Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Biochim Biophys Acta Mol Basis Dis ; 1870(4): 167107, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38430974

RESUMEN

Hereditary renal cell carcinoma (RCC) is caused by germline mutations in a subset of genes, including VHL, MET, FLCN, and FH. However, many familial RCC cases do not harbor mutations in the known predisposition genes. Using Whole Exome Sequencing, we identified two germline missense variants in the DCLRE1B/Apollo gene (ApolloN246I and ApolloY273H) in two unrelated families with several RCC cases. Apollo encodes an exonuclease involved in DNA Damage Response and Repair (DDRR) and telomere integrity. We characterized these two functions in the human renal epithelial cell line HKC8. The decrease or inhibition of Apollo expression sensitizes these cells to DNA interstrand crosslink damage (ICLs). HKC8 Apollo-/- cells appear defective in the DDRR and present an accumulation of telomere damage. Wild-type and mutated Apollo forms could interact with TRF2, a shelterin protein involved in telomere protection. However, only ApolloWT can rescue the telomere damage in HKC8 Apollo-/- cells. Our results strongly suggest that ApolloN246I and ApolloY273H are loss-of-function mutants that cause impaired telomere integrity and could lead to genomic instability. Altogether, our results suggest that mutations in Apollo could induce renal oncogenesis.


Asunto(s)
Carcinoma de Células Renales , Humanos , Carcinoma de Células Renales/genética , Mutación de Línea Germinal , Telómero/genética , Daño del ADN , Reparación del ADN/genética , Exodesoxirribonucleasas/genética
2.
Hum Mol Genet ; 33(7): 612-623, 2024 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-38176734

RESUMEN

Telomeres are nucleoprotein structures that protect the chromosome ends from degradation and fusion. Telomerase is a ribonucleoprotein complex essential to maintain the length of telomeres. Germline defects that lead to short and/or dysfunctional telomeres cause telomere biology disorders (TBDs), a group of rare and heterogeneous Mendelian diseases including pulmonary fibrosis, dyskeratosis congenita, and Høyeraal-Hreidarsson syndrome. TPP1, a telomeric factor encoded by the gene ACD, recruits telomerase at telomere and stimulates its activity via its TEL-patch domain that directly interacts with TERT, the catalytic subunit of telomerase. TBDs due to TPP1 deficiency have been reported only in 11 individuals. We here report four unrelated individuals with a wide spectrum of TBD manifestations carrying either heterozygous or homozygous ACD variants consisting in the recurrent and previously described in-frame deletion of K170 (K170∆) and three novel missense mutations G179D, L184R, and E215V. Structural and functional analyses demonstrated that the four variants affect the TEL-patch domain of TPP1 and impair telomerase activity. In addition, we identified in the ACD gene several motifs associated with small deletion hotspots that could explain the recurrence of the K170∆ mutation. Finally, we detected in a subset of blood cells from one patient, a somatic TERT promoter-activating mutation that likely provides a selective advantage over non-modified cells, a phenomenon known as indirect somatic genetic rescue. Together, our results broaden the genetic and clinical spectrum of TPP1 deficiency and specify new residues in the TEL-patch domain that are crucial for length maintenance and stability of human telomeres in vivo.


Asunto(s)
Complejo Shelterina , Telomerasa , Proteínas de Unión a Telómeros , Humanos , Biología , Mutación , Complejo Shelterina/genética , Telomerasa/genética , Telómero/genética , Telómero/metabolismo , Proteínas de Unión a Telómeros/genética , Proteínas de Unión a Telómeros/metabolismo
4.
Blood ; 139(16): 2427-2440, 2022 04 21.
Artículo en Inglés | MEDLINE | ID: mdl-35007328

RESUMEN

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.


Asunto(s)
Disqueratosis Congénita , Discapacidad Intelectual , Microcefalia , Disqueratosis Congénita/genética , Disqueratosis Congénita/metabolismo , Retardo del Crecimiento Fetal , Humanos , Discapacidad Intelectual/genética , Microcefalia/genética , Microcefalia/metabolismo , Mutación , Telómero/genética , Telómero/metabolismo
5.
Nat Commun ; 12(1): 5044, 2021 08 19.
Artículo en Inglés | MEDLINE | ID: mdl-34413298

RESUMEN

Indirect somatic genetic rescue (SGR) of a germline mutation is thought to be rare in inherited Mendelian disorders. Here, we establish that acquired mutations in the EIF6 gene are a frequent mechanism of SGR in Shwachman-Diamond syndrome (SDS), a leukemia predisposition disorder caused by a germline defect in ribosome assembly. Biallelic mutations in the SBDS or EFL1 genes in SDS impair release of the anti-association factor eIF6 from the 60S ribosomal subunit, a key step in the translational activation of ribosomes. Here, we identify diverse mosaic somatic genetic events (point mutations, interstitial deletion, reciprocal chromosomal translocation) in SDS hematopoietic cells that reduce eIF6 expression or disrupt its interaction with the 60S subunit, thereby conferring a selective advantage over non-modified cells. SDS-related somatic EIF6 missense mutations that reduce eIF6 dosage or eIF6 binding to the 60S subunit suppress the defects in ribosome assembly and protein synthesis across multiple SBDS-deficient species including yeast, Dictyostelium and Drosophila. Our data suggest that SGR is a universal phenomenon that may influence the clinical evolution of diverse Mendelian disorders and support eIF6 suppressor mimics as a therapeutic strategy in SDS.


Asunto(s)
Mutación , Subunidades Ribosómicas Grandes de Eucariotas/metabolismo , Ribosomas/genética , Ribosomas/patología , Síndrome de Shwachman-Diamond/genética , Síndrome de Shwachman-Diamond/patología , Adolescente , Adulto , Animales , Fenómenos Biológicos , Células Cultivadas , Niño , Preescolar , Dictyostelium , Drosophila , Factores Eucarióticos de Iniciación/genética , Factores Eucarióticos de Iniciación/metabolismo , Células Germinativas , Humanos , Lactante , Simulación de Dinámica Molecular , Factores de Elongación de Péptidos/genética , Factores de Elongación de Péptidos/metabolismo , Unión Proteica , Biosíntesis de Proteínas , Proteínas/genética , Proteínas/metabolismo , Ribonucleoproteína Nuclear Pequeña U5/genética , Ribonucleoproteína Nuclear Pequeña U5/metabolismo , Ribosomas/metabolismo , Saccharomyces cerevisiae , Homología de Secuencia de Aminoácido , Síndrome de Shwachman-Diamond/metabolismo , Adulto Joven
6.
Cell Rep ; 33(13): 108559, 2020 12 29.
Artículo en Inglés | MEDLINE | ID: mdl-33378670

RESUMEN

The MRE11-RAD50-NBS1 complex plays a central role in response to DNA double-strand breaks. Here, we identify a patient with bone marrow failure and developmental defects caused by biallelic RAD50 mutations. One of the mutations creates a null allele, whereas the other (RAD50E1035Δ) leads to the loss of a single residue in the heptad repeats within the RAD50 coiled-coil domain. This mutation represents a human RAD50 separation-of-function mutation that impairs DNA repair, DNA replication, and DNA end resection without affecting ATM-dependent DNA damage response. Purified recombinant proteins indicate that RAD50E1035Δ impairs MRE11 nuclease activity. The corresponding mutation in Saccharomyces cerevisiae causes severe thermosensitive defects in both DNA repair and Tel1ATM-dependent signaling. These findings demonstrate that a minor heptad break in the RAD50 coiled coil suffices to impede MRE11 complex functions in human and yeast. Furthermore, these results emphasize the importance of the RAD50 coiled coil to regulate MRE11-dependent DNA end resection in humans.


Asunto(s)
Ácido Anhídrido Hidrolasas/genética , Ácido Anhídrido Hidrolasas/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Endodesoxirribonucleasas/metabolismo , Exodesoxirribonucleasas/metabolismo , Proteína Homóloga de MRE11/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/fisiología , Trastornos de Fallo de la Médula Ósea/genética , Niño , Preescolar , Roturas del ADN de Doble Cadena , Reparación del ADN , Replicación del ADN , Discapacidades del Desarrollo/genética , Humanos , Unión Proteica , Dominios Proteicos , Análisis de Secuencia de Proteína , Eliminación de Secuencia , Transducción de Señal
7.
Hum Mol Genet ; 29(6): 907-922, 2020 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-31985013

RESUMEN

Telomeres are nucleoprotein structures at the end of chromosomes. The telomerase complex, constituted of the catalytic subunit TERT, the RNA matrix hTR and several cofactors, including the H/ACA box ribonucleoproteins Dyskerin, NOP10, GAR1, NAF1 and NHP2, regulates telomere length. In humans, inherited defects in telomere length maintenance are responsible for a wide spectrum of clinical premature aging manifestations including pulmonary fibrosis (PF), dyskeratosis congenita (DC), bone marrow failure and predisposition to cancer. NHP2 mutations have been so far reported only in two patients with DC. Here, we report the first case of Høyeraal-Hreidarsson syndrome, the severe form of DC, caused by biallelic missense mutations in NHP2. Additionally, we identified three unrelated patients with PF carrying NHP2 heterozygous mutations. Strikingly, one of these patients acquired a somatic mutation in the promoter of TERT that likely conferred a selective advantage in a subset of blood cells. Finally, we demonstrate that a functional deficit of human NHP2 affects ribosomal RNA biogenesis. Together, our results broaden the functional consequences and clinical spectrum of NHP2 deficiency.


Asunto(s)
Disqueratosis Congénita/patología , Retardo del Crecimiento Fetal/patología , Discapacidad Intelectual/patología , Microcefalia/patología , Mutación , Proteínas Nucleares/deficiencia , Proteínas Nucleares/genética , Fibrosis Pulmonar/patología , ARN Ribosómico/biosíntesis , Ribonucleoproteínas Nucleares Pequeñas/deficiencia , Ribonucleoproteínas Nucleares Pequeñas/genética , Anciano , Secuencia de Aminoácidos , Disqueratosis Congénita/etiología , Femenino , Retardo del Crecimiento Fetal/etiología , Humanos , Recién Nacido , Discapacidad Intelectual/etiología , Masculino , Microcefalia/etiología , Persona de Mediana Edad , Proteínas Nucleares/química , Linaje , Regiones Promotoras Genéticas , Fibrosis Pulmonar/etiología , Ribonucleoproteínas Nucleares Pequeñas/química , Homología de Secuencia , Telomerasa/genética , Transcripción Genética
8.
EMBO Mol Med ; 11(7): e10201, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31273937

RESUMEN

PARN, poly(A)-specific ribonuclease, regulates the turnover of mRNAs and the maturation and stabilization of the hTR RNA component of telomerase. Biallelic PARN mutations were associated with Høyeraal-Hreidarsson (HH) syndrome, a rare telomere biology disorder that, because of its severity, is likely not exclusively due to hTR down-regulation. Whether PARN deficiency was affecting the expression of telomere-related genes was still unclear. Using cells from two unrelated HH individuals carrying novel PARN mutations and a human PARN knock-out (KO) cell line with inducible PARN complementation, we found that PARN deficiency affects both telomere length and stability and down-regulates the expression of TRF1, TRF2, TPP1, RAP1, and POT1 shelterin transcripts. Down-regulation of dyskerin-encoding DKC1 mRNA was also observed and found to result from p53 activation in PARN-deficient cells. We further showed that PARN deficiency compromises ribosomal RNA biogenesis in patients' fibroblasts and cells from heterozygous Parn KO mice. Homozygous Parn KO however resulted in early embryonic lethality that was not overcome by p53 KO. Our results refine our knowledge on the pleiotropic cellular consequences of PARN deficiency.


Asunto(s)
Disqueratosis Congénita/metabolismo , Exorribonucleasas/deficiencia , Retardo del Crecimiento Fetal/metabolismo , Discapacidad Intelectual/metabolismo , Microcefalia/metabolismo , ARN Ribosómico/biosíntesis , Homeostasis del Telómero , Telómero/metabolismo , Animales , Preescolar , Modelos Animales de Enfermedad , Disqueratosis Congénita/genética , Disqueratosis Congénita/patología , Exorribonucleasas/metabolismo , Femenino , Retardo del Crecimiento Fetal/genética , Retardo del Crecimiento Fetal/patología , Humanos , Discapacidad Intelectual/genética , Discapacidad Intelectual/patología , Masculino , Ratones , Ratones Noqueados , Microcefalia/genética , Microcefalia/patología , ARN Ribosómico/genética , Complejo Shelterina , Telómero/genética , Telómero/patología , Proteínas de Unión a Telómeros
9.
Blood ; 134(3): 277-290, 2019 07 18.
Artículo en Inglés | MEDLINE | ID: mdl-31151987

RESUMEN

Shwachman-Diamond syndrome (SDS) is a recessive disorder typified by bone marrow failure and predisposition to hematological malignancies. SDS is predominantly caused by deficiency of the allosteric regulator Shwachman-Bodian-Diamond syndrome that cooperates with elongation factor-like GTPase 1 (EFL1) to catalyze release of the ribosome antiassociation factor eIF6 and activate translation. Here, we report biallelic mutations in EFL1 in 3 unrelated individuals with clinical features of SDS. Cellular defects in these individuals include impaired ribosomal subunit joining and attenuated global protein translation as a consequence of defective eIF6 eviction. In mice, Efl1 deficiency recapitulates key aspects of the SDS phenotype. By identifying biallelic EFL1 mutations in SDS, we define this leukemia predisposition disorder as a ribosomopathy that is caused by corruption of a fundamental, conserved mechanism, which licenses entry of the large ribosomal subunit into translation.


Asunto(s)
Mutación , Factores de Elongación de Péptidos/genética , Factores de Iniciación de Péptidos/biosíntesis , Ribonucleoproteína Nuclear Pequeña U5/genética , Síndrome de Shwachman-Diamond/genética , Síndrome de Shwachman-Diamond/metabolismo , Adolescente , Animales , Células Cultivadas , Análisis Mutacional de ADN , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Femenino , Estudio de Asociación del Genoma Completo , Humanos , Lactante , Masculino , Ratones , Ratones Transgénicos , Modelos Moleculares , Linaje , Factores de Elongación de Péptidos/química , Factores de Elongación de Péptidos/metabolismo , Fenotipo , Conformación Proteica , Ribonucleoproteína Nuclear Pequeña U5/química , Ribonucleoproteína Nuclear Pequeña U5/metabolismo , Síndrome de Shwachman-Diamond/diagnóstico , Relación Estructura-Actividad , Secuenciación Completa del Genoma
10.
Neuron ; 97(2): 326-340.e4, 2018 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-29346753

RESUMEN

At the optic chiasm choice point, ipsilateral retinal ganglion cells (RGCs) are repelled away from the midline by guidance cues, including Ephrin-B2 and Sonic Hedgehog (Shh). Although guidance cues are normally produced by cells residing at the choice point, the mRNA for Shh is not found at the optic chiasm. Here we show that Shh protein is instead produced by contralateral RGCs at the retina, transported anterogradely along the axon, and accumulates at the optic chiasm to repel ipsilateral RGCs. In vitro, contralateral RGC axons, which secrete Shh, repel ipsilateral RGCs in a Boc- and Smo-dependent manner. Finally, knockdown of Shh in the contralateral retina causes a decrease in the proportion of ipsilateral RGCs in a non-cell-autonomous manner. These findings reveal a role for axon-axon interactions in ipsilateral RGC guidance, and they establish that remotely produced cues can act at axon guidance midline choice points.


Asunto(s)
Transporte Axonal/fisiología , Axones/fisiología , Proteínas Hedgehog/metabolismo , Proteínas del Tejido Nervioso/fisiología , Quiasma Óptico/embriología , Células Ganglionares de la Retina/metabolismo , Animales , Inmunoglobulina G/fisiología , Ratones , Quiasma Óptico/metabolismo , Transporte de Proteínas , Receptores de Superficie Celular/fisiología , Receptor Smoothened/fisiología
11.
Cell Death Differ ; 25(2): 444-452, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29077092

RESUMEN

The repair of DNA double-stranded breaks (DNAdsb) through non-homologous end joining (NHEJ) is a prerequisite for the proper development of the central nervous system and the adaptive immune system. Yet, mice with Xlf or PAXX loss of function are viable and present with very mild immune phenotypes, although their lymphoid cells are sensitive to ionizing radiation attesting for the role of these factors in NHEJ. In contrast, we show here that mice defective for both Xlf and PAXX are embryonically lethal owing to a massive apoptosis of post-mitotic neurons, a situation reminiscent to XRCC4 or DNA Ligase IV KO conditions. The development of the adaptive immune system in Xlf-/-PAXX-/- E18.5 embryos is severely affected with the block of B- and T-cell maturation at the stage of IgH and TCRß gene rearrangements, respectively. This damaging phenotype highlights the functional nexus between Xlf and PAXX, which is critical for the completion of NHEJ-dependent mechanisms during mouse development.


Asunto(s)
Sistema Nervioso Central/crecimiento & desarrollo , Proteínas de Unión al ADN/metabolismo , Síndromes de Inmunodeficiencia/metabolismo , Animales , Sistema Nervioso Central/metabolismo , Reparación del ADN por Unión de Extremidades , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación , Fenotipo , Cementos de Resina/metabolismo
12.
Malar J ; 15: 248, 2016 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-27129434

RESUMEN

BACKGROUND: The asexual intra-erythrocytic multiplication of the malaria parasite Plasmodium falciparum is regulated by various molecular mechanisms. In eukaryotic cells, protein kinases are known to play key roles in cell cycle regulation and signaling pathways. The activity of cAMP-dependent protein kinase (PKA) depends on A-kinase anchoring proteins (AKAPs) through protein interactions. While several components of the cAMP dependent pathway-including the PKA catalytic and regulatory subunits-have been characterized in P. falciparum, whether AKAPs are involved in this pathway remains unclear. Here, PfAKAL, an open reading frame of a potential AKAP-like protein in the P. falciparum genome was identified, and its protein partners and putative cellular functions characterized. METHODS: The expression of PfAKAL throughout the erythrocytic cycle of the 3D7 strain was assessed by RT-qPCR and the presence of the corresponding protein by immunofluorescence assays. In order to study physical interactions between PfAKAL and other proteins, pull down experiments were performed using a recombinant PfAKAL protein and parasite protein extracts, or with recombinant proteins. These interactions were also tested by combining biochemical and proteomic approaches. As phosphorylation could be involved in the regulation of protein complexes, both PfAKAL and Pf14-3-3I phosphorylation was studied using a radiolabel kinase activity assay. Finally, to identify a potential function of the protein, PfAKAL sequence was aligned and structurally modeled, revealing a conserved nucleotide-binding pocket; confirmed by qualitative nucleotide binding experiments. RESULTS: PfAKAL is the first AKAP-like protein in P. falciparum to be identified, and shares 23 % sequence identity with the central domain of human AKAP18δ. PfAKAL is expressed in mature asexual stages, merozoites and gametocytes. In spite of homology to AKAP18, biochemical and immunochemical analyses demonstrated that PfAKAL does not interact directly with the P. falciparum PKA regulatory subunit (PfPKA-R), but instead binds and colocalizes with Pf14-3-3I, which in turn interacts with PfPKA-R. In vivo, these different interactions could be regulated by phosphorylation, as PfPKA-R and Pf14-3-3I, but not PfAKAL, are phosphorylated in vitro by PKA. Interestingly, PfAKAL binds nucleotides such as AMP and cAMP, suggesting that this protein may be involved in the AMP-activated protein kinase (AMPK) pathway, or associated with phosphodiesterase activities. CONCLUSION: PfAKAL is an atypical AKAP that shares common features with human AKAP18, such as nucleotides binding. The interaction of PfAKAL with PfPKA-R could be indirectly mediated through a join interaction with Pf14-3-3I. Therefore, PfPKA localization could not depend on PfAKAL, but rather involves other partners.


Asunto(s)
Proteínas de Anclaje a la Quinasa A/genética , Proteínas Quinasas Dependientes de AMP Cíclico/genética , Plasmodium falciparum/enzimología , Plasmodium falciparum/genética , Proteínas Protozoarias/genética , Proteínas de Anclaje a la Quinasa A/química , Proteínas de Anclaje a la Quinasa A/metabolismo , Secuencia de Aminoácidos , Proteínas Quinasas Dependientes de AMP Cíclico/química , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Plasmodium falciparum/metabolismo , Proteínas Protozoarias/química , Proteínas Protozoarias/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Alineación de Secuencia , Transducción de Señal
13.
Hum Mutat ; 37(5): 469-72, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26847928

RESUMEN

The DNA helicase RTEL1 participates in telomere maintenance and genome stability. Biallelic mutations in the RTEL1 gene account for the severe telomere biology disorder characteristic of the Hoyeraal-Hreidarsson syndrome (HH). Here, we report a HH patient (P4) carrying two novel compound heterozygous mutations in RTEL1: a premature stop codon (c.949A>T, p.Lys317*) and an intronic deletion leading to an exon skipping and an in-frame deletion of 25 amino-acids (p.Ile398_Lys422). P4's cells exhibit short and dysfunctional telomeres similarly to other RTEL1-deficient patients. 3D structure predictions indicated that the p.Ile398_Lys422 deletion affects a part of the helicase ARCH domain, which lines the pore formed with the core HD and the iron-sulfur cluster domains and is highly specific of sequences from the eukaryotic XPD family members.


Asunto(s)
ADN Helicasas/química , ADN Helicasas/genética , Disqueratosis Congénita/genética , Retardo del Crecimiento Fetal/genética , Discapacidad Intelectual/genética , Microcefalia/genética , Mutación , Niño , Codón de Terminación , Femenino , Humanos , Modelos Moleculares , Dominios Proteicos , Eliminación de Secuencia
14.
Blood Adv ; 1(1): 36-46, 2016 Nov 29.
Artículo en Inglés | MEDLINE | ID: mdl-29296694

RESUMEN

Telomeres are repetitive hexameric sequences located at the end of linear chromosomes. They adopt a lariat-like structure, the T-loop, to prevent them from being recognized as DNA breaks by the DNA repair machinery. RTEL1 is a DNA helicase required for proper telomere replication and stability. In particular, it has been postulated that RTEL1 is involved in the opening of the T-loop during telomere replication to avoid sudden telomere deletion and telomere circle (T-circle) formation. In humans, biallelic RTEL1 mutations cause Hoyeraal-Hreidarsson syndrome (HH), a rare and severe telomere biology disorder characterized by intrauterine growth retardation, bone marrow failure, microcephaly and/or cerebellar hypoplasia, and immunodeficiency. To date, 18 different RTEL1 mutations have been described in 19 cases of HH with short telomeres. The impaired T-loop resolution has been proposed to be a major cause of telomere shortening in RTEL1 deficiency. However, the biological and clinical consequences of this disorder remain incompletely documented. Here, we describe 4 new patients harboring biallelic RTEL1 mutations, including 2 novel missense mutations located in the C-terminal end of RTEL1 (p.Cys1268Arg and p.Val1294Phe). Clinical characteristics from these 4 patients were collected as those from 4 other RTEL1-deficient patients previously reported. In addition, we assessed whether T-circles, the product of improper T-loop resolution, were detected in our RTEL1-deficient patients. Overall, our study broadens and refines the clinical and biological spectrum of human RTEL1 deficiency.

15.
J Allergy Clin Immunol ; 136(6): 1619-1626.e5, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26220525

RESUMEN

BACKGROUND: Myb-Like, SWIRM, and MPN domains 1 (MYSM1) is a metalloprotease that deubiquitinates the K119-monoubiquitinated form of histone 2A (H2A), a chromatin marker associated with gene transcription silencing. Likewise, it has been reported that murine Mysm1 participates in transcription derepression of genes, among which are transcription factors involved in hematopoietic stem cell homeostasis, hematopoiesis, and lymphocyte differentiation. However, whether MYSM1 has a similar function in human subjects remains unclear. Here we describe a patient presenting with a complete lack of B lymphocytes, T-cell lymphopenia, defective hematopoiesis, and developmental abnormalities. OBJECTIVES: We sought to characterize the underlying genetic cause of this syndrome. METHODS: We performed genome-wide homozygosity mapping, followed by whole-exome sequencing. RESULTS: Genetic analysis revealed that this novel disorder is caused by a homozygous MYSM1 missense mutation affecting the catalytic site within the deubiquitinase JAB1/MPN/Mov34 (JAMM)/MPN domain. Remarkably, during the course of our study, the patient recovered a normal immunohematologic phenotype. Genetic analysis indicated that this improvement originated from a spontaneous genetic reversion of the MYSM1 mutation in a hematopoietic stem cell. CONCLUSIONS: We here define a novel human immunodeficiency and provide evidence that MYSM1 is essential for proper immunohematopoietic development in human subjects. In addition, we describe one of the few examples of spontaneous in vivo genetic cure of a human immunodeficiency.


Asunto(s)
Proteínas de Unión al ADN/genética , Síndromes de Inmunodeficiencia/genética , Factores de Transcripción/genética , Linfocitos B/citología , Diferenciación Celular , Hematopoyesis/genética , Humanos , Lactante , Linfopenia/genética , Masculino , Mutación , Linfocitos T/citología , Transactivadores , Proteasas Ubiquitina-Específicas
16.
Med Sci (Paris) ; 29(12): 1138-44, 2013 Dec.
Artículo en Francés | MEDLINE | ID: mdl-24356145

RESUMEN

RTEL1 (regulator of telomere length helicase 1) is a DNA helicase that has been identified more than 10 years ago. Many works since, mainly in the nematode Caenorhabditis elegans and the mouse, have highlighted its role in chromosomal stability, maintenance of telomere length, and DNA repair. Recently, four laboratories have characterized RTEL1 mutations in patients with dyskeratosis congenita (DC) and Hoyeraal-Hreidarsson (HH) syndrome, a rare and severe variant of DC. We here summarize the current knowledge on RTEL1 and discuss the possible other functions that RTEL1 could play.


Asunto(s)
ADN Helicasas/fisiología , Inestabilidad Genómica/fisiología , Animales , ADN Helicasas/genética , Reparación del ADN , Disqueratosis Congénita/genética , Retardo del Crecimiento Fetal/genética , Fenómenos Genéticos/genética , Fenómenos Genéticos/fisiología , Inestabilidad Genómica/genética , Humanos , Discapacidad Intelectual/genética , Ratones , Microcefalia/genética , Mutación , Telómero/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...