal de Orientação para Peroxissomos , Peroxissomos/ultraestrutura , Fagossomos/metabolismo , Fagossomos/ultraestrutura , Fosforilação/efeitos dos fármacos , Ligação Proteica , Interferência de RNA , Receptores Citoplasmáticos e Nucleares/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteína Sequestossoma-1 , Serina/genética , Serina/metabolismo , Serina-Treonina Quinases TOR/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA