Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Br J Haematol ; 204(1): 292-305, 2024 01.
Artículo en Inglés | MEDLINE | ID: mdl-37876306

RESUMEN

Shwachman-Diamond syndrome (SDS) is characterized by neutropenia, exocrine pancreatic insufficiency and skeletal abnormalities. SDS bone marrow haematopoietic progenitors show increased apoptosis and impairment in granulocytic differentiation. Loss of Shwachman-Bodian-Diamond syndrome (SBDS) expression results in reduced eukaryotic 80S ribosome maturation. Biallelic mutations in the SBDS gene are found in ~90% of SDS patients, ~55% of whom carry the c.183-184TA>CT nonsense mutation. Several translational readthrough-inducing drugs aimed at suppressing nonsense mutations have been developed. One of these, ataluren, has received approval in Europe for the treatment of Duchenne muscular dystrophy. We previously showed that ataluren can restore full-length SBDS protein synthesis in SDS-derived bone marrow cells. Here, we extend our preclinical study to assess the functional restoration of SBDS capabilities in vitro and ex vivo. Ataluren improved 80S ribosome assembly and total protein synthesis in SDS-derived cells, restored myelopoiesis in myeloid progenitors, improved neutrophil chemotaxis in vitro and reduced neutrophil dysplastic markers ex vivo. Ataluren also restored full-length SBDS synthesis in primary osteoblasts, suggesting that its beneficial role may go beyond the myeloid compartment. Altogether, our results strengthened the rationale for a Phase I/II clinical trial of ataluren in SDS patients who harbour the nonsense mutation.


Asunto(s)
Enfermedades de la Médula Ósea , Insuficiencia Pancreática Exocrina , Lipomatosis , Humanos , Síndrome de Shwachman-Diamond , Proteína p53 Supresora de Tumor/genética , Lipomatosis/genética , Codón sin Sentido , Mielopoyesis , Neutrófilos/metabolismo , Quimiotaxis , Enfermedades de la Médula Ósea/genética , Enfermedades de la Médula Ósea/terapia , Insuficiencia Pancreática Exocrina/genética , Ribosomas/metabolismo
2.
Am J Med Genet A ; : e63901, 2024 Oct 11.
Artículo en Inglés | MEDLINE | ID: mdl-39392177

RESUMEN

Mosaic variegated aneuploidy (MVA) is an autosomal recessive disorder characterized by mosaic aneuploidies, predominantly trisomies, involving multiple different chromosomes and tissues. The proportion of aneuploid cells varies, and most patients present with intrauterine growth delay, microcephaly, and a broad spectrum of congenital abnormalities. We report a patient with a distinctive type of MVA discovered in bone marrow (BM) when she was 3-month-old due to neutropenia and hypocellular bone marrow. She was followed up for more than 20 years, and different trisomic cells were repeatedly discovered in different tissues, whereas her clinical picture has never been severe. The main sign remained intermittent neutropenia, not cyclic and often not too severe, occasionally with anemia and thrombocytopenia. Retromicrognathia was the only dysmorphic sign. Unlike other patients with MVA, the trisomies in all tissues involved almost invariably chromosomes 18 and 19. Therefore, the peculiarities of our patient were the clinical and the atypical cytogenetic pictures. Nevertheless, we looked for mutations in the seven causative genes of the known types of MVA, but the results were negative. Then, we analyzed the entire exome to find out other possible causing mutations, but also this attempt failed to discover a possible cause of this distinctive form of MVA.

3.
Int J Mol Sci ; 22(24)2021 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-34948128

RESUMEN

Shwachman-Diamond syndrome (SDS) is a rare autosomal recessive disorder characterized by bone marrow failure, exocrine pancreatic insufficiency, and skeletal abnormalities, caused by loss-of-function mutations in the SBDS gene, a factor involved in ribosome biogenesis. By analyzing osteoblasts from SDS patients (SDS-OBs), we show that SDS-OBs displayed reduced SBDS gene expression and reduced/undetectable SBDS protein compared to osteoblasts from healthy subjects (H-OBs). SDS-OBs cultured in an osteogenic medium displayed a lower mineralization capacity compared to H-OBs. Whole transcriptome analysis showed significant differences in the gene expression of SDS-OBs vs. H-OBs, particularly in the ossification pathway. SDS-OBs expressed lower levels of the main genes responsible for osteoblastogenesis. Of all downregulated genes, Western blot analyses confirmed lower levels of alkaline phosphatase and collagen type I in SDS-OBs than in H-OBs. Interestingly, SDS-OBs showed higher protein levels of p53, an inhibitor of osteogenesis, compared to H-OBs. Silencing of Tp53 was associated with higher collagen type I and alkaline phosphatase protein levels and an increase in SDS-OB mineralization capacity. In conclusion, our results show that the reduced capacity of SDS-OBs to mineralize is mediated, at least in part, by the high levels of p53 and highlight an important role of SBDS in osteoblast functions.


Asunto(s)
Calcificación Fisiológica , Osteoblastos/metabolismo , Síndrome de Shwachman-Diamond/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Células Cultivadas , Femenino , Humanos , Masculino , Osteoblastos/patología , Proteínas/genética , Proteínas/metabolismo , Síndrome de Shwachman-Diamond/genética , Síndrome de Shwachman-Diamond/patología , Proteína p53 Supresora de Tumor/genética
4.
Int J Mol Sci ; 21(6)2020 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-32178390

RESUMEN

The growing trend for women to postpone childbearing has resulted in a dramatic increase in the incidence of aneuploid pregnancies. Despite the importance to human reproductive health, the events precipitating female age-related meiotic errors are poorly understood. To gain new insight into the molecular basis of age-related chromosome missegregation in human oocytes, we combined the transcriptome profiles of twenty single oocytes (derived from females divided into two groups according to age <35 and ≥35 years) with their chromosome status obtained by array comparative genomic hybridization (aCGH). Furthermore, we compared the transcription profile of the single oocyte with the surrounding cumulus cells (CCs). RNA-seq data showed differences in gene expression between young and old oocytes. Dysregulated genes play a role in important biological processes such as gene transcription regulation, cytoskeleton organization, pathways related to RNA maturation and translation. The comparison of the transcription profile of the oocyte and the corresponding CCs highlighted the differential expression of genes belonging to the G protein-coupled receptor superfamily. Finally, we detected the loss of a X chromosome in two oocytes derived from women belonging to the ≥35 years age group. These aneuploidies may be caused by the detriment of REEP4, an endoplasmic reticulum protein, in women aged ≥35 years. Here we gained new insight into the complex regulatory circuit between the oocyte and the surrounding CCs and uncovered a new putative molecular basis of age-related chromosome missegregation in human oocytes.


Asunto(s)
Cromosomas/genética , Oocitos/fisiología , Transcriptoma/genética , Adulto , Aneuploidia , Células Cultivadas , Hibridación Genómica Comparativa/métodos , Células del Cúmulo/fisiología , Femenino , Perfilación de la Expresión Génica/métodos , Regulación de la Expresión Génica/genética , Humanos , Masculino , Análisis por Micromatrices/métodos , Embarazo , Adulto Joven
5.
Br J Haematol ; 184(6): 974-981, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30585299

RESUMEN

In Shwachman-Diamond syndrome (SDS), deletion of the long arm of chromosome 20, del(20)(q), often acquired in bone marrow (BM), may imply a lower risk of developing myelodysplastic syndrome/acute myeloid leukaemia (MDS/AML), due to the loss of the EIF6 gene. The genes L3MBTL1 and SGK2, also on chromosome 20, are in a cluster of imprinted genes, and their loss implies dysregulation of BM function. We report here the results of array comparative genomic hybridization (a-CGH) performed on BM DNA of six patients which confirmed the consistent loss of EIF6 gene. Interestingly, array single nucleotide polymorphisms (SNPs) showed copy neutral loss of heterozygosity for EIF6 region in cases without del(20)(q). No preferential parental origin of the deleted chromosome 20 was detected by microsatellite analysis in six SDS patients. Our patients showed a very mild haematological condition, and none evolved into BM aplasia or MDS/AML. We extend the benign prognostic significance of del(20)(q) and loss of EIF6 to the haematological features of these patients, consistently characterized by mild hypoplastic BM, no or mild neutropenia, anaemia and thrombocytopenia. Some odd results obtained in microsatellite and SNP-array analysis demonstrate a peculiar genomic instability, in an attempt to improve BM function through the acquisition of the del(20)(q).


Asunto(s)
Cromosomas Humanos Par 20/genética , Inestabilidad Genómica/genética , Síndrome de Shwachman-Diamond/genética , Adolescente , Adulto , Niño , Femenino , Humanos , Masculino , Pronóstico , Síndrome de Shwachman-Diamond/patología , Adulto Joven
6.
Pediatr Blood Cancer ; 64(8)2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28130858

RESUMEN

BACKGROUND: Two chromosome anomalies are frequent in the bone marrow (BM) of patients with Shwachman-Diamond syndrome (SDS): an isochromosome of the long arm of chromosome 7, i(7)(q10), and an interstitial deletion of the long arm of chromosome 20, del(20)(q). These anomalies are associated with a lower risk of developing myelodysplasia (MDS) and/or acute myeloid leukemia. The chromosome anomalies may be due to an SDS-specific karyotype instability, reflected also by anomalies that are not clonal, but found in single cells in the BM or in peripheral blood (PB). PROCEDURE: Starting in 1999, we have monitored the cytogenetic picture of a cohort of 91 Italian patients with SDS by all suitable cytogenetic and molecular methods. RESULTS: Here, we report clonal chromosome anomalies that are different from the aforementioned, as well as changes found in single cells in BM/PB of the same patients. CONCLUSIONS: Some of the newly recognized clonal anomalies in BM reported here are recurrent, especially unbalanced structural anomalies of chromosome 7, a further complex rearrangement of the del(20)(q) with duplicated and deleted portions, and an unbalanced translocation t(3;6), with partial trisomy of the long arm of chromosome 3 and partial monosomy of the long arm of chromosome 6. Firm conclusions on the possible prognostic relevance of these anomalies would require further study with larger patient cohorts, but our data are sufficient to suggest that these patients necessitate more frequent cytogenetic monitoring. The results on anomalies found in single cells confirm the presence of an SDS-specific karyotype instability.


Asunto(s)
Enfermedades de la Médula Ósea/genética , Aberraciones Cromosómicas , Insuficiencia Pancreática Exocrina/genética , Lipomatosis/genética , Cariotipo Anormal , Adolescente , Adulto , Niño , Femenino , Humanos , Hibridación Fluorescente in Situ , Lactante , Estudios Longitudinales , Masculino , Análisis de Secuencia por Matrices de Oligonucleótidos , Recurrencia , Síndrome de Shwachman-Diamond , Adulto Joven
7.
Nat Genet ; 39(8): 960-2, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17632511

RESUMEN

Autosomal recessive osteopetrosis is usually associated with normal or elevated numbers of nonfunctional osteoclasts. Here we report mutations in the gene encoding RANKL (receptor activator of nuclear factor-KB ligand) in six individuals with autosomal recessive osteopetrosis whose bone biopsy specimens lacked osteoclasts. These individuals did not show any obvious defects in immunological parameters and could not be cured by hematopoietic stem cell transplantation; however, exogenous RANKL induced formation of functional osteoclasts from their monocytes, suggesting that they could, theoretically, benefit from exogenous RANKL administration.


Asunto(s)
Osteopetrosis/genética , Ligando RANK/genética , Animales , Consanguinidad , Femenino , Genes Recesivos , Humanos , Masculino , Ratones , Osteoclastos , Linaje
8.
Genes Chromosomes Cancer ; 54(12): 771-5, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26355525

RESUMEN

Olfactory neuroblastoma is a rare tumor arising from the basal layer of the olfactory epithelium in the superior recesses of the nasal cavity. The rarity of this tumor, and the difficulties in culturing tumor cells has limited the generation of conventional cytogenetic data, whereas consistent results have been obtained by recent molecular methods. We report the results of an array-based comparative genomic hybridization analysis (a-CGH) obtained on 11 samples from 10 subjects: 8 primary and 3 relapsed tumors. In one patient, both the primary and relapsed tumors were available. Our results on chromosome imbalances highlight the highly heterogeneous presentation: six of eleven samples showed multiple numerical changes and very few structural ones, while four samples showed an opposite pattern; one sample out of eleven showed no imbalances. We did not reach firm evidence of any recurrent specific imbalances either at level of entire chromosomes or chromosome segments. A review of the literature indicates a number of recurrent gains, and losses, mostly not confirmed by our results. Gain of chromosome 19 was the only correspondence with literature data concerning an entire chromosome, and most segmental gains and losses found in our cohort of patients were different from those indicated in the literature: the only similarities concerned the gain of 20q13 and the loss of segments of chromosomes 15 and 22.


Asunto(s)
Estesioneuroblastoma Olfatorio/genética , Cavidad Nasal/patología , Neoplasias Nasales/genética , Enfermedades Raras/genética , Adulto , Anciano , Anciano de 80 o más Años , Aberraciones Cromosómicas , Cromosomas Humanos Par 15 , Cromosomas Humanos Par 19 , Cromosomas Humanos Par 22 , Hibridación Genómica Comparativa/métodos , Femenino , Humanos , Masculino , Persona de Mediana Edad , Recurrencia Local de Neoplasia/genética , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos
9.
J Exp Clin Cancer Res ; 43(1): 49, 2024 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-38365745

RESUMEN

BACKGROUND: SMC1A is a subunit of the cohesin complex that participates in many DNA- and chromosome-related biological processes. Previous studies have established that SMC1A is involved in cancer development and in particular, is overexpressed in chromosomally unstable human colorectal cancer (CRC). This study aimed to investigate whether SMC1A could serve as a therapeutic target for CRC. METHODS: At first, we studied the effects of either SMC1A overexpression or knockdown in vitro. Next, the outcome of SMC1A knocking down (alone or in combination with bevacizumab, a monoclonal antibody against vascular endothelial growth factor) was analyzed in vivo. RESULTS: We found that SMC1A knockdown affects cell proliferation and reduces the ability to grow in anchorage-independent manner. Next, we demonstrated that the silencing of SMC1A and the combo treatment were effective in increasing overall survival in a xenograft mouse model. Functional analyses indicated that both treatments lead to atypical mitotic figures and gene expression dysregulation. Differentially expressed genes were implicated in several pathways including gene transcription regulation, cellular proliferation, and other transformation-associated processes. CONCLUSIONS: These results indicate that SMC1A silencing, in combination with bevacizumab, can represent a promising therapeutic strategy for human CRC.


Asunto(s)
Cohesinas , Neoplasias Colorrectales , Animales , Humanos , Ratones , Bevacizumab/farmacología , Bevacizumab/uso terapéutico , Proteínas de Ciclo Celular/metabolismo , Proliferación Celular , Proteínas Cromosómicas no Histona/genética , Cohesinas/genética , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Silenciador del Gen , Factor A de Crecimiento Endotelial Vascular/genética
10.
Genes (Basel) ; 14(7)2023 06 27.
Artículo en Inglés | MEDLINE | ID: mdl-37510256

RESUMEN

Chronic myeloid leukemia (CML) is a rare myeloproliferative disorder caused by the reciprocal translocation t(9;22)(q34;q11) in hematopoietic stem cells (HSCs). This chromosomal translocation results in the formation of an extra-short chromosome 22, called a Philadelphia chromosome (Ph), containing the BCR-ABL1 fusion gene responsible for the expression of a constitutively active tyrosine kinase that causes uncontrolled growth and replication of leukemic cells. Mechanisms behind the formation of this chromosomal rearrangement are not well known, even if, as observed in tumors, repetitive DNA may be involved as core elements in chromosomal rearrangements. We have participated in the explorative investigations of the PhilosoPhi34 study to evaluate residual Ph+ cells in patients with negative FISH analysis on CD34+/lin- cells with gDNA qPCR. Using targeted next-generation deep sequencing strategies, we analyzed the genomic region around the t(9;22) translocations of 82 CML patients and one CML cell line and assessed the relevance of interspersed repeat elements at breakpoints (BP). We found a statistically higher presence of LINE elements, in particular belonging to the subfamily L1M, in BP cluster regions of both chromosome 22 and 9 compared to the whole human genome. These data suggest that L1M elements could be potential drivers of t(9;22) translocation leading to the generation of the BCR-ABL1 chimeric gene and the expression of the active BCR-ABL1-controlled tyrosine kinase chimeric protein responsible for CML.


Asunto(s)
Leucemia Mielógena Crónica BCR-ABL Positiva , Trastornos Mieloproliferativos , Humanos , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Cromosoma Filadelfia , Translocación Genética , Proteínas de Fusión bcr-abl/genética , Trastornos Mieloproliferativos/genética
11.
Genes (Basel) ; 14(11)2023 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-38003028

RESUMEN

The patient reported here underwent hematopoietic stem cell transplantation (HSCT) due to chronic granulomatous disease (CGD) caused by biallelic mutations of the NCF1 gene. Two years later, he developed AML, which was unexpected and was recognized via sex-mismatched chromosomes as deriving from the donor cells; the patient was male, and the donor was his sister. Donor cell leukemia (DCL) is very rare, and it had never been reported in patients with CGD after HSCT. In the subsequent ten years, the AML relapsed three times and the patient underwent chemotherapy and three further HSCTs; donors were the same sister from the first HSCT, an unrelated donor, and his mother. The patient died during the third relapse. The DCL was characterized since onset by an acquired translocation between chromosomes 9 and 11, with a molecular rearrangement between the MLL and MLLT3 genes-a quite frequent cause of AML. In all of the relapses, the malignant clone had XX sex chromosomes and this rearrangement, thus indicating that it was always the original clone derived from the transplanted sister's cells. It exhibited the ability to remain quiescent in the BM during repeated chemotherapy courses, remission periods and HSCT. The leukemic clone then acquired different additional anomalies during the ten years of follow-up, with cytogenetic results characterized both by anomalies frequent in AML and by different, non-recurrent changes. This type of cytogenetic course is uncommon in AML.


Asunto(s)
Enfermedad Granulomatosa Crónica , Trasplante de Células Madre Hematopoyéticas , Leucemia Mieloide Aguda , Humanos , Masculino , Enfermedad Granulomatosa Crónica/genética , Enfermedad Granulomatosa Crónica/terapia , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Trasplante de Células Madre Hematopoyéticas/métodos , Donante no Emparentado , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/terapia , Leucemia Mieloide Aguda/patología , Translocación Genética
12.
Calcif Tissue Int ; 91(4): 250-4, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22847576

RESUMEN

Human malignant autosomal recessive osteopetrosis (ARO) is a genetically heterogeneous disorder caused by reduced bone resorption by osteoclasts. Mutations in the CLCN7 gene are responsible not only for a substantial portion of ARO patients but also for other forms of osteopetrosis characterized by different severity and inheritance. The lack of a clear genotype/phenotype correlation makes genetic counseling a tricky issue for CLCN7-dependent osteopetrosis. Here, we characterize the first homozygous interstitial deletion in 16p13.3, detected by array comparative genomic hybridization in an ARO patient of Jordanian origin. The deletion involved other genes besides CLCN7, while the proband displayed a classic ARO phenotype; however, her early death did not allow more extensive clinical investigations. The identification of this novel genomic deletion involving a large part of the CLCN7 gene is of clinical relevance, especially in prenatal diagnosis, and suggests the possibility that this kind of mutation has been underestimated so far. These data highlight the need for alternative approaches to genetic analysis also in other ARO-causative genes.


Asunto(s)
Cromosomas Humanos Par 16/genética , Eliminación de Gen , Genes Recesivos , Homocigoto , Osteopetrosis/genética , Secuencia de Bases , Canales de Cloruro/genética , Hibridación Genómica Comparativa , Humanos , Lactante , Datos de Secuencia Molecular , Mutación , Fenotipo
13.
Front Genet ; 13: 896749, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36035165

RESUMEN

Background: Shwachman-Diamond syndrome (SDS) is a rare autosomal recessive ribosomopathy mainly characterized by exocrine pancreatic insufficiency, skeletal alterations, neutropenia, and a relevant risk of hematological transformation. At least 90% of SDS patients have pathogenic variants in SBDS, the first gene associated with the disease with very low allelic heterogeneity; three variants, derived from events of genetic conversion between SBDS and its pseudogene, SBDSP1, provided the alleles observed in about 62% of SDS patients. Methods: We performed a reanalysis of the available WES files of a group of SDS patients with biallelic SBDS pathogenic variants, studying the results by next bioinformatic and protein structural analysis. Parallelly, careful clinical attention was given to the patient focused in this study. Results: We found and confirmed in one SDS patient a germline heterozygous missense variant (c.100T>C; p.Phe34Leu) in the EIF6 gene. This variant, inherited from his mother, has a very low frequency, and it is predicted as pathogenic, according to several in silico prediction tools. The protein structural analysis also envisages the variant could reduce the binding to the nascent 60S ribosomal. Conclusion: This study focused on the hypothesis that the EIF6 germline variant mimics the effect of somatic deletions of chromosome 20, always including the locus of this gene, and similarly may rescue the ribosomal stress and ribosomal dysfunction due to SBDS mutations. It is likely that this rescue may contribute to the stable and not severe hematological status of the proband, but a definite answer on the role of this EIF6 variant can be obtained only by adding a functional layer of evidence. In the future, these results are likely to be useful for selected cases in personalized medicine and therapy.

14.
Hum Mol Genet ; 18(3): 418-27, 2009 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-18996922

RESUMEN

Cornelia de Lange syndrome (CdLS) is a clinically heterogeneous developmental disorder characterized by facial dysmorphia, upper limb malformations, growth and cognitive retardation. Mutations in the sister chromatid cohesion factor genes NIPBL, SMC1A and SMC3 are present in approximately 65% of CdLS patients. In addition to their canonical roles in chromosome segregation, the cohesin proteins are involved in other biological processes such as regulation of gene expression, DNA repair and maintenance of genome stability. To gain insights into the molecular basis of CdLS, we analyzed the affinity of mutated SMC1A and SMC3 hinge domains for DNA. Mutated hinge dimers bind DNA with higher affinity than wild-type proteins. SMC1A- and SMC3-mutated CdLS cell lines display genomic instability and sensitivity to ionizing radiation and interstrand crosslinking agents. We propose that SMC1A and SMC3 CdLS mutations affect the dynamic association between SMC proteins and DNA, providing new clues to the underlying molecular cause of CdLS.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Proteoglicanos Tipo Condroitín Sulfato/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Proteínas de Unión al ADN/metabolismo , Síndrome de Cornelia de Lange/genética , Mutación , Proteínas de Ciclo Celular/genética , Línea Celular , Células Cultivadas , Proteoglicanos Tipo Condroitín Sulfato/genética , Proteínas Cromosómicas no Histona/genética , ADN/genética , ADN/metabolismo , Daño del ADN , Proteínas de Unión al ADN/genética , Síndrome de Cornelia de Lange/metabolismo , Femenino , Humanos , Unión Proteica , Cohesinas
15.
Am J Hum Genet ; 83(1): 64-76, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18606301

RESUMEN

Autosomal-Recessive Osteopetrosis (ARO) comprises a heterogeneous group of bone diseases for which mutations in five genes are known as causative. Most ARO are classified as osteoclast-rich, but recently a subset of osteoclast-poor ARO has been recognized as due to a defect in TNFSF11 (also called RANKL or TRANCE, coding for the RANKL protein), a master gene driving osteoclast differentiation along the RANKL-RANK axis. RANKL and RANK (coded for by the TNFRSF11A gene) also play a role in the immune system, which raises the possibility that defects in this pathway might cause osteopetrosis with immunodeficiency. From a large series of ARO patients we selected a Turkish consanguineous family with two siblings affected by ARO and hypogammaglobulinemia with no defects in known osteopetrosis genes. Sequencing of genes involved in the RANKL downstream pathway identified a homozygous mutation in the TNFRSF11A gene in both siblings. Their monocytes failed to differentiate in vitro into osteoclasts upon exposure to M-CSF and RANKL, in keeping with an osteoclast-intrinsic defect. Immunological analysis showed that their hypogammaglobulinemia was associated with impairment in immunoglobulin-secreting B cells. Investigation of other patients revealed a defect in both TNFRSF11A alleles in six additional, unrelated families. Our results indicate that TNFRSF11A mutations can cause a clinical condition in which severe ARO is associated with an immunoglobulin-production defect.


Asunto(s)
Agammaglobulinemia/sangre , Osteoclastos/patología , Osteopetrosis/genética , Receptor Activador del Factor Nuclear kappa-B/genética , Fosfatasa Ácida/metabolismo , Actinas/metabolismo , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Argentina , Arginina/metabolismo , Biopsia , Estudios de Casos y Controles , Línea Celular Transformada , Proliferación Celular , Transformación Celular Viral , Células Cultivadas , Estudios de Cohortes , Consanguinidad , Cisteína/metabolismo , Análisis Mutacional de ADN , Dendritas/fisiología , Femenino , Genes Recesivos , Herpesvirus Humano 4/fisiología , Heterocigoto , Homocigoto , Humanos , Ilion/cirugía , Isoenzimas/metabolismo , Antígenos Comunes de Leucocito/metabolismo , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/patología , Lipopolisacáridos/farmacología , Factor Estimulante de Colonias de Macrófagos/farmacología , Masculino , Modelos Inmunológicos , Datos de Secuencia Molecular , Mutación Missense , Osteoclastos/metabolismo , Osteoclastos/ultraestructura , Osteopetrosis/diagnóstico , Osteopetrosis/diagnóstico por imagen , Osteopetrosis/patología , Osteopetrosis/fisiopatología , Osteoprotegerina/metabolismo , Pakistán , Linaje , Polimorfismo Genético , Estructura Terciaria de Proteína , Ligando RANK/metabolismo , Radiografía Torácica/métodos , Receptor Activador del Factor Nuclear kappa-B/química , Receptor Activador del Factor Nuclear kappa-B/inmunología , Receptores de Vitronectina/metabolismo
16.
Blood ; 114(2): 459-68, 2009 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-19414862

RESUMEN

Autosomal dominant osteogenesis imperfecta (OI) caused by glycine substitutions in type I collagen is a paradigmatic disorder for stem cell therapy. Bone marrow transplantation in OI children has produced a low engraftment rate, but surprisingly encouraging symptomatic improvements. In utero transplantation (IUT) may hold even more promise. However, systematic studies of both methods have so far been limited to a recessive mouse model. In this study, we evaluated intrauterine transplantation of adult bone marrow into heterozygous BrtlIV mice. Brtl is a knockin mouse with a classical glycine substitution in type I collagen [alpha1(I)-Gly349Cys], dominant trait transmission, and a phenotype resembling moderately severe and lethal OI. Adult bone marrow donor cells from enhanced green fluorescent protein (eGFP) transgenic mice engrafted in hematopoietic and nonhematopoietic tissues differentiated to trabecular and cortical bone cells and synthesized up to 20% of all type I collagen in the host bone. The transplantation eliminated the perinatal lethality of heterozygous BrtlIV mice. At 2 months of age, femora of treated Brtl mice had significant improvement in geometric parameters (P < .05) versus untreated Brtl mice, and their mechanical properties attained wild-type values. Our results suggest that the engrafted cells form bone with higher efficiency than the endogenous cells, supporting IUT as a promising approach for the treatment of genetic bone diseases.


Asunto(s)
Envejecimiento/fisiología , Trasplante de Médula Ósea/métodos , Investigación Fetal , Osteogénesis Imperfecta/prevención & control , Osteogénesis Imperfecta/terapia , Útero/fisiología , Animales , Células de la Médula Ósea/citología , Colágeno/metabolismo , Modelos Animales de Enfermedad , Espacio Extracelular/química , Femenino , Técnicas de Sustitución del Gen , Genes Dominantes , Supervivencia de Injerto , Ratones , Ratones Transgénicos , Osteogénesis Imperfecta/metabolismo , Osteogénesis Imperfecta/patología , Fenotipo , Embarazo , Espectrometría Raman , Tasa de Supervivencia , Donantes de Tejidos
17.
Nat Med ; 9(4): 399-406, 2003 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-12627228

RESUMEN

The spontaneous mouse grey-lethal (gl) mutation is responsible for a coat color defect and for the development of the most severe autosomal recessive form of osteopetrosis. Using a positional cloning approach, we have mapped and isolated the gl locus from a approximately 1.5 cM genetic interval. The gl locus was identified in a bacterial artificial chromosome (BAC) contig by functional genetic complementation in transgenic mice. Genomic sequence analysis revealed that the gl mutation is a deletion resulting in complete loss of function. The unique approximately 3 kb wild-type transcript is expressed primarily in osteoclasts and melanocytes as well as in brain, kidney, thymus and spleen. The gl gene is predicted to encode a 338-amino acid type I transmembrane protein that localizes to the intracellular compartment. Mutation in the human GL gene leads to severe recessive osteopetrosis. Our studies show that mouse Gl protein function is absolutely required for osteoclast and melanocyte maturation and function.


Asunto(s)
Genes Letales , Genes Recesivos , Proteínas de la Membrana/genética , Osteopetrosis/genética , Trastornos de la Pigmentación/genética , Secuencia de Aminoácidos , Animales , Cromosomas Artificiales Bacterianos , Eliminación de Gen , Humanos , Líquido Intracelular/metabolismo , Melanocitos/fisiología , Proteínas de la Membrana/química , Ratones , Datos de Secuencia Molecular , Osteoclastos/fisiología , Osteopetrosis/patología , Mapeo Físico de Cromosoma , Homología de Secuencia de Aminoácido
18.
Mol Cytogenet ; 14(1): 54, 2021 Nov 24.
Artículo en Inglés | MEDLINE | ID: mdl-34819134

RESUMEN

BACKGROUND: An isochromosome of the long arm of chromosome 7, i(7)(q10), and an interstitial deletion of the long arm of chromosome 20, del(20)(q), are the most frequent anomalies in the bone marrow of patients with Shwachman-Diamond syndrome, which is caused in most cases by mutations of the SBDS gene. These clonal changes imply milder haematological symptoms and lower risk of myelodysplastic syndromes and acute myeloid leukaemia, thanks to already postulated rescue mechanisms. RESULTS: Bone marrow from fourteen patients exhibiting either the i(7)(q10) or the del(20)(q) and coming from two large cohorts of patients, were subjected to chromosome analyses, Fluorescent In Situ Hybridization with informative probes and array-Comparative Genomic Hybridization. One patient with the i(7)(q10) showed a subsequent clonal rearrangement of the normal chromosome 7 across years. Four patients carrying the del(20)(q) evolved further different del(20)(q) independent clones, within a single bone marrow sample, or across sequential samples. One patient with the del(20)(q), developed a parallel different clone with a duplication of chromosome 3 long arm. Eight patients bore the del(20)(q) as the sole chromosomal abnormality. An overall overview of patients with the del(20)(q), also including cases already reported, confirmed that all the deletions were interstitial. The loss of material varied from 1.7 to 26.9 Mb and resulted in the loss of the EIF6 gene in all patients. CONCLUSIONS: Although the i(7)(q) and the del(20)(q) clones are frequent and clinically benign in Shwachman Diamond-syndrome, in the present work we show that they may rearrange, may be lost and then reconstructed de novo, or may evolve with independent clones across years. These findings unravel a striking selective pressure exerted by SBDS deficiency driving to karyotype instability and to specific clonal abnormalities.

19.
Hum Mutat ; 31(1): E1071-80, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19953639

RESUMEN

The "Osteopetroses" are genetic diseases whose clinical picture is caused by a defect in bone resorption by osteoclasts. Three main forms can be distinguished on the basis of severity, age of onset and means of inheritance: the dominant benign, the intermediate and the recessive severe form. While several genes have been involved in the pathogenesis of the different types of osteopetroses, the CLCN7 gene has drawn the attention of many researchers, as mutations within this gene are associated with very different phenotypes. We report here the characterization of 25 unpublished patients which has resulted in the identification of 20 novel mutations, including 11 missense mutations, 6 causing premature termination, 1 small deletion and 2 putative splice site defects. Careful analysis of clinical and molecular data led us to several conclusions. First, intermediate osteopetrosis is not homogeneous, since it can comprise both severe dominant forms with an early onset and recessive ones without central nervous system involvement. Second, the appropriateness of haematopoietic stem cell transplantation in CLCN7-dependent ARO patients has to be carefully evaluated and exhaustive CNS examination is strongly suggested, as transplantation can almost completely cure the disease in situations where no primary neurological symptoms are present. Finally, the analysis of this largest cohort of CLCN7-dependent ARO patients together with some ADO II families allowed us to draw preliminary genotype-phenotype correlations suggesting that haploinsufficiency is not the mechanism causing ADO II. The availability of biochemical assays to characterize ClC-7 function will help to confirm this hypothesis.


Asunto(s)
Canales de Cloruro/genética , Mutación , Osteopetrosis/genética , Osteopetrosis/fisiopatología , Índice de Severidad de la Enfermedad , Adulto , Edad de Inicio , Niño , Preescolar , Canales de Cloruro/química , Canales de Cloruro/metabolismo , Cristalización , Femenino , Genotipo , Humanos , Lactante , Recién Nacido , Masculino , Modelos Moleculares , Fenotipo
20.
J Clin Invest ; 117(4): 919-30, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17404618

RESUMEN

This study illustrates that Plekhm1 is an essential protein for bone resorption, as loss-of-function mutations were found to underlie the osteopetrotic phenotype of the incisors absent rat as well as an intermediate type of human osteopetrosis. Electron and confocal microscopic analysis demonstrated that monocytes from a patient homozygous for the mutation differentiated into osteoclasts normally, but when cultured on dentine discs, the osteoclasts failed to form ruffled borders and showed little evidence of bone resorption. The presence of both RUN and pleckstrin homology domains suggests that Plekhm1 may be linked to small GTPase signaling. We found that Plekhm1 colocalized with Rab7 to late endosomal/lysosomal vesicles in HEK293 and osteoclast-like cells, an effect that was dependent on the prenylation of Rab7. In conclusion, we believe PLEKHM1 to be a novel gene implicated in the development of osteopetrosis, with a putative critical function in vesicular transport in the osteoclast.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Cromosomas Humanos Par 10 , Glicoproteínas de Membrana/genética , Osteopetrosis/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Adaptadoras del Transporte Vesicular/genética , Animales , Proteínas Relacionadas con la Autofagia , Mapeo Cromosómico , Femenino , Regulación de la Expresión Génica , Humanos , Riñón/fisiología , Riñón/fisiopatología , Masculino , Glicoproteínas de Membrana/metabolismo , Monocitos/fisiología , Mutación , Especificidad de Órganos , Linaje , Ratas , Proteínas de Unión al GTP rab/metabolismo , Proteínas de Unión a GTP rab7
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA