Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Hum Mol Genet ; 33(4): 318-332, 2024 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-37879098

RESUMEN

Dyskerin is a component of the human telomerase complex and is involved in stabilizing the human telomerase RNA (hTR). Many mutations in the DKC1 gene encoding dyskerin are found in X-linked dyskeratosis congenita (X-DC), a premature aging disorder and other related diseases. The C-terminal extension (CTE) of dyskerin contributes to its interaction with the molecular chaperone SHQ1 during the early stage of telomerase biogenesis. Disease mutations in this region were proposed to disrupt dyskerin-SHQ1 interaction and destabilize dyskerin, reducing hTR levels indirectly. However, biochemical evidence supporting this hypothesis is still lacking. In addition, the effects of many CTE disease mutations on hTR have not been examined. In this study, we tested eight dyskerin CTE variants and showed that they failed to maintain hTR levels. These mutants showed slightly reduced but not abolished interaction with SHQ1, and caused defective binding to hTR. Deletion of the CTE further reduced binding to hTR, and perturbed localization of dyskerin to the Cajal bodies and the nucleolus, and the interaction with TCAB1 as well as GAR1. Our findings suggest impaired dyskerin-hTR interaction in cells as a previously overlooked mechanism through which dyskerin CTE mutations cause X-DC and related telomere syndromes.


Asunto(s)
Disqueratosis Congénita , Telomerasa , Humanos , Telomerasa/genética , Disqueratosis Congénita/genética , Telómero/genética , Telómero/metabolismo , Proteínas Nucleares/metabolismo , ARN/genética , ARN/metabolismo , Mutación , Proteínas de Unión al ARN/genética , Proteínas de Ciclo Celular/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética
2.
J Biol Chem ; 299(3): 102916, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36649908

RESUMEN

In the majority of human cancer cells, cellular immortalization depends on the maintenance of telomere length by telomerase. An essential step required for telomerase function is its recruitment to telomeres, which is regulated by the interaction of the telomere protein, TPP1, with the telomerase essential N-terminal (TEN) domain of the human telomerase reverse transcriptase, hTERT. We previously reported that the hTERT 'insertion in fingers domain' (IFD) recruits telomerase to telomeres in a TPP1-dependent manner. Here, we use hTERT truncations and the IFD domain containing mutations in conserved residues or premature aging disease-associated mutations to map the interactions between the IFD and TPP1. We find that the hTERT-IFD domain can interact with TPP1. However, deletion of the IFD motif in hTERT lacking the N-terminus and the C-terminal extension does not abolish interaction with TPP1, suggesting the IFD is not essential for hTERT interaction with TPP1. Several conserved residues in the central IFD-TRAP region that we reported regulate telomerase recruitment to telomeres, and cell immortalization compromise interaction of the hTERT-IFD domain with TPP1 when mutated. Using a similar approach, we find that the IFD domain interacts with the TEN domain but is not essential for intramolecular hTERT interactions with the TEN domain. IFD-TEN interactions are not disrupted by multiple amino acid changes in the IFD or TEN, thus highlighting a complex regulation of IFD-TEN interactions as suggested by recent cryo-EM structures of human telomerase.


Asunto(s)
Complejo Shelterina , Telomerasa , Proteínas de Unión a Telómeros , Humanos , Línea Celular , Mutación , Telomerasa/química , Telomerasa/metabolismo , Telómero/química , Telómero/metabolismo , Proteínas de Unión a Telómeros/química , Proteínas de Unión a Telómeros/metabolismo , Complejo Shelterina/química , Complejo Shelterina/metabolismo
3.
RNA ; 27(12): 1441-1458, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34556550

RESUMEN

Dyskerin and its homologs are ancient and conserved enzymes that catalyze the most common post-transcriptional modification found in cells, pseudouridylation. The resulting pseudouridines provide stability to RNA molecules and regulate ribosome biogenesis and splicing events. Dyskerin does not act independently-it is the core component of a protein heterotetramer, which associates with RNAs that contain the H/ACA motif. The variety of H/ACA RNAs that guide the function of this ribonucleoprotein (RNP) complex highlights the diversity of cellular processes in which dyskerin participates. When associated with small nucleolar (sno) RNAs, it regulates ribosomal (r) RNAs and ribosome biogenesis. By interacting with small Cajal body (sca) RNAs, it targets small nuclear (sn) RNAs to regulate pre-mRNA splicing. As a component of the telomerase holoenzyme, dyskerin binds to the telomerase RNA to modulate telomere maintenance. In a disease context, dyskerin malfunction can result in multiple detrimental phenotypes. Mutations in DKC1, the gene that encodes dyskerin, cause the premature aging syndrome X-linked dyskeratosis congenita (X-DC), a still incurable disorder that typically leads to bone marrow failure. In this review, we present the classical and most recent findings on this essential protein, discussing the evolutionary, structural, and functional aspects of dyskerin and the H/ACA RNP. The latest research underscores the role that dyskerin plays in the regulation of gene expression, translation efficiency, and telomere maintenance, along with the impacts that defective dyskerin has on aging, cell proliferation, haematopoietic potential, and cancer.


Asunto(s)
Empalme Alternativo , Proteínas de Ciclo Celular/metabolismo , Transferasas Intramoleculares/metabolismo , Mutación , Proteínas Nucleares/metabolismo , Procesamiento Postranscripcional del ARN , Ribosomas/metabolismo , Telómero/fisiología , Proteínas de Ciclo Celular/genética , Disqueratosis Congénita , Humanos , Transferasas Intramoleculares/genética , Proteínas Nucleares/genética
4.
Nucleic Acids Res ; 49(20): 11690-11707, 2021 11 18.
Artículo en Inglés | MEDLINE | ID: mdl-34725692

RESUMEN

Loss of telomeric DNA leads to telomere uncapping, which triggers a persistent, p53-centric DNA damage response that sustains a stable senescence-associated proliferation arrest. Here, we show that in normal cells telomere uncapping triggers a focal telomeric DNA damage response accompanied by a transient cell cycle arrest. Subsequent cell division with dysfunctional telomeres resulted in sporadic telomeric sister chromatid fusions that gave rise to next-mitosis genome instability, including non-telomeric DNA lesions responsible for a stable, p53-mediated, senescence-associated proliferation arrest. Unexpectedly, the blocking of Rad51/RPA-mediated homologous recombination, but not non-homologous end joining (NHEJ), prevented senescence despite multiple dysfunctional telomeres. When cells approached natural replicative senescence, interphase senescent cells displayed genome instability, whereas near-senescent cells that underwent mitosis despite the presence of uncapped telomeres did not. This suggests that these near-senescent cells had not yet acquired irreversible telomeric fusions. We propose a new model for telomere-initiated senescence where tolerance of telomere uncapping eventually results in irreversible non-telomeric DNA lesions leading to stable senescence. Paradoxically, our work reveals that senescence-associated tumor suppression from telomere shortening requires irreversible genome instability at the single-cell level, which suggests that interventions to repair telomeres in the pre-senescent state could prevent senescence and genome instability.


Asunto(s)
Inestabilidad Genómica , Recombinación Homóloga , Acortamiento del Telómero/genética , Células Cultivadas , Daño del ADN , Reparación del ADN por Unión de Extremidades , Humanos , Recombinasa Rad51/metabolismo
5.
RNA Biol ; 18(3): 305-315, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-32813614

RESUMEN

Maintenance of telomeres is essential for genome integrity and replicative capacity in eukaryotic cells. Telomerase, the ribonucleoprotein complex that catalyses telomere synthesis is minimally composed of a reverse transcriptase and an RNA component. The sequence and structural domains of human telomerase RNA (hTR) have been extensively characterized, while the regulation of hTR transcription, maturation, and localization, is not fully understood. Here, we provide an up-to-date review of hTR, with an emphasis on current breakthroughs uncovering the mechanisms of hTR maturation and localization.


Asunto(s)
Regulación de la Expresión Génica , ARN/genética , ARN/metabolismo , Telomerasa/genética , Telomerasa/metabolismo , Humanos , Metilación , Mutación , Conformación de Ácido Nucleico , Unión Proteica , ARN/química , Caperuzas de ARN , Procesamiento Postranscripcional del ARN , Estabilidad del ARN , Transporte de ARN , Relación Estructura-Actividad , Telomerasa/química , Transcripción Genética
6.
Nucleic Acids Res ; 47(10): 5368-5380, 2019 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-30931479

RESUMEN

The telomerase holoenzyme responsible for maintaining telomeres in vertebrates requires many components in vivo, including dyskerin. Dyskerin binds and regulates the accumulation of the human telomerase RNA, hTR, as well as other non-coding RNAs that share the conserved H/ACA box motif. The precise mechanism by which dyskerin controls hTR levels is unknown, but is evidenced by defective hTR accumulation caused by substitutions in dyskerin, that are observed in the X-linked telomere biology disorder dyskeratosis congenita (X-DC). To understand the role of dyskerin in hTR accumulation, we analyzed X-DC substitutions K39E and K43E in the poorly characterized dyskerin N-terminus, and A353V within the canonical RNA binding domain (the PUA). These variants exhibited impaired binding to hTR and polyadenylated hTR species, while interactions with other H/ACA RNAs appear largely unperturbed by the N-terminal substitutions. hTR accumulation and telomerase activity defects of dyskerin-deficient cells were rescued by wildtype dyskerin but not the variants. hTR 3' extended or polyadenylated species did not accumulate, suggesting hTR precursor degradation occurs upstream of mature complex assembly in the absence of dyskerin binding. Our findings demonstrate that the dyskerin-hTR interaction mediated by PUA and N-terminal residues of dyskerin is crucial to prevent unchecked hTR degradation.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Proteínas Nucleares/metabolismo , Estabilidad del ARN , ARN/genética , Telomerasa/metabolismo , Secuencias de Aminoácidos , Proteínas de Ciclo Celular/genética , Núcleo Celular/metabolismo , Citosol/metabolismo , Disqueratosis Congénita/genética , Células HEK293 , Humanos , Mutación , Proteínas Nucleares/genética , Dominios Proteicos , ARN Interferente Pequeño/metabolismo , Proteínas de Unión al ARN/metabolismo , Telomerasa/genética , Telómero/metabolismo
7.
J Biol Chem ; 291(16): 8374-86, 2016 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-26887940

RESUMEN

Normal human stem cells rely on low levels of active telomerase to sustain their high replicative requirements. Deficiency in telomere maintenance mechanisms leads to the development of premature aging diseases, such as dyskeratosis congenita and aplastic anemia. Mutations in the unique "insertion in fingers domain" (IFD) in the human telomerase reverse transcriptase catalytic subunit (hTERT) have previously been identified and shown to be associated with dyskeratosis congenita and aplastic anemia. However, little is known about the molecular mechanisms impacted by these IFD mutations. We performed comparative functional analyses of disease-associated IFD variants at the molecular and cellular levels. We report that hTERT-P721R- and hTERT-R811C-expressing cells exhibited growth defects likely due to impaired TPP1-mediated recruitment of these variant enzymes to telomeres. We showed that activity and processivity of hTERT-T726M failed to be stimulated by TPP1-POT1 overexpression and that dGTP usage by this variant was less efficient compared with the wild-type enzyme. hTERT-P785L-expressing cells did not show growth defects, and this variant likely confers cell survival through increased DNA synthesis and robust activity stimulation by TPP1-POT1. Altogether, our data suggest that multiple mechanisms contribute to cell growth defects conferred by the IFD variants.


Asunto(s)
Envejecimiento Prematuro/enzimología , Mutación Missense , Telomerasa/metabolismo , Envejecimiento Prematuro/genética , Envejecimiento Prematuro/patología , Sustitución de Aminoácidos , Aminopeptidasas/genética , Aminopeptidasas/metabolismo , Dipeptidil-Peptidasas y Tripeptidil-Peptidasas/genética , Dipeptidil-Peptidasas y Tripeptidil-Peptidasas/metabolismo , Células HEK293 , Células HeLa , Humanos , Estructura Terciaria de Proteína , Serina Proteasas/genética , Serina Proteasas/metabolismo , Complejo Shelterina , Telomerasa/genética , Proteínas de Unión a Telómeros/genética , Proteínas de Unión a Telómeros/metabolismo
8.
Biol Cell ; 108(4): 96-112, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26787169

RESUMEN

BACKGROUND INFORMATION: Naturally occurring telomerase reverse transcriptase (TERT) isoforms may regulate telomerase activity, and possibly function independently of telomeres to modulate embryonic stem (ES) cell self-renewal and differentiation. RESULTS: We report the characterisation of two novel mouse TERT (mTERT) splice variants, Ins-i1[1-102] (Insi1 for short) and Del-e12[1-40] (Dele12 for short) that have not been previously described. Insi1 represents an in-frame insertion of nucleotides 1-102 from intron 1, encoding a 34 amino acid insertion at amino acid 73. Based on known functions of this region in human and Tetrahymena TERTs, the insertion interrupts the RNA interaction domain 1 implicated in low-affinity RNA binding and the telomerase essential N-terminal domain implicated in DNA substrate interactions. Dele12 contains a 40 nucleotide deletion of exon 12 which generates a premature stop codon, and possible protein lacking the C-terminus. We found Insi1 expressed in adult mouse brain and kidney and Dele12 expressed in adult mouse ovary. Dele12 was inactive in vitro and in mTERT(-/-) ES cells and Insi1 retained 26-48% of telomerase activity reconstituted by wild-type mTERT in vitro and in mTERT(-/-) ES cells. The Insi1 variant exhibited reduced DNA substrate binding in vitro and both variants exhibited a reduction in binding the telomerase RNA, mTR, when expressed in mTERT(-/-) ES cells. Stable expression of Dele12 in the mouse fibroblast CB17 cell line inhibited telomerase activity and slowed cell growth, suggesting a potential dominant-negative effect. Levels of signal-free ends, representing short telomeres, and end-to-end fusions were higher in mTERT(-/-) ES cells expressing mTERT-Insi1 and mTERT-Dele12, compared with levels observed in mTERT(-/-) ES cells expressing wild-type mTERT. In addition, in mTERT(-/-) cells expressing mTERT-Insi1, we observed chromosomes that were products of repeated breakage-bridge-fusion cycles and other telomere dysfunction-related aberrations. CONCLUSION AND SIGNIFICANCE: An intact mTERT N-terminus which contributes to mTR binding, DNA binding and telomerase activity is necessary for elongation of short telomeres and the maintenance of functional telomeres. It is reasonable to speculate that relative levels of mTERT-Insi1 may regulate telomere function in specific tissues.


Asunto(s)
Telomerasa/genética , Telomerasa/metabolismo , Telómero/metabolismo , Empalme Alternativo , Animales , Línea Celular , Ratones , Biosíntesis de Proteínas , Isoformas de Proteínas/análisis , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , ARN/metabolismo , Telomerasa/análisis , Transcripción Genética
9.
Hum Mol Genet ; 22(17): 3498-507, 2013 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-23660516

RESUMEN

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.


Asunto(s)
Proteínas de Ciclo Celular/genética , Disqueratosis Congénita/genética , Proteínas Nucleares/genética , ARN/metabolismo , Telomerasa/metabolismo , Homeostasis del Telómero , Telómero/metabolismo , Secuencia de Aminoácidos , Disqueratosis Congénita/metabolismo , Técnicas de Silenciamiento del Gen , Células HEK293 , Humanos , Datos de Secuencia Molecular , Mutación , Filogenia , Estabilidad Proteica , ARN/genética , Alineación de Secuencia , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/metabolismo , Sumoilación , Telomerasa/genética
10.
J Cell Sci ; 126(Pt 2): 676-87, 2013 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-23178942

RESUMEN

Telomerase is a ribonucleoprotein consisting of a catalytic subunit, the telomerase reverse transcriptase (TERT), and an integrally associated RNA that contains a template for the synthesis of short repetitive G-rich DNA sequences at the ends of telomeres. Telomerase can repetitively reverse transcribe its short RNA template, acting processively to add multiple telomeric repeats onto the same DNA substrate. The contribution of enzyme processivity to telomere length regulation in human cells is not well characterized. In cancer cells, under homeostatic telomere length-maintenance conditions, telomerase acts processively, whereas under nonequilibrium conditions, telomerase acts distributively on the shortest telomeres. To investigate the role of increased telomerase processivity on telomere length regulation in human cells with limited lifespan that are dependent on human TERT for lifespan extension and immortalization, we mutated the leucine at position 866 in the reverse transcriptase C motif of human TERT to a tyrosine (L866Y), which is the amino acid found at the equivalent position in HIV-1 reverse transcriptase. We report that, similar to the previously reported gain-of-function Tetrahymena telomerase mutant (L813Y), the human telomerase variant displays increased processivity. Human TERT-L866Y, like wild-type human TERT, can immortalize and extend the lifespan of limited-lifespan cells. Moreover, cells expressing human TERT-L866Y display heterogenous telomere lengths, telomere elongation, multiple telomeric signals indicative of fragile sites and replicative stress, and an increase in short telomeres, which is accompanied by telomere trimming events. Our results suggest that telomere length and homeostasis in human cells may be regulated by telomerase enzyme processivity.


Asunto(s)
Telomerasa/metabolismo , Homeostasis del Telómero , Animales , Células HEK293 , Homeostasis , Humanos , Conejos , Telomerasa/genética , Telómero/metabolismo
11.
BMC Cancer ; 15: 621, 2015 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-26345285

RESUMEN

BACKGROUND: The identification of oncogenic driver mutations has largely relied on the assumption that genes that exhibit more mutations than expected by chance are more likely to play an active role in tumorigenesis. Major cancer sequencing initiatives have therefore focused on recurrent mutations that are more likely to be drivers. However, in specific genetic contexts, low frequency mutations may also be capable of participating in oncogenic processes. Reliable strategies for identifying these rare or even patient-specific (private) mutations are needed in order to elucidate more personalized approaches to cancer diagnosis and treatment. METHODS: Here we performed whole-exome sequencing on three cases of childhood pre-B acute lymphoblastic leukemia (cALL), representing three cytogenetically-defined subgroups (high hyperdiploid, t(12;21) translocation, and cytogenetically normal). We applied a data reduction strategy to identify both common and rare/private somatic events with high functional potential. Top-ranked candidate mutations were subsequently validated at high sequencing depth on an independent platform and in vitro expression assays were performed to evaluate the impact of identified mutations on cell growth and survival. RESULTS: We identified 6 putatively damaging non-synonymous somatic mutations among the three cALL patients. Three of these mutations were well-characterized common cALL mutations involved in constitutive activation of the mitogen-activated protein kinase pathway (FLT3 p.D835Y, NRAS p.G13D, BRAF p.G466A). The remaining three patient-specific mutations (ACD p.G223V, DOT1L p.V114F, HCFC1 p.Y103H) were novel mutations previously undescribed in public cancer databases. Cytotoxicity assays demonstrated a protective effect of the ACD p.G223V mutation against apoptosis in leukemia cells. ACD plays a key role in protecting telomeres and recruiting telomerase. Using a telomere restriction fragment assay, we also showed that this novel mutation in ACD leads to increased telomere length in leukemia cells. CONCLUSION: This study identified ACD as a novel gene involved in cALL and points to a functional role for ACD in enhancing leukemia cell survival. These results highlight the importance of rare/private somatic mutations in understanding cALL etiology, even within well-characterized molecular subgroups.


Asunto(s)
Apoptosis/genética , Análisis Mutacional de ADN/métodos , Mutación , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Homeostasis del Telómero/genética , Proteínas de Unión a Telómeros/genética , Niño , Preescolar , Exoma/genética , Femenino , Humanos , Masculino , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Complejo Shelterina
12.
RNA Biol ; 12(10): 1078-82, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26291128

RESUMEN

The key step of carcinogenesis is the malignant transformation which is fundamentally a telomere biology dysfunction permitting cells to bypass the Hayflick limit and to divide indefinitely and uncontrollably. Thus all partners and structures involved in normal and abnormal telomere maintenance, protection and lengthening can be considered as potential anti-cancer therapeutic targets. In this Point of View we discuss, highlight and provide new perspectives from the current knowledge and understanding to position the different aspects of telomere biology and dysfunction as diagnostic, preventive and curative tools in the field of cancer.


Asunto(s)
Transformación Celular Neoplásica/genética , Neoplasias/genética , Telómero/genética , Humanos , Neoplasias/diagnóstico , Neoplasias/patología , Neoplasias/terapia , Telomerasa/genética , Telómero/patología
13.
Nat Struct Mol Biol ; 30(7): 878-890, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37400652

RESUMEN

Telomerase is a special reverse transcriptase ribonucleoprotein dedicated to the synthesis of telomere repeats that protect chromosome ends. Among reverse transcriptases, telomerase is unique in using a stably associated RNA with an embedded template to synthesize a specified sequence. Moreover, it is capable of iteratively copying the same template region (repeat addition processivity) through multiple rounds of RNA-DNA unpairing and reannealing, that is, the translocation reaction. Biochemical analyses of telomerase over the past 3 decades in protozoa, fungi and mammals have identified structural elements that underpin telomerase mechanisms and have led to models that account for the special attributes of telomerase. Notably, these findings and models can now be interpreted and adjudicated through recent cryo-EM structures of Tetrahymena and human telomerase holoenzyme complexes in association with substrates and regulatory proteins. Collectively, these structures reveal the intricate protein-nucleic acid interactions that potentiate telomerase's unique translocation reaction and clarify how this enzyme reconfigures the basic reverse transcriptase scaffold to craft a polymerase dedicated to the synthesis of telomere DNA. Among the many new insights is the resolution of the telomerase 'anchor site' proposed more than 3 decades ago. The structures also highlight the nearly universal conservation of a protein-protein interface between an oligonucleotide/oligosaccharide-binding (OB)-fold regulatory protein and the telomerase catalytic subunit, which enables spatial and temporal regulation of telomerase function in vivo. In this Review, we discuss key features of the structures in combination with relevant functional analyses. We also examine conserved and divergent aspects of telomerase mechanisms as gleaned from studies in different model organisms.


Asunto(s)
Ácidos Nucleicos , Telomerasa , Animales , Humanos , Telomerasa/química , Telómero/metabolismo , ADN Polimerasa Dirigida por ARN/genética , ADN Polimerasa Dirigida por ARN/metabolismo , ARN/metabolismo , ADN , Mamíferos/genética
14.
J Cell Sci ; 123(Pt 10): 1693-704, 2010 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-20427319

RESUMEN

Telomerase synthesizes telomeric sequences and is minimally composed of a reverse transcriptase (RT) known as TERT and an RNA known as TR. We reconstituted heterologous mouse (m) and human (h) TERT-TR complexes and chimeric mTERT-hTERT-hTR complexes in vitro and in immortalized human alternative lengthening of telomere (ALT) cells. Our data suggest that species-specific determinants of activity, processivity and telomere function map not only to the TR but also to the TERT component. The presence of hTERT-hTR, but not heterologous TERT-TR complexes or chimeric mTERT-hTERT-hTR complexes, significantly reduced the percentage of chromosomes without telomeric signals in ALT cells. Moreover, heterologous and chimeric complexes were defective in recruitment to telomeres. Our results suggest a requirement for several hTERT domains and interaction with multiple proteins for proper recruitment of telomerase to the shortest telomeres in human ALT cells. Late-passage mTERT(-/-) mouse embryonic stem (ES) cells ectopically expressing hTERT or mTERT harboured fewer chromosome ends without telomeric signals and end-to-end fusions than typically observed in late-passage mTERT(-/-) ES cells. The ability of hTERT to function at mouse telomeres and the inability of mTERT to function at human telomeres suggest that mechanisms regulating the recruitment and activity of hTERT at mouse telomeres might be less stringent than the mechanisms regulating mTERT at human telomeres.


Asunto(s)
ADN/biosíntesis , Fibroblastos/metabolismo , Telomerasa/metabolismo , Telómero/metabolismo , Animales , Línea Celular Tumoral , Clonación Molecular , Fibroblastos/patología , Regulación Enzimológica de la Expresión Génica , Humanos , Ratones , Multimerización de Proteína , Especificidad de la Especie , Especificidad por Sustrato , Telomerasa/genética , Telómero/genética , Transgenes/genética
15.
Cell Mol Biol Lett ; 17(4): 598-615, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22941205

RESUMEN

The minimal vertebrate telomerase enzyme is composed of a protein component (telomerase reverse transcriptase, TERT) and an RNA component (telomerase RNA, TR). Expression of these two subunits is sufficient to reconstitute telomerase activity in vitro, while the formation of a holoenzyme comprising telomerase-associated proteins is necessary for proper telomere length maintenance. Previous reports demonstrated the high processivity of the human telomerase complex and the interspecies compatibility of human TERT (hTERT). In this study, we tested the function of the only known viral telomerase RNA subunit (vTR) in association with human telomerase, both in a cell-free system and in human cells. When vTR is assembled with hTERT in a cell-free environment, it is able to interact with hTERT and to reconstitute telomerase activity. However, in human cells, vTR does not reconstitute telomerase activity and could not be detected in the human telomerase complex, suggesting that vTR is not able to interact properly with the proteins constituting the human telomerase holoenzyme.


Asunto(s)
ARN/metabolismo , Telomerasa/metabolismo , Secuencia de Bases , Dominio Catalítico , Sistema Libre de Células , Clonación Molecular , Herpesvirus Gallináceo 2/enzimología , Holoenzimas/química , Holoenzimas/genética , Holoenzimas/metabolismo , Humanos , Inmunoprecipitación , ARN/genética , Telomerasa/genética
16.
J Clin Invest ; 132(8)2022 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-35426375

RESUMEN

Mice with experimental nerve damage can display long­lasting neuropathic pain behavior. We show here that 4 months and later after nerve injury, male but not female mice displayed telomere length (TL) reduction and p53­mediated cellular senescence in the spinal cord, resulting in maintenance of pain and associated with decreased lifespan. Nerve injury increased the number of p53­positive spinal cord neurons, astrocytes, and microglia, but only in microglia was the increase male­specific, matching a robust sex specificity of TL reduction in this cell type, which has been previously implicated in male­specific pain processing. Pain hypersensitivity was reversed by repeated intrathecal administration of a p53­specific senolytic peptide, only in male mice and only many months after injury. Analysis of UK Biobank data revealed sex-specific relevance of this pathway in humans, featuring male­specific genetic association of the human p53 locus (TP53) with chronic pain and a male-specific effect of chronic pain on mortality. Our findings demonstrate the existence of a biological mechanism maintaining pain behavior, at least in males, occurring much later than the time span of virtually all extant preclinical studies.


Asunto(s)
Dolor Crónico , Neuralgia , Animales , Senescencia Celular , Dolor Crónico/genética , Dolor Crónico/metabolismo , Femenino , Hiperalgesia/metabolismo , Masculino , Ratones , Microglía/metabolismo , Neuralgia/genética , Neuralgia/metabolismo , Médula Espinal/metabolismo , Telómero/genética , Telómero/metabolismo , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
17.
DNA Repair (Amst) ; 100: 103055, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33581499

RESUMEN

The maintenance of telomeres, which are specialized stretches of DNA found at the ends of linear chromosomes, is a crucial step for the immortalization of cancer cells. Approximately 10-15 % of cancer cells use a homologous recombination-based mechanism known as the Alternative Lengthening of Telomeres (ALT) pathway to maintain their telomeres. Telomeres in general pose a challenge to DNA replication owing to their repetitive nature and potential for forming secondary structures. Telomeres in ALT+ cells especially are subject to elevated levels of replication stress compared to telomeres that are maintained by the enzyme telomerase, in part due to the incorporation of telomeric variant repeats at ALT+ telomeres, their on average longer lengths, and their modified chromatin states. Many DNA metabolic strategies exist to counter replication stress and to protect stalled replication forks. The role of proliferating cell nuclear antigen (PCNA) as a platform for recruiting protein partners that participate in several of these DNA replication and repair pathways has been well-documented. We propose that many of these pathways may be active at ALT+ telomeres, either to facilitate DNA replication, to manage replication stress, or during telomere extension. Here, we summarize recent evidence detailing the role of PCNA in pathways including DNA secondary structure resolution, DNA damage bypass, replication fork restart, and DNA damage synthesis. We propose that an examination of PCNA and its post-translational modifications (PTMs) may offer a unique lens by which we might gain insight into the DNA metabolic landscape that is distinctively present at ALT+ telomeres.


Asunto(s)
Antígeno Nuclear de Célula en Proliferación/metabolismo , Reparación del ADN por Recombinación , Homeostasis del Telómero , ADN/metabolismo , Replicación del ADN , Eucariontes/genética , Eucariontes/metabolismo , Humanos , Neoplasias/genética , Neoplasias/metabolismo
18.
JCI Insight ; 6(4)2021 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-33470989

RESUMEN

Triple-negative breast cancers (TNBCs) lack effective targeted therapies, and cytotoxic chemotherapies remain the standard of care for this subtype. Owing to their increased genomic instability, poly (ADP-ribose) polymerase (PARP) inhibitors (PARPi) are being tested against TNBCs. In particular, clinical trials are now interrogating the efficacy of PARPi combined with chemotherapies. Intriguingly, while response rates are low, cohort of patients do respond to PARPi in combination with chemotherapies. Moreover, recent studies suggest that an increase in levels of ROS may sensitize cells to PARPi. This represents a therapeutic opportunity, as several chemotherapies, including doxorubicin, function in part by producing ROS. We previously demonstrated that the p66ShcA adaptor protein is variably expressed in TNBCs. We now show that, in response to therapy-induced stress, p66ShcA stimulated ROS production, which, in turn, potentiated the synergy of PARPi in combination with doxorubicin in TNBCs. This p66ShcA-induced sensitivity relied on the accumulation of oxidative damage in TNBCs, rather than genomic instability, to potentiate cell death. These findings suggest that increasing the expression of p66ShcA protein levels in TNBCs represents a rational approach to bolster the synergy between PARPi and doxorubicin.


Asunto(s)
Antineoplásicos/farmacología , Poli(ADP-Ribosa) Polimerasa-1/efectos de los fármacos , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Neoplasias de la Mama Triple Negativas/genética , Apoptosis , Proteína BRCA1/genética , Proteína BRCA2/genética , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Sistemas CRISPR-Cas , Línea Celular Tumoral , Supervivencia Celular , Daño del ADN , Inestabilidad Genómica , Humanos , Células MCF-7 , Inhibidores de Poli(ADP-Ribosa) Polimerasas/uso terapéutico , Proteína Transformadora 1 que Contiene Dominios de Homología 2 de Src , Ensayos Antitumor por Modelo de Xenoinjerto
19.
J Cell Biol ; 159(6): 957-69, 2002 Dec 23.
Artículo en Inglés | MEDLINE | ID: mdl-12486110

RESUMEN

The nuclear structures that contain symmetrical dimethylated arginine (sDMA)-modified proteins and the role of this posttranslational modification is unknown. Here we report that the Cajal body is a major epitope in HeLa cells for an sDMA-specific antibody and that coilin is an sDMA-containing protein as analyzed by using the sDMA-specific antibody and matrix-assisted laser desorption ionization time of flight mass spectrometry. The methylation inhibitor 5'-deoxy-5'-methylthioadenosine reduces the levels of coilin methylation and causes the appearance of SMN-positive gems. In cells devoid of Cajal bodies, such as primary fibroblasts, sDMA-containing proteins concentrated in speckles. Cells from a patient with spinal muscular atrophy, containing low levels of the methyl-binding protein SMN, localized sDMA-containing proteins in the nucleoplasm as a discrete granular pattern. Splicing reactions are efficiently inhibited by using the sDMA-specific antibody or by using hypomethylated nuclear extracts, showing that active spliceosomes contain sDMA polypeptides and suggesting that arginine methylation is important for efficient pre-mRNA splicing. Our findings support a model in which arginine methylation is important for the localization of coilin and SMN in Cajal bodies.


Asunto(s)
Arginina/metabolismo , Cuerpos Enrollados/metabolismo , Anticuerpos/metabolismo , Arginina/análogos & derivados , Núcleo Celular/metabolismo , ADN/metabolismo , Relación Dosis-Respuesta a Droga , Regulación hacia Abajo , Electroforesis en Gel de Poliacrilamida , Ensayo de Inmunoadsorción Enzimática , Fibroblastos/metabolismo , Técnica del Anticuerpo Fluorescente Indirecta , Células HeLa , Humanos , Espectrometría de Masas , Metilación , Microscopía Fluorescente , Atrofia Muscular Espinal/metabolismo , Pruebas de Precipitina , Empalme del ARN , ARN Mensajero/metabolismo , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Transfección
20.
J Am Chem Soc ; 130(31): 10040-1, 2008 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-18616250

RESUMEN

In this contribution, we report that a self-assembled platinum molecular square [Pt(en)(4,4'-dipyridyl)]4 can act as an efficient G-quadruplex binder and telomerase inhibitor. Molecular modeling studies show that the square arrangement of the four bipyridyl ligands, the highly electropositive nature of the overall complex, as well as hydrogen bonding interactions between the ethylenediamine ligands and phosphates of the DNA backbone all contribute to the observed strong binding affinity to the G-quadruplex. Through thermal denaturation studies with duplex and quadruplex FRET probes and enzymatic assays, we demonstrate that this platinum square strongly binds to G-quadruplexes and can act as an inhibitor of telomerase. This study thus shows the potential of supramolecular self-assembly to readily generate scaffolds of unique geometries for effective targeting of G-quadruplexes and for the ultimate development of selective antitumor therapies.


Asunto(s)
Antineoplásicos/química , G-Cuádruplex , Compuestos Organoplatinos/química , Telomerasa/antagonistas & inhibidores , 2,2'-Dipiridil , Sitios de Unión , Inhibidores Enzimáticos/química , Modelos Moleculares , Estructura Molecular , Desnaturalización de Ácido Nucleico/efectos de los fármacos , Compuestos Organoplatinos/metabolismo , Compuestos Organoplatinos/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA