Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 93
Filtrar
Mais filtros

Tipo de documento
Intervalo de ano de publicação
1.
Cell ; 159(1): 148-162, 2014 Sep 25.
Artigo em Inglês | MEDLINE | ID: mdl-25219674

RESUMO

Pseudouridine is the most abundant RNA modification, yet except for a few well-studied cases, little is known about the modified positions and their function(s). Here, we develop Ψ-seq for transcriptome-wide quantitative mapping of pseudouridine. We validate Ψ-seq with spike-ins and de novo identification of previously reported positions and discover hundreds of unique sites in human and yeast mRNAs and snoRNAs. Perturbing pseudouridine synthases (PUS) uncovers which pseudouridine synthase modifies each site and their target sequence features. mRNA pseudouridinylation depends on both site-specific and snoRNA-guided pseudouridine synthases. Upon heat shock in yeast, Pus7p-mediated pseudouridylation is induced at >200 sites, and PUS7 deletion decreases the levels of otherwise pseudouridylated mRNA, suggesting a role in enhancing transcript stability. rRNA pseudouridine stoichiometries are conserved but reduced in cells from dyskeratosis congenita patients, where the PUS DKC1 is mutated. Our work identifies an enhanced, transcriptome-wide scope for pseudouridine and methods to dissect its underlying mechanisms and function.


Assuntos
Pseudouridina/análise , RNA Mensageiro/química , RNA não Traduzido/química , Animais , Candida albicans/genética , Candida albicans/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Disceratose Congênita/genética , Disceratose Congênita/metabolismo , Perfilação da Expressão Gênica , Humanos , Transferases Intramoleculares/química , Transferases Intramoleculares/metabolismo , Camundongos , Dados de Sequência Molecular , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Pseudouridina/metabolismo , RNA/química , RNA/genética , RNA Ribossômico/química , RNA Ribossômico/genética , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/metabolismo , Especificidade por Substrato , Telomerase/química , Telomerase/genética
2.
Hum Mol Genet ; 33(9): 818-834, 2024 Apr 18.
Artigo em Inglês | MEDLINE | ID: mdl-38641551

RESUMO

Telomeres are nucleoprotein structures at the end of chromosomes that maintain their integrity. Mutations in genes coding for proteins involved in telomere protection and elongation produce diseases such as dyskeratosis congenita or idiopathic pulmonary fibrosis known as telomeropathies. These diseases are characterized by premature telomere shortening, increased DNA damage and oxidative stress. Genetic diagnosis of telomeropathy patients has identified mutations in the genes TERT and TERC coding for telomerase components but the functional consequences of many of these mutations still have to be experimentally demonstrated. The activity of twelve TERT and five TERC mutants, five of them identified in Spanish patients, has been analyzed. TERT and TERC mutants were expressed in VA-13 human cells that express low telomerase levels and the activity induced was analyzed. The production of reactive oxygen species, DNA oxidation and TRF2 association at telomeres, DNA damage response and cell apoptosis were determined. Most mutations presented decreased telomerase activity, as compared to wild-type TERT and TERC. In addition, the expression of several TERT and TERC mutants induced oxidative stress, DNA oxidation, DNA damage, decreased recruitment of the shelterin component TRF2 to telomeres and increased apoptosis. These observations might indicate that the increase in DNA damage and oxidative stress observed in cells from telomeropathy patients is dependent on their TERT or TERC mutations. Therefore, analysis of the effect of TERT and TERC mutations of unknown function on DNA damage and oxidative stress could be of great utility to determine the possible pathogenicity of these variants.


Assuntos
Disceratose Congênita , Telomerase , Humanos , Apoptose/genética , DNA/metabolismo , Dano ao DNA/genética , Disceratose Congênita/genética , Disceratose Congênita/metabolismo , Disceratose Congênita/patologia , Mutação , Estresse Oxidativo/genética , RNA/genética , Telomerase/genética , Telomerase/metabolismo , Telômero/genética , Telômero/metabolismo
3.
EMBO J ; 39(21): e103420, 2020 11 02.
Artigo em Inglês | MEDLINE | ID: mdl-32935380

RESUMO

Short telomeres are a principal defining feature of telomere biology disorders, such as dyskeratosis congenita (DC), for which there are no effective treatments. Here, we report that primary fibroblasts from DC patients and late generation telomerase knockout mice display lower nicotinamide adenine dinucleotide (NAD) levels, and an imbalance in the NAD metabolome that includes elevated CD38 NADase and reduced poly(ADP-ribose) polymerase and SIRT1 activities, respectively, affecting many associated biological pathways. Supplementation with the NAD precursor, nicotinamide riboside, and CD38 inhibition improved NAD homeostasis, thereby alleviating telomere damage, defective mitochondrial biosynthesis and clearance, cell growth retardation, and cellular senescence of DC fibroblasts. These findings reveal a direct, underlying role of NAD dysregulation when telomeres are short and underscore its relevance to the pathophysiology and interventions of human telomere-driven diseases.


Assuntos
Disceratose Congênita/genética , Disceratose Congênita/metabolismo , Fibroblastos/metabolismo , NAD/metabolismo , Telomerase/genética , Telômero/metabolismo , ADP-Ribosil Ciclase 1/genética , Animais , Encéfalo/patologia , Linhagem Celular , Senescência Celular , Disceratose Congênita/patologia , Feminino , Homeostase , Humanos , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Knockout , Mitocôndrias/metabolismo , Niacinamida/análogos & derivados , Niacinamida/metabolismo , Fenótipo , Poli(ADP-Ribose) Polimerase-1/metabolismo , Compostos de Piridínio/metabolismo , Telomerase/metabolismo
4.
Blood ; 139(16): 2427-2440, 2022 04 21.
Artigo em Inglês | MEDLINE | ID: mdl-35007328

RESUMO

Inherited bone marrow failure syndromes (IBMFSs) are a group of disorders typified by impaired production of 1 or several blood cell types. The telomere biology disorders dyskeratosis congenita (DC) and its severe variant, Høyeraal-Hreidarsson (HH) syndrome, are rare IBMFSs characterized by bone marrow failure, developmental defects, and various premature aging complications associated with critically short telomeres. We identified biallelic variants in the gene encoding the 5'-to-3' DNA exonuclease Apollo/SNM1B in 3 unrelated patients presenting with a DC/HH phenotype consisting of early-onset hypocellular bone marrow failure, B and NK lymphopenia, developmental anomalies, microcephaly, and/or intrauterine growth retardation. All 3 patients carry a homozygous or compound heterozygous (in combination with a null allele) missense variant affecting the same residue L142 (L142F or L142S) located in the catalytic domain of Apollo. Apollo-deficient cells from patients exhibited spontaneous chromosome instability and impaired DNA repair that was complemented by CRISPR/Cas9-mediated gene correction. Furthermore, patients' cells showed signs of telomere fragility that were not associated with global reduction of telomere length. Unlike patients' cells, human Apollo KO HT1080 cell lines showed strong telomere dysfunction accompanied by excessive telomere shortening, suggesting that the L142S and L142F Apollo variants are hypomorphic. Collectively, these findings define human Apollo as a genome caretaker and identify biallelic Apollo variants as a genetic cause of a hitherto unrecognized severe IBMFS that combines clinical hallmarks of DC/HH with normal telomere length.


Assuntos
Disceratose Congênita , Deficiência Intelectual , Microcefalia , Disceratose Congênita/genética , Disceratose Congênita/metabolismo , Retardo do Crescimento Fetal , Humanos , Deficiência Intelectual/genética , Microcefalia/genética , Microcefalia/metabolismo , Mutação , Telômero/genética , Telômero/metabolismo
5.
Hereditas ; 160(1): 38, 2023 Dec 11.
Artigo em Inglês | MEDLINE | ID: mdl-38082360

RESUMO

BACKGROUND: Dyskeratosis congenita 1 (DKC1), a critical component of telomerase complex, is highly expressed in a variety of human cancers. However, the association of DKC1 with cancer occurrence and development stages is not clear, making a pan-cancer analysis crucial. METHODS: We conducted a study using various bioinformatic databases such as TIMER, GEPIA, UALCAN, and KM plotter Analysis to examine the different expressions of DKC1 in multiple tissues and its correlation with pathological stages. Through KEGG analysis, GO enrichment analysis and Venn analysis, we were able to reveal DKC1-associated genes and signaling pathways. In addition, we performed several tests including the CCK, wound healing assay, cell cycle arrest assay, transwell assay and Sa-ß-gal staining on DKC1-deleted MDA-231 cells. RESULTS: Our study demonstrates that DKC1 has relatively low expression specificity in different tissues. Furthermore, we found that in ACC, KICH, KIRP and LIHC, the expression level of DKC1 is positively correlated with pathological stages. Conversely, in NHSC, KIRP, LGG, LIHC, MESO and SARC, we observed a negative influence of DKC1 expression level on the overall survival rate. We also found a significant positive correlation between DKC1 expression and Tumor Mutational Burden in 14 tumors. Additionally, we observed a significantly negative impact of DKC1 DNA methylation on gene expression at the promoter region in BRCA. We also identified numerous phosphorylation sites concentrated at the C-terminus of the DKC1 protein. Our GO analysis revealed a correlation between DKC1 and ribosomal biosynthesis pathways, and the common element UTP14A was identified. We also observed decreased rates of cell proliferation, migration and invasion abilities in DKC1-knockout MDA-MB-231 cell lines. Furthermore, DKC1-knockout induced cell cycle arrest and caused cell senescence. CONCLUSIONS: Our findings suggest that the precise expression of DKC1 is closely associated with the occurrence and developmental stages of cancer in multiple tissues. Depletion of DKC1 can inhibit the abilities of cancer cells to proliferate, migrate, and invade by arresting the cell cycle and inducing cell senescence. Therefore, DKC1 may be a valuable prognostic biomarker for the diagnosis and treatment of cancer in various tissues.


Assuntos
Disceratose Congênita , Neoplasias , Humanos , Prognóstico , Proteínas de Ciclo Celular/genética , Disceratose Congênita/genética , Disceratose Congênita/metabolismo , Disceratose Congênita/patologia , Neoplasias/genética , Biomarcadores , Proteínas Nucleares/genética
6.
Blood ; 133(12): 1308-1312, 2019 03 21.
Artigo em Inglês | MEDLINE | ID: mdl-30728146

RESUMO

Reduced levels of TERC, the telomerase RNA component, cause dyskeratosis congenita (DC) in patients harboring mutations in TERC, PARN, NOP10, NHP2, NAF1, or DKC1. Inhibition of the noncanonical poly(A) polymerase PAPD5, or the exosome RNA degradation complex, partially restores TERC levels in immortalized DKC1 mutant cells, but it remains unknown if modulation of posttranscriptional processing of TERC could improve hematopoietic output in DC. We used human embryonic stem cells (hESCs) with a common dyskerin mutation (DKC1_A353V), which have defective telomere maintenance and reduced definitive hematopoietic potential, to understand the effects of reducing EXOSC3 activity, or silencing PAPD5-mediated oligoadenylation, on hematopoietic progenitor specification and function in DC. Reduction of EXOSC3 or PAPD5 levels in DKC1 mutant hESCs led to functional improvements in TERC levels and telomerase activity, with concomitant telomere elongation and reduced levels of DNA damage signaling. Interestingly, the silencing of PAPD5, but not EXOSC3, significantly restored definitive hematopoietic potential in DKC1 mutant cells. Mechanistically, we show that PAPD5 inhibition is sustained in differentiated CD34+ cells, with a concomitant increase in mature, functional, forms of TERC, indicating that regulation of PAPD5 is a potential strategy to reverse hematologic dysfunction in DC patients.


Assuntos
Disceratose Congênita/prevenção & controle , Células-Tronco Embrionárias/citologia , Hematopoese , Mutação , RNA Nucleotidiltransferases/antagonistas & inibidores , Processamento Pós-Transcricional do RNA , RNA/metabolismo , Telomerase/metabolismo , Proteínas de Ciclo Celular/genética , Disceratose Congênita/metabolismo , Disceratose Congênita/patologia , Células-Tronco Embrionárias/metabolismo , Complexo Multienzimático de Ribonucleases do Exossomo/genética , Complexo Multienzimático de Ribonucleases do Exossomo/metabolismo , Humanos , Proteínas Nucleares/genética , RNA/genética , RNA Nucleotidiltransferases/genética , RNA Nucleotidiltransferases/metabolismo , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , Telomerase/genética , Telômero
7.
J Biol Chem ; 292(11): 4593-4601, 2017 03 17.
Artigo em Inglês | MEDLINE | ID: mdl-28154186

RESUMO

Naturally occurring mutations in the ribonucleoprotein reverse transcriptase, telomerase, are associated with the bone marrow failure syndromes dyskeratosis congenita, aplastic anemia, and idiopathic pulmonary fibrosis. However, the mechanism by which these mutations impact telomerase function remains unknown. Here we present the structure of the human telomerase C-terminal extension (or thumb domain) determined by the method of single-wavelength anomalous diffraction to 2.31 Å resolution. We also used direct telomerase activity and nucleic acid binding assays to explain how naturally occurring mutations within this portion of telomerase contribute to human disease. The single mutations localize within three highly conserved regions of the telomerase thumb domain referred to as motifs E-I (thumb loop and helix), E-II, and E-III (the FVYL pocket, comprising the hydrophobic residues Phe-1012, Val-1025, Tyr-1089, and Leu-1092). Biochemical data show that the mutations associated with dyskeratosis congenita, aplastic anemia, and idiopathic pulmonary fibrosis disrupt the binding between the protein subunit reverse transcriptase of the telomerase and its nucleic acid substrates leading to loss of telomerase activity and processivity. Collectively our data show that although these mutations do not alter the overall stability or expression of telomerase reverse transcriptase, these rare genetic disorders are associated with an impaired telomerase holoenzyme that is unable to correctly assemble with its nucleic acid substrates, leading to incomplete telomere extension and telomere attrition, which are hallmarks of these diseases.


Assuntos
Anemia Aplástica/genética , Doenças da Medula Óssea/genética , Hemoglobinúria Paroxística/genética , Mutação Puntual , Telomerase/genética , Sequência de Aminoácidos , Anemia Aplástica/metabolismo , Medula Óssea/metabolismo , Doenças da Medula Óssea/metabolismo , Transtornos da Insuficiência da Medula Óssea , Cristalografia por Raios X , Disceratose Congênita/genética , Disceratose Congênita/metabolismo , Células HEK293 , Hemoglobinúria Paroxística/metabolismo , Humanos , Fibrose Pulmonar Idiopática/genética , Fibrose Pulmonar Idiopática/metabolismo , Modelos Moleculares , Ácidos Nucleicos/metabolismo , Conformação Proteica , Domínios Proteicos , Telomerase/química , Telomerase/metabolismo , Telômero/metabolismo
8.
RNA Biol ; 13(8): 696-706, 2016 08 02.
Artigo em Inglês | MEDLINE | ID: mdl-26400640

RESUMO

The importance of telomere function for human health is exemplified by a collection of Mendelian disorders referred to as the telomere biology disorders (TBDs), telomeropathies, or syndromes of telomere shortening. Collectively, the TBDs cover a spectrum of conditions from multisystem disease presenting in infancy to isolated disease presentations in adulthood, most notably idiopathic pulmonary fibrosis. Eleven genes have been found mutated in the TBDs to date, each of which is linked to some aspect of telomere maintenance. This review summarizes the molecular defects that result from mutations in these genes, highlighting recent advances, including the addition of PARN to the TBD gene family and the discovery of heterozygous mutations in RTEL1 as a cause of familial pulmonary fibrosis.


Assuntos
Estudos de Associação Genética , Predisposição Genética para Doença , Encurtamento do Telômero , Telômero/genética , Telômero/metabolismo , Animais , Replicação do DNA , Disceratose Congênita/genética , Disceratose Congênita/metabolismo , Ativação Enzimática/genética , Exorribonucleases/genética , Exorribonucleases/metabolismo , Instabilidade Genômica , Humanos , Mutação , Conformação de Ácido Nucleico , Ligação Proteica , Transporte Proteico , Síndrome , Telomerase/genética , Telomerase/metabolismo
9.
Proc Natl Acad Sci U S A ; 110(36): E3408-16, 2013 Sep 03.
Artigo em Inglês | MEDLINE | ID: mdl-23959892

RESUMO

Telomeres repress the DNA damage response at the natural chromosome ends to prevent cell-cycle arrest and maintain genome stability. Telomeres are elongated by telomerase in a tightly regulated manner to ensure a sufficient number of cell divisions throughout life, yet prevent unlimited cell division and cancer development. Hoyeraal-Hreidarsson syndrome (HHS) is characterized by accelerated telomere shortening and a broad range of pathologies, including bone marrow failure, immunodeficiency, and developmental defects. HHS-causing mutations have previously been found in telomerase and the shelterin component telomeric repeat binding factor 1 (TRF1)-interacting nuclear factor 2 (TIN2). We identified by whole-genome exome sequencing compound heterozygous mutations in four siblings affected with HHS, in the gene encoding the regulator of telomere elongation helicase 1 (RTEL1). Rtel1 was identified in mouse by its genetic association with telomere length. However, its mechanism of action and whether it regulates telomere length in human remained unknown. Lymphoblastoid cell lines obtained from a patient and from the healthy parents carrying heterozygous RTEL1 mutations displayed telomere shortening, fragility and fusion, and growth defects in culture. Ectopic expression of WT RTEL1 suppressed the telomere shortening and growth defect, confirming the causal role of the RTEL1 mutations in HHS and demonstrating the essential function of human RTEL1 in telomere protection and elongation. Finally, we show that human RTEL1 interacts with the shelterin protein TRF1, providing a potential recruitment mechanism of RTEL1 to telomeres.


Assuntos
DNA Helicases/genética , Disceratose Congênita/genética , Retardo do Crescimento Fetal/genética , Deficiência Intelectual/genética , Microcefalia/genética , Mutação , Telômero/genética , Animais , Sequência de Bases , Western Blotting , Proliferação de Células , Células Cultivadas , DNA Helicases/metabolismo , Disceratose Congênita/metabolismo , Disceratose Congênita/patologia , Saúde da Família , Feminino , Retardo do Crescimento Fetal/metabolismo , Retardo do Crescimento Fetal/patologia , Expressão Gênica , Instabilidade Genômica/genética , Células HeLa , Humanos , Hibridização in Situ Fluorescente , Deficiência Intelectual/metabolismo , Deficiência Intelectual/patologia , Masculino , Camundongos , Microcefalia/metabolismo , Microcefalia/patologia , Linhagem , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Encurtamento do Telômero/genética , Proteína 1 de Ligação a Repetições Teloméricas/genética , Proteína 1 de Ligação a Repetições Teloméricas/metabolismo
10.
Hum Mol Genet ; 22(17): 3498-507, 2013 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-23660516

RESUMO

Mutations in the dyskerin gene (DKC1) cause X-linked dyskeratosis congenita (DC), a rare and fatal premature aging syndrome characterized by defective telomere maintenance. Dyskerin is a highly conserved nucleolar protein, and a component of the human telomerase complex that is essential for human telomerase RNA (hTR) stability. However, its regulation remains poorly understood. Here, we report that dyskerin can be modified by small ubiquitin-like modifiers (SUMOs). We find that human DC-causing mutations in highly conserved dyskerin SUMOylation consensus sites lead to impaired hTR accumulation, telomerase activity and telomere maintenance. Finally, we show that modification of dyskerin by SUMOylation is required for its stability. Our findings provide the first evidence that dyskerin stability is regulated by SUMOylation and that mutations altering dyskerin SUMOylation can lead to defects in telomere maintenance that are characteristics of DC.


Assuntos
Proteínas de Ciclo Celular/genética , Disceratose Congênita/genética , Proteínas Nucleares/genética , RNA/metabolismo , Telomerase/metabolismo , Homeostase do Telômero , Telômero/metabolismo , Sequência de Aminoácidos , Disceratose Congênita/metabolismo , Técnicas de Silenciamento de Genes , Células HEK293 , Humanos , Dados de Sequência Molecular , Mutação , Filogenia , Estabilidade Proteica , RNA/genética , Alinhamento de Sequência , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/metabolismo , Sumoilação , Telomerase/genética
11.
Hum Mol Genet ; 22(16): 3239-49, 2013 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-23591994

RESUMO

Hoyeraal-Hreidarsson syndrome (HHS), a severe variant of dyskeratosis congenita (DC), is characterized by early onset bone marrow failure, immunodeficiency and developmental defects. Several factors involved in telomere length maintenance and/or protection are defective in HHS/DC, underlining the relationship between telomere dysfunction and these diseases. By combining whole-genome linkage analysis and exome sequencing, we identified compound heterozygous RTEL1 (regulator of telomere elongation helicase 1) mutations in three patients with HHS from two unrelated families. RTEL1 is a DNA helicase that participates in DNA replication, DNA repair and telomere integrity. We show that, in addition to short telomeres, RTEL1-deficient cells from patients exhibit hallmarks of genome instability, including spontaneous DNA damage, anaphase bridges and telomeric aberrations. Collectively, these results identify RTEL1 as a novel HHS-causing gene and highlight its role as a genomic caretaker in humans.


Assuntos
DNA Helicases/genética , Disceratose Congênita/genética , Retardo do Crescimento Fetal/genética , Instabilidade Genômica , Deficiência Intelectual/genética , Microcefalia/genética , Homeostase do Telômero/genética , Encurtamento do Telômero , Telômero/metabolismo , Células Cultivadas , Pré-Escolar , Dano ao DNA , DNA Helicases/química , DNA Helicases/deficiência , DNA Helicases/metabolismo , Replicação do DNA , Disceratose Congênita/metabolismo , Exoma , Feminino , Retardo do Crescimento Fetal/metabolismo , Ligação Genética , Humanos , Lactente , Deficiência Intelectual/metabolismo , Masculino , Microcefalia/metabolismo , Mutação , Alinhamento de Sequência , Análise de Sequência de RNA , Telomerase/genética , Telomerase/metabolismo , Telômero/ultraestrutura
12.
EMBO J ; 30(24): 5010-20, 2011 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-22117216

RESUMO

Shq1 is a conserved protein required for the biogenesis of eukaryotic H/ACA ribonucleoproteins (RNPs), including human telomerase. We report the structure of the Shq1-specific domain alone and in complex with H/ACA RNP proteins Cbf5, Nop10 and Gar1. The Shq1-specific domain adopts a novel helical fold and primarily contacts the PUA domain and the otherwise disordered C-terminal extension (CTE) of Cbf5. The structure shows that dyskeratosis congenita mutations found in the CTE of human Cbf5 likely interfere with Shq1 binding. However, most mutations in the PUA domain are not located at the Shq1-binding surface and also have little effect on the yeast Cbf5-Shq1 interaction. Shq1 binds Cbf5 independently of the H/ACA RNP proteins Nop10, Gar1 and Nhp2 and the assembly factor Naf1, but shares an overlapping binding surface with H/ACA RNA. Shq1 point mutations that disrupt Cbf5 interaction suppress yeast growth particularly at elevated temperatures. Our results suggest that Shq1 functions as an assembly chaperone that protects the Cbf5 protein complexes from non-specific RNA binding and aggregation before assembly of H/ACA RNA.


Assuntos
Disceratose Congênita/metabolismo , Hidroliases/química , Proteínas Associadas aos Microtúbulos/química , Proteínas Nucleares/química , Proteínas de Ligação a RNA/química , Ribonucleoproteínas Nucleares Pequenas/química , Ribonucleoproteínas Nucleolares Pequenas/química , Ribonucleoproteínas/biossíntese , Proteínas de Saccharomyces cerevisiae/química , Sequência de Aminoácidos , Cristalografia por Raios X , Disceratose Congênita/genética , Humanos , Hidroliases/genética , Proteínas Associadas aos Microtúbulos/genética , Dados de Sequência Molecular , Mutação , Proteínas Nucleares/genética , Conformação Proteica , Proteínas de Ligação a RNA/genética , Ribonucleoproteínas Nucleares Pequenas/genética , Ribonucleoproteínas Nucleolares Pequenas/genética , Proteínas de Saccharomyces cerevisiae/genética
13.
Br J Haematol ; 170(4): 457-71, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25940403

RESUMO

Hoyeraal-Hreidarsson (HH) syndrome is a multisystem genetic disorder characterized by very short telomeres and considered a clinically severe variant of dyskeratosis congenita. The main cause of mortality, usually in early childhood, is bone marrow failure. Mutations in several telomere biology genes have been reported to cause HH in about 60% of the HH patients, but the genetic defects in the rest of the patients are still unknown. Understanding the aetiology of HH and its diverse manifestations is challenging because of the complexity of telomere biology and the multiple telomeric and non-telomeric functions played by telomere-associated proteins in processes such as telomere replication, telomere protection, DNA damage response and ribosome and spliceosome assembly. Here we review the known clinical complications, molecular defects and germline mutations associated with HH, and elucidate possible mechanistic explanations and remaining questions in our understanding of the disease.


Assuntos
Disceratose Congênita/genética , Disceratose Congênita/metabolismo , Retardo do Crescimento Fetal/genética , Retardo do Crescimento Fetal/metabolismo , Deficiência Intelectual/genética , Deficiência Intelectual/metabolismo , Microcefalia/genética , Microcefalia/metabolismo , Mutação , Homeostase do Telômero/genética , Telômero/genética , Telômero/metabolismo , Disceratose Congênita/patologia , Retardo do Crescimento Fetal/patologia , Humanos , Deficiência Intelectual/patologia , Microcefalia/patologia , Telômero/patologia
14.
Clin Chem Lab Med ; 53(11): 1661-78, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25803084

RESUMO

Aging is a complex biological process characterized by a progressive decline of organ functions leading to an increased risk of age-associated diseases and death. Decades of intensive research have identified a range of molecular and biochemical pathways contributing to aging. However, many aspects regarding the regulation and interplay of these pathways are insufficiently understood. Telomere dysfunction and genomic instability appear to be of critical importance for aging at a cellular level. For example, age-related diseases and premature aging syndromes are frequently associated with telomere shortening. Telomeres are repetitive nucleotide sequences that together with the associated sheltrin complex protect the ends of chromosomes and maintain genomic stability. Recent studies suggest that micronutrients, such as vitamin D, folate and vitamin B12, are involved in telomere biology and cellular aging. In particular, vitamin D is important for a range of vital cellular processes including cellular differentiation, proliferation and apoptosis. As a result of the multiple functions of vitamin D it has been speculated that vitamin D might play a role in telomere biology and genomic stability. Here we review existing knowledge about the link between telomere biology and cellular aging with a focus on the role of vitamin D. We searched the literature up to November 2014 for human studies, animal models and in vitro experiments that addressed this topic.


Assuntos
Envelhecimento , Doenças Cardiovasculares/metabolismo , Senescência Celular , Diabetes Mellitus Tipo 2/metabolismo , Disceratose Congênita/metabolismo , Doenças Neurodegenerativas/metabolismo , Telômero/metabolismo , Vitamina D/metabolismo , Envelhecimento/genética , Envelhecimento/metabolismo , Envelhecimento/patologia , Animais , Doenças Cardiovasculares/genética , Diabetes Mellitus Tipo 2/genética , Disceratose Congênita/genética , Humanos , Doenças Neurodegenerativas/genética , Telômero/genética
15.
Hum Mol Genet ; 21(4): 721-9, 2012 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-22058290

RESUMO

X-linked dyskeratosis congenita (X-DC) is caused by mutations in the housekeeping nucleolar protein dyskerin. Amino acid changes associated with X-DC are remarkably heterogeneous. Peripheral mononuclear blood cells and fibroblasts isolated from X-DC patients harbor lower steady-state telomerase RNA (TER) levels and shorter telomeres than healthy age-matched controls. Previously, we showed that retroviral expression of recombinant TER, together with expression of recombinant telomerase reverse transcriptase, restored telomere maintenance and proliferative capacity in X-DC patient cells. Using rare X-DC isoforms (ΔL37 and A386T dyskerin), we showed that telomere maintenance defects observed in X-DC are solely due to decreased steady-state levels of TER. Disease-associated reductions in steady-state TER levels cause deficiencies in telomere maintenance. Here, we confirm these findings in other primary X-DC patient cell lines coding for the most common (A353V dyskerin) and more clinically severe (K314R and A353V dyskerin) X-DC isoforms. Using cell lines derived from these patients, we also examined the steady-state levels of other hinge-ACA motif RNAs and did not find differences in their in vivo accumulations. We show, for the first time, that purified telomerase holoenzyme complexes from different X-DC cells have normal catalytic activity. Our data confirm that dyskerin promotes TER stability in vivo, endorsing the development of TER supplementation strategies for the treatment of X-DC.


Assuntos
Proteínas de Ciclo Celular/genética , Disceratose Congênita/genética , Disceratose Congênita/metabolismo , Proteínas Nucleares/genética , Telomerase/metabolismo , Telômero/genética , Telômero/metabolismo , Biocatálise , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular , Disceratose Congênita/enzimologia , Disceratose Congênita/patologia , Fibroblastos/enzimologia , Fibroblastos/metabolismo , Fibroblastos/patologia , Expressão Gênica , Genótipo , Holoenzimas/isolamento & purificação , Holoenzimas/metabolismo , Humanos , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Mutação , Proteínas Nucleares/metabolismo , Motivos de Nucleotídeos , RNA/genética , RNA/metabolismo , Estabilidade de RNA , Telomerase/genética , Telomerase/isolamento & purificação , Homeostase do Telômero
16.
EMBO J ; 29(11): 1865-76, 2010 Jun 02.
Artigo em Inglês | MEDLINE | ID: mdl-20453831

RESUMO

Defects in ribosome biogenesis and function are present in a growing list of human syndromes associated with cancer susceptibility. One example is X-linked dyskeratosis congenita (X-DC) in which the DKC1 gene, encoding for an enzyme that modifies ribosomal RNA, is found to be mutated. How ribosome dysfunction leads to cancer remains poorly understood. A critical cellular response that counteracts cellular transformation is oncogene-induced senescence (OIS). Here, we show that during OIS, a switch between cap- and internal ribosome entry site (IRES)-dependent translation occurs. During this switch, an IRES element positioned in the 5'untranslated region of p53 is engaged and facilitates p53 translation. We further show that in DKC1(m) cells, p53 IRES-dependent translation is impaired during OIS ex vivo and on DNA damage in vivo. This defect in p53 translation perturbs the cellular response that counteracts oncogenic insult. We extend these findings to X-DC human patient cells in which similar impairments in p53 IRES-dependent translation are observed. Importantly, re-introduction of wild-type DKC1 restores p53 expression in these cells. These results provide insight into the basis for cancer susceptibility in human syndromes associated with ribosome dysfunction.


Assuntos
Envelhecimento/genética , Disceratose Congênita/genética , Genes p53 , Ribossomos/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Dano ao DNA , Disceratose Congênita/metabolismo , Humanos , Oncogenes , RNA Ribossômico/genética , RNA Ribossômico/metabolismo , Ribossomos/genética , Proteína Supressora de Tumor p53/genética , Regiões não Traduzidas
18.
Br J Haematol ; 165(3): 349-57, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24666134

RESUMO

Dyskeratosis congenita (DC) is an inherited bone marrow failure syndrome and telomere biology disorder characterized by dysplastic nails, reticular skin pigmentation and oral leucoplakia. Androgens are a standard therapeutic option for bone marrow failure in those patients with DC who are unable to undergo haematopoietic stem cell transplantation, but there are no systematic data on its use in those patients. We evaluated haematological response and side effects of androgen therapy in 16 patients with DC in our observational cohort study. Untreated DC patients served as controls. Seventy percent of treated DC patients had a haematological response with red blood cell and/or platelet transfusion independence. The expected age-related decline in telomere length was noted in androgen-treated patients. All treated DC patients had at least one significant lipid abnormality. Additional treatment-related findings included a significant decrease in thyroid binding globulin, accelerated growth in pre-pubertal children and splenic peliosis in two patients. Liver enzymes were elevated in both androgen-treated and untreated patients, suggesting underlying liver involvement in DC. This study suggests that androgen therapy can be effectively used to treat bone marrow failure in DC, but that side effects need to be closely monitored.


Assuntos
Androgênios/uso terapêutico , Disceratose Congênita/tratamento farmacológico , Adolescente , Adulto , Idoso , Criança , Pré-Escolar , Estudos de Coortes , Disceratose Congênita/genética , Disceratose Congênita/metabolismo , Feminino , Humanos , Lactente , Masculino , Pessoa de Meia-Idade , Estudos Prospectivos , Estudos Retrospectivos , Telômero/metabolismo , Adulto Jovem
19.
Biol Chem ; 395(6): 593-610, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24468621

RESUMO

Human dyskerin is an evolutively conserved protein that participates in diverse nuclear complexes: the H/ACA snoRNPs, that control ribosome biogenesis, RNA pseudouridylation, and stability of H/ACA snoRNAs; the scaRNPs, that control pseudouridylation of snRNAs; and the telomerase active holoenzyme, which safeguards telomere integrity. The biological importance of dyskerin is further outlined by the fact that its deficiency causes the X-linked dyskeratosis congenita disease, while its over-expression characterizes several types of cancers and has been proposed as prognostic marker. The role of dyskerin in telomere maintenance has widely been discussed, while its functions as H/ACA sno/scaRNP component has been so far mostly overlooked and represent the main goal of this review. Here we summarize how increasing evidence indicates that the snoRNA/microRNA pathways can be interlaced, and that dyskerin-dependent RNA pseudouridylation represents a flexible mechanism able to modulate RNA function in different ways, including modulation of splicing, change of mRNA coding properties, and selective regulation of IRES-dependent translation. We also propose a speculative model that suggests that the dynamics of pre-assembly and nuclear import of H/ACA RNPs are crucial regulatory steps that can be finely controlled in the cytoplasm in response to developmental, differentiative and stress stimuli.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Disceratose Congênita/metabolismo , Proteínas Nucleares/metabolismo , Ribonucleoproteínas Nucleolares Pequenas/metabolismo , Proteínas de Ciclo Celular/genética , Disceratose Congênita/genética , Humanos , Proteínas Nucleares/genética , Telômero
20.
RNA Biol ; 11(12): 1483-94, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25590339

RESUMO

Box H/ACA ribonucleoproteins (RNPs), each consisting of one unique guide RNA and 4 common core proteins, constitute a family of complex enzymes that catalyze, in an RNA-guided manner, the isomerization of uridines to pseudouridines (Ψs) in RNAs, a reaction known as pseudouridylation. Over the years, box H/ACA RNPs have been extensively studied revealing many important aspects of these RNA modifying machines. In this review, we focus on the composition, structure, and biogenesis of H/ACA RNPs. We explain the mechanism of how this enzyme family recognizes and specifies its target uridine in a substrate RNA. We discuss the substrates of box H/ACA RNPs, focusing on rRNA (rRNA) and spliceosomal small nuclear RNA (snRNA). We describe the modification product Ψ and its contribution to RNA function. Finally, we consider possible mechanisms of the bone marrow failure syndrome dyskeratosis congenita and of prostate and other cancers linked to mutations in H/ACA RNPs.


Assuntos
Disceratose Congênita/metabolismo , Neoplasias da Próstata/metabolismo , Pseudouridina/metabolismo , Processamento Pós-Transcricional do RNA , RNA Guia de Cinetoplastídeos/metabolismo , Uridina/metabolismo , Disceratose Congênita/genética , Disceratose Congênita/patologia , Humanos , Isomerismo , Masculino , Mutação , Conformação de Ácido Nucleico , Neoplasias da Próstata/genética , Neoplasias da Próstata/patologia , RNA Guia de Cinetoplastídeos/genética , RNA Ribossômico/genética , RNA Ribossômico/metabolismo , RNA Nuclear Pequeno/genética , RNA Nuclear Pequeno/metabolismo , RNA de Transferência Aminoácido-Específico/genética , RNA de Transferência Aminoácido-Específico/metabolismo , Ribonucleoproteínas Nucleares Pequenas/genética , Ribonucleoproteínas Nucleares Pequenas/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA