Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Cancer Genet ; 272-273: 23-28, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36657266

RESUMEN

ETS-related gene (ERG) amplification, observed in 4-6% of acute myeloid leukemia (AML), is associated with unfavorable prognosis. To determine coincident effects of additional genomic abnormalities in AML with ERG amplification (ERGamp), we examined 11 ERGamp cases of 205 newly diagnosed AML using chromosomal microarray analysis and next generation sequencing. ERGamp cases demonstrated a distinct pattern of high genetic complexity: loss of 5q, chromothripsis and TP53 loss of function variants. Remarkably, allelic TP53 loss or loss of heterozygosity (LOH) co-occurring with TP53 inactivating mutation dramatically effected ERGamp tumor patient outcome. In the presence of homozygous TP53 loss of function, ERGamp patients demonstrated no response to induction chemotherapy with median overall survival (OS) of 3.8 months (N = 9). Two patients with heterozygous loss of TP53 function underwent alloSCT without evidence of relapse at one year. Similarly, a validation TCGA cohort, 6 of the 8 ERGamp cases with TP53 loss of function demonstrated median OS of 2.5 months. This suggests that with TP53 mutant ERGamp AML, successive loss of the second TP53 allele, typically by 17p deletion or LOH identifies a specific high-risk subtype of AML patients who are resistant to standard induction chemotherapy and need novel approaches to avert the very poor prognosis.


Asunto(s)
Leucemia Mieloide Aguda , Proteína p53 Supresora de Tumor , Humanos , Proteína p53 Supresora de Tumor/genética , Leucemia Mieloide Aguda/patología , Pérdida de Heterocigocidad , Pronóstico , Hibridación Fluorescente in Situ , Mutación/genética , Regulador Transcripcional ERG/genética
3.
Cancer Genet ; 260-261: 18-22, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34808593

RESUMEN

Richter transformation (RT) refers to the development of an aggressive lymphoma in patients with pre-existing chronic lymphocytic leukemia/small lymphocytic lymphoma (CLL/SLL). It carries a poor prognosis secondary to poor response to therapy or rapid disease relapse. Currently there are no randomized trials to guide treatment. Therapeutic decisions are often influenced by the presence or absence of a clonal relationship between the underlying CLL/SLL and the new lymphoma given the poor prognosis of patients with clonally related RT. Chromosomal microarray analysis (CMA) can help to establish clonality while also detecting genomic complexity and clinically relevant genetic variants such as loss of CDKN2A and/or TP53. As a result, CMA has potential prognostic and therapeutic implications. For this study, CMA results from patients with Richter transformation were evaluated in paired CLL/SLL and transformed lymphoma samples. CMA revealed that 86% of patients had common aberrations in the two samples indicating evidence of common clonality. CMA was also useful in detecting aberrations associated with a poor prognosis in 71% of patients with RT. This study highlights the potential clinical utility of CMA to investigate the clonal relationship between CLL/SLL and RT, provide prognostic information, and possibly guide therapeutic decision making for patients with Richter transformation.


Asunto(s)
Cromosomas Humanos/genética , Células Clonales/química , Leucemia Linfocítica Crónica de Células B/genética , Linfoma de Células B Grandes Difuso/genética , Análisis por Micromatrices/métodos , Anciano , Progresión de la Enfermedad , Femenino , Inestabilidad Genómica , Humanos , Masculino , Persona de Mediana Edad , Pronóstico , Estudios Retrospectivos
4.
Sci Rep ; 11(1): 5690, 2021 03 11.
Artículo en Inglés | MEDLINE | ID: mdl-33707600

RESUMEN

Malignant peripheral nerve sheath tumors (MPNSTs) are aggressive Schwann cell-derived neoplasms that occur sporadically or in patients with neurofibromatosis type 1 (NF1). Preclinical research on sporadic MPNSTs has been limited as few cell lines exist. We generated and characterized a new sporadic MPNST cell line, 2XSB, which shares the molecular and genomic features of the parent tumor. These cells have a highly complex karyotype with extensive chromothripsis. 2XSB cells show robust invasive 3-dimensional and clonogenic culture capability and form solid tumors when xenografted into immunodeficient mice. High-density single nucleotide polymorphism array and whole exome sequencing analyses indicate that, unlike NF1-associated MPNSTs, 2XSB cells have intact, functional NF1 alleles with no evidence of mutations in genes encoding components of Polycomb Repressor Complex 2. However, mutations in other genes implicated in MPNST pathogenesis were identified in 2XSB cells including homozygous deletion of CDKN2A and mutations in TP53 and PTEN. We also identified mutations in genes not previously associated with MPNSTs but associated with the pathogenesis of other human cancers. These include DNMT1, NUMA1, NTRK1, PDE11A, CSMD3, LRP5 and ACTL9. This sporadic MPNST-derived cell line provides a useful tool for investigating the biology and potential treatment regimens for sporadic MPNSTs.


Asunto(s)
Genoma Humano , Neoplasias de la Vaina del Nervio/genética , Neoplasias de la Vaina del Nervio/patología , Secuencias Repetitivas de Ácidos Nucleicos , Línea Celular Tumoral , Proliferación Celular , Dosificación de Gen , Genes Relacionados con las Neoplasias , Humanos , Cariotipificación , Mutación/genética , Polimorfismo de Nucleótido Simple/genética , Secuencias Repetitivas de Ácidos Nucleicos/genética , Secuenciación del Exoma
5.
Cancer Genet ; 246-247: 44-47, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32827877

RESUMEN

The 2016 World Health Organization entity 'Myeloid/lymphoid neoplasms with eosinophilia and rearrangement of PDGFRA, PDGFRB or FGFR1, or with PCM1-JAK2' encompasses a group of rare neoplasms that result from the formation of a fusion gene that leads to expression of an aberrant tyrosine kinase. This entity also contains variant JAK2 fusion partners, and detection of this defining event can be facilitated by various cytogenetic and molecular methods. Cryptic rearrangements of 9p24/JAK2 can be particularly challenging to identify. We describe the use of chromosomal microarray analysis (CMA), fluorescence in situ hybridization (FISH) with a probe for JAK2, and genomic mate pair analysis to describe a complex karyotype with a t(9;22) that produced a functional BCR-JAK2 fusion, leading to the appropriate diagnosis for the patient. This case highlights the importance of using an integrated genomic approach to fully define complex aberrations to assign proper diagnoses.


Asunto(s)
Cromosomas Humanos Par 22/genética , Cromosomas Humanos Par 9/genética , Eosinofilia/patología , Janus Quinasa 2/genética , Trastornos Mieloproliferativos/patología , Proteínas Proto-Oncogénicas c-bcr/genética , Translocación Genética , Eosinofilia/genética , Genómica/métodos , Humanos , Hibridación Fluorescente in Situ/métodos , Masculino , Análisis por Micromatrices/métodos , Persona de Mediana Edad , Trastornos Mieloproliferativos/genética , Pronóstico
6.
Genes Chromosomes Cancer ; 59(7): 422-427, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32196814

RESUMEN

Infant leukemias are a rare group of neoplasms that are clinically and biologically distinct from their pediatric and adult counterparts. Unlike leukemia in older children where survival rates are generally favorable, infants with leukemia have a 5-year event-free survival rate of <50%. The majority of infant leukemias are characterized by KMT2A (MLL) rearrangements (~70 to 80% in acute lymphoblastic leukemia), which appear to be drivers of early leukemogenesis. In this report, we describe three cases: a 9-month-old female infant with B-acute lymphoblastic leukemia (B-ALL), an 8-month-old female presenting with B/myeloid mixed phenotype acute leukemia (MPAL), and a 16-month-old male with B-ALL. The first case had a normal karyotype and B-ALL FISH results consistent with an atypical KMT2A rearrangement. The second case had trisomy 10 as the sole chromosomal abnormality and a normal KMT2A FISH result. Case 3 had trisomy 8 and a t(11;15)(q23;q21), an atypical KMT2A rearrangement by FISH studies, and a focal deletion of 15q with a breakpoint within the USP8 gene by chromosomal microarray. Mate pair sequencing was performed on all three cases and identified a KMT2A-USP2 rearrangement (cases 1 and 2) or a KMT2A-USP8 rearrangement (case 3). These recently characterized KMT2A fusions have been described exclusively in infant and pediatric leukemia cases where the incidence varies vary according to leukemia subtype, are considered high-risk, with a high incidence of central nervous system involvement, poor response to initial prednisone treatment, and poor event free survival. Additionally, approximately half of cases are unable to be resolved using standard cytogenetic approaches and are likely under recognized. Therefore, targeted molecular approaches are suggested in genetically unresolved infant leukemia cases to characterize these prognostically relevant clones.


Asunto(s)
Reordenamiento Génico , N-Metiltransferasa de Histona-Lisina/genética , Proteína de la Leucemia Mieloide-Linfoide/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Endopeptidasas/genética , Complejos de Clasificación Endosomal Requeridos para el Transporte/genética , Femenino , Pruebas Genéticas/métodos , Humanos , Lactante , Masculino , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Ubiquitina Tiolesterasa/genética
7.
Neurooncol Adv ; 2(1): vdaa155, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33392505

RESUMEN

BACKGROUND: Giant cell glioblastoma (gcGBM) is a rare histologic subtype of glioblastoma characterized by numerous bizarre multinucleate giant cells and increased reticulin deposition. Compared with conventional isocitrate dehydrogenase (IDH)-wildtype glioblastomas, gcGBMs typically occur in younger patients and are generally associated with an improved prognosis. Although prior studies of gcGBMs have shown enrichment of genetic events, such as TP53 alterations, no defining aberrations have been identified. The aim of this study was to evaluate the genomic profile of gcGBMs to facilitate more accurate diagnosis and prognostication for this entity. METHODS: Through a multi-institutional collaborative effort, we characterized 10 gcGBMs by chromosome studies, single nucleotide polymorphism microarray analysis, and targeted next-generation sequencing. These tumors were subsequently compared to the genomic and epigenomic profile of glioblastomas described in The Cancer Genome Atlas (TCGA) dataset. RESULTS: Our analysis identified a specific pattern of genome-wide massive loss of heterozygosity (LOH) driven by near haploidization in a subset of glioblastomas with giant cell histology. We compared the genomic signature of these tumors against that of all glioblastomas in the TCGA dataset (n = 367) and confirmed that our cohort of gcGBMs demonstrated a significantly different genomic profile. Integrated genomic and histologic review of the TCGA cohort identified 3 additional gcGBMs with a near haploid genomic profile. CONCLUSIONS: Massive LOH driven by haploidization represents a defining molecular hallmark of a subtype of gcGBM. This unusual mechanism of tumorigenesis provides a diagnostic genomic hallmark to evaluate in future cases, may explain reported differences in survival, and suggests new therapeutic vulnerabilities.

8.
Ophthalmic Genet ; 40(2): 110-117, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30870047

RESUMEN

BACKGROUND: Leber congenital amaurosis (LCA) and early-onset retinal dystrophy (EORD), are primary causes of inherited childhood blindness. Both are autosomal recessive diseases, with mutations in more than 25 genes explaining approximately ~70% of cases. However, the genetic cause for many cases remains unclear. Sequencing studies from genetically isolated populations with increased prevalence of a disorder has proven useful for rare variant studies, making Costa Rica an ideal place to study LCA/EORD genetics. MATERIALS AND METHODS: Twenty-eight affected children (25 LCA, three EORD) and their immediate family members, totaling 52 individuals (30 affected) from 22 families, were sequenced. Whole exome sequencing was performed on all affected individuals. Available parents were analyzed either by whole exome sequencing (WES) or Sanger sequencing to determine transmission. RESULTS: All affected individuals demonstrated compound heterozygous or homozygous mutations in known Inherited Retinal Disease (IRD) associated genes. Twelve variants were identified in at least one individual in three genes, RDH12, RPE65, and USH2A. Four recurrent RPE65 mutations were observed in 97% of individuals and 95% of families. All patients with LCA and two of the three individuals with EORD had biallelic mutations in RPE65; one child with EORD had a homozygous RDH12 mutation. CONCLUSIONS: These data suggest that the majority of LCA/EORD in Costa Rica is due to four founder mutations in RPE65 which have been maintained in this genetically isolated population. This finding is of great clinical significance due to the availability of gene therapy recently approved in the US and European Union for patients with biallelic RPE65 defects.


Asunto(s)
Mutación del Sistema de Lectura , Amaurosis Congénita de Leber/genética , Mutación Missense , Distrofias Retinianas/genética , cis-trans-Isomerasas/genética , Adolescente , Oxidorreductasas de Alcohol/genética , Niño , Preescolar , Costa Rica/epidemiología , Electrorretinografía , Femenino , Efecto Fundador , Humanos , Lactante , Amaurosis Congénita de Leber/epidemiología , Amaurosis Congénita de Leber/fisiopatología , Masculino , Prevalencia , Retina/fisiopatología , Distrofias Retinianas/epidemiología , Distrofias Retinianas/fisiopatología , Secuenciación del Exoma
9.
Methods Mol Biol ; 1908: 89-111, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30649723

RESUMEN

The basis of cancer biology is built upon two fundamental processes that result in uncontrolled cell proliferation and tumor formation: loss of tumor suppressor gene function and gain of oncogene function. Somatic DNA copy number variants (CNVs), which generally range in size from kilobases to entire chromosomes, facilitate gains and losses of chromosomal material incorporating oncogenes and tumor suppressor genes, respectively. In fact, many cancer types are characterized by DNA copy number changes and relatively few single nucleotide mutations (Ciriello et al. Nat Genet 45:1127-1133, 2013). Currently, the optimal method to detect such somatic copy number changes across the cancer genome is whole-genome single nucleotide polymorphism (SNP) microarray analysis.


Asunto(s)
Variaciones en el Número de Copia de ADN , Pérdida de Heterocigocidad , Neoplasias/genética , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , Análisis de Secuencia de ADN/métodos , Genómica/métodos , Humanos , Mutación , Adhesión en Parafina , Polimorfismo de Nucleótido Simple , Fijación del Tejido
10.
Acta Neuropathol Commun ; 6(1): 107, 2018 10 19.
Artículo en Inglés | MEDLINE | ID: mdl-30340515

RESUMEN

Endolymphatic sac tumor (ELST) is a rare neoplasm arising in the temporal petrous region thought to originate from endolymphatic sac epithelium. It may arise sporadically or in association with Von-Hippel-Lindau syndrome (VHL). The ELST prevalence in VHL ranges from 3 to 16% and may be the initial presentation of the disease. Onset is usually in the 3rd to 5th decade with hearing loss and an indolent course. ELSTs present as locally destructive lesions with characteristic computed tomography imaging features. Histologically, they show papillary, cystic or glandular architectures. Immunohistochemically, they express keratin, EMA, and variably S100 and GFAP. Currently it is recommended that, given its rarity, ELST needs to be differentiated from other entities with similar morphologic patterns, particularly other VHL-associated neoplasms such as metastatic clear cell renal cell carcinoma (ccRCC). Nineteen ELST cases were studied. Immunohistochemistry (18/19) and single nucleotide polymorphism microarray testing was performed (12/19). Comparison with the immunophenotype and copy number profile in RCC is discussed. Patients presented with characteristic bone destructive lesions in the petrous temporal bones. Pathology of tumors showed characteristic ELST morphology with immunoexpression of CK7, GFAP, S100, PAX-8, PAX-2, CA-9 in the tumor cells. Immunostaines for RCC, CD10, CK20, chromogranin A, synaptophysin, TTF-1, thyroglobulin, and transthyretin were negative in the tumor cells. Molecular testing showed loss of 3p and 9q in 66% (8/12) and 58% (7/12) cases, respectively. Immunoreactivity for renal markers in ELST is an important diagnostic caveat and has not been previously reported. In fact, renal markers are currently recommended in order to rule out metastatic RCC although PAX gene complex and CA-9 have been implicated in the development of the inner ear. Importantly copy number assessment of ELST has not been previously reported. Loss of 3p (including the VHL locus) in ELST suggests similar mechanistic origins as ccRCC.


Asunto(s)
Carcinoma de Células Renales/diagnóstico , Neoplasias del Oído/diagnóstico , Saco Endolinfático/patología , Neoplasias Renales/diagnóstico , Proteínas de Neoplasias/metabolismo , Adolescente , Adulto , Carcinoma de Células Renales/cirugía , Estudios de Cohortes , Biología Computacional , Citocinas/genética , Citocinas/metabolismo , Neoplasias del Oído/cirugía , Saco Endolinfático/diagnóstico por imagen , Saco Endolinfático/cirugía , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Queratinas/metabolismo , Neoplasias Renales/cirugía , Imagen por Resonancia Magnética , Masculino , Persona de Mediana Edad , Proteínas del Tejido Nervioso/metabolismo , Factor de Transcripción PAX2/metabolismo , Adulto Joven
11.
Pediatr Dev Pathol ; 21(6): 561-567, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29216801

RESUMEN

The SOX10 gene plays a vital role in neural crest cell development and migration. Abnormalities in SOX10 are associated with Waardenburg syndrome Types II and IV, and these patients have recognizable clinical features. This case report highlights the first ever reported homozygous loss of function of the SOX10 gene in a human. This deletion is correlated using family history, prenatal ultrasound, microarray analysis of amniotic fluid, and ultimately, a medical autopsy examination to further elucidate phenotypic effects of this genetic variation. Incorporating the use of molecular pathology into the autopsy examination of fetuses with suspected congenital anomalies is vital for appropriate family counseling, and with the ability to use formalin-fixed and paraffin-embedded tissues, has become a practical approach in autopsy pathology.


Asunto(s)
Homocigoto , Mutación con Pérdida de Función , Diagnóstico Prenatal/métodos , Factores de Transcripción SOXE/genética , Síndrome de Waardenburg/diagnóstico , Autopsia , Resultado Fatal , Femenino , Marcadores Genéticos , Humanos , Fenotipo , Embarazo , Síndrome de Waardenburg/genética , Adulto Joven
13.
Cancer Genet ; 208(11): 525-36, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26454669

RESUMEN

Cytogenomic microarray analysis (CMA) offers high resolution, genome-wide copy number information and is widely used in clinical laboratories for diagnosis of constitutional abnormalities. The Cancer Genomics Consortium (CGC) conducted a multiplatform, multicenter clinical validation project to compare the reliability and inter- and intralaboratory reproducibility of this technology for clinical oncology applications. Four specimen types were processed on three different microarray platforms-from Affymetrix, Agilent, and Illumina. Each microarray platform was employed at two independent test sites. The results were compared in a blinded manner with current standard methods, including karyotype, FISH, or morphology. Twenty-nine chronic lymphocytic leukemia blood, 34 myelodysplastic syndrome bone marrow, and 30 fresh frozen renal epithelial tumor samples were assessed by all six laboratories. Thirty formalin fixed paraffin embedded renal tumor samples were analyzed at the Affymetrix and Agilent test sites only. All study samples were initial diagnostic samples. Array data were analyzed at each participating site and were submitted to caArray for central analysis. Laboratory interpretive results were submitted to the central analysis team for comparison with the standard-of-care assays and for calculation of intraplatform reproducibility and cross-platform concordance. The results demonstrated that the three microarray platforms 1) detect clinically actionable genomic changes in cancer compatible to standard-of-care methods; 2) further define cytogenetic aberrations; 3) identify submicroscopic alterations and loss of heterozygosity (LOH); and 4) yield consistent results within and between laboratories. Based on this study, the CGC concludes that CMA is a sensitive and reliable technique for copy number and LOH assessment that may be used for clinical oncology genomic analysis.


Asunto(s)
Hibridación Genómica Comparativa/métodos , Análisis Citogenético/métodos , Neoplasias/diagnóstico , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , Aberraciones Cromosómicas , Dosificación de Gen , Humanos , Hibridación Fluorescente in Situ , Cariotipo , Neoplasias Renales/diagnóstico , Neoplasias Renales/genética , Leucemia Linfocítica Crónica de Células B/diagnóstico , Leucemia Linfocítica Crónica de Células B/genética , Pérdida de Heterocigocidad , Síndromes Mielodisplásicos/diagnóstico , Síndromes Mielodisplásicos/genética , Neoplasias/genética , Neoplasias Glandulares y Epiteliales/diagnóstico , Neoplasias Glandulares y Epiteliales/genética , Reproducibilidad de los Resultados , Nivel de Atención
14.
Cancer Genet ; 204(12): 654-65, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22285017

RESUMEN

Chronic lymphocytic leukemia (CLL) is a clinically heterogeneous disease. The methods currently used for monitoring CLL and determining conditions for treatment are limited in their ability to predict disease progression, patient survival, and response to therapy. Although clonal diversity and the acquisition of new chromosomal abnormalities during the disease course (clonal evolution) have been associated with disease progression, their prognostic potential has been underappreciated because cytogenetic and fluorescence in situ hybridization (FISH) studies have a restricted ability to detect genomic abnormalities and clonal evolution. We hypothesized that whole genome analysis using high resolution single nucleotide polymorphism (SNP) microarrays would be useful to detect diversity and infer clonal evolution to offer prognostic information. In this study, we used the Infinium Omni1 BeadChip (Illumina, San Diego, CA) array for the analysis of genetic variation and percent mosaicism in 25 non-selected CLL patients to explore the prognostic value of the assessment of clonal diversity in patients with CLL. We calculated the percentage of mosaicism for each abnormality by applying a mathematical algorithm to the genotype frequency data and by manual determination using the Simulated DNA Copy Number (SiDCoN) tool, which was developed from a computer model of mosaicism. At least one genetic abnormality was identified in each case, and the SNP data was 98% concordant with FISH results. Clonal diversity, defined as the presence of two or more genetic abnormalities with differing percentages of mosaicism, was observed in 12 patients (48%), and the diversity correlated with the disease stage. Clonal diversity was present in most cases of advanced disease (Rai stages III and IV) or those with previous treatment, whereas 9 of 13 patients without detected clonal diversity were asymptomatic or clinically stable. In conclusion, SNP microarray studies with simultaneous evaluation of genomic alterations and mosaic distribution of clones can be used to assess apparent clonal evolution via analysis of clonal diversity. Since clonal evolution in CLL is strongly correlated with disease progression, whole genome SNP microarray analysis provides a new comprehensive and reliable prognostic tool for CLL patients.


Asunto(s)
Leucemia Linfocítica Crónica de Células B/diagnóstico , Análisis de Secuencia por Matrices de Oligonucleótidos , Polimorfismo de Nucleótido Simple , Adulto , Anciano , Femenino , Genotipo , Humanos , Hibridación Fluorescente in Situ , Leucemia Linfocítica Crónica de Células B/genética , Masculino , Persona de Mediana Edad , Mosaicismo , Pronóstico
15.
Cancer Genet Cytogenet ; 183(1): 64-8, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18474300

RESUMEN

Tetraploidy is a very rare cytogenetic abnormality in myelocytic malignancies, and its significance is unclear to date. We report here on a 68-year-old male diagnosed with myelodysplastic syndrome/refractory anemia with excess blasts (MDS/RAEB). Cytogenetic analysis of his bone marrow biopsy at initial clinical presentation and in subsequent studies revealed the presence of two abnormal clones, 92,XXYY and 92,XXYY,del(5)(q13q33). Interphase fluorescence in situ hybridization analysis of abnormal cells confirmed interstitial deletion in 5q, demonstrated predominance of the tetraploid clone and persistent presence of the tetraploid clone with 5q deletion. The patient was not responsive to Revlimid (lenalidomide) treatment, which is routinely used in patients with 5q- syndrome. However, a subsequent course of therapy with the methyl-transferase inhibitor decitabine resulted in clinical and cytogenetic remission. Our data suggest that the unique complex abnormality of tetraploidy and 5q deletion described here for the first time in MDS is characterized by distinct disease etiology, the mechanism of which could involve epigenetic inactivation of gene expression via methylation.


Asunto(s)
Deleción Cromosómica , Cromosomas Humanos Par 5 , Síndromes Mielodisplásicos/genética , Poliploidía , Anciano , Bandeo Cromosómico , Humanos , Hibridación Fluorescente in Situ , Masculino
16.
Mol Cell Biochem ; 282(1-2): 89-99, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16317516

RESUMEN

Activation of hepatic stellate cells from quiescence to myofibroblast-like cells (MFBs) is a pivotal event in hepatic fibrogenesis. Plastic-cultured stellate cells (an established in vitro model of the activated phenotype) recultured on Matrigel revert to quiescence. In the present study we analyzed the molecular mechanism underlying this process, focusing on the effect of collagen receptors alpha(2)beta(1) and alpha(1)beta(1) integrin signaling on the expression of Ets-1 transcription factor and its target gene MMP1 in cultured human MFBs. Cells grown in 3-dimensional (3D) substrates (Matrigel) or collagen type I gel) markedly upregulated Ets-1 and MMP1 messages, in comparison to cells cultured on plastic. A similar effect but less intense was mimicked by stimulation of alpha(2)beta(1) or blocking of alpha(1)beta(1) integrin in cells grown on plastic. We observed increased expression of MMP1 transcripts with parallel changes in MMP1 promoter activity, and in mRNA and protein levels of upstream transcription factors Ets-1 and c-Jun. Interference with alpha(2)beta(1) and alpha(1)beta(1) integrin function in cells cultured in a 3D collagen substrate resulted in an even greater effect. Morphologically, stimulation of alpha(2)beta(1) integrin resulted in formation of multicellular networks, probably by facilitation of cell migration. Thus, we report the novel observation that in cultured human MFBs reverting to quiescence, the expression of transcription factor Ets-1 and its downstream target MMP1 can be modulated by changes in the microenvironment, which are mediated, at least in part, by the balance between collagen receptor integrin alpha(2)beta(1) and alpha(1)beta(1) activities.


Asunto(s)
Colágeno Tipo I/metabolismo , Fibroblastos/metabolismo , Hepatocitos/metabolismo , Integrina alfa1beta1/metabolismo , Integrina alfa2beta1/metabolismo , Proteína Proto-Oncogénica c-ets-1/biosíntesis , Diferenciación Celular , Células Cultivadas , Colágeno/metabolismo , Combinación de Medicamentos , Fibroblastos/citología , Hepatocitos/citología , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Laminina/metabolismo , Metaloproteinasa 1 de la Matriz/genética , Metaloproteinasa 1 de la Matriz/metabolismo , Regiones Promotoras Genéticas , Unión Proteica , Proteoglicanos/metabolismo , Transducción de Señal , Regulación hacia Arriba
17.
J Hepatol ; 43(5): 893-900, 2005 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16169119

RESUMEN

BACKGROUND/AIMS: In the cirrhotic liver, gene expression of the multifunctional cytokine oncostatin M (OSM) is up-regulated, but its cellular origin is unknown. Therefore, we investigated the expression of OSM protein and its specific receptor subunits, OSMRbeta and LIFRbeta in normal and cirrhotic human liver using immunohistochemical and Western blot analysis. RESULTS: OSM protein was expressed in Kupffer cells, variably in normal liver but consistently in cirrhosis. OSMRbeta was expressed at low level in hepatocytes of all normal livers examined, but in no cirrhotic sample. In contrast, LIFRbeta receptor was expressed weakly in normal livers, but much more intensely in cirrhosis, in reactive ductules, bile duct epithelial cells and perisinusoidal areas. Double immunostaining showed co-localization of LIFRbeta with cytokeratin 7, proliferating cell nuclear antigen (PCNA) and leukemia inhibitory factor (LIF), in bile duct epithelial cells, but not with alpha-smooth muscle actin, a myofibroblast marker. CONCLUSIONS: In human liver, OSM protein is expressed in Kupffer cells, variably in normals but universally in cirrhosis. The differential expression pattern of OSM and its receptors could allow for differential OSM signaling by alternative utilization of receptors to promote hepatocyte proliferation in acute injury and, with its homologue LIF, for the bile ductular reaction in cirrhosis.


Asunto(s)
Expresión Génica , Inhibidores de Crecimiento/metabolismo , Cirrosis Hepática/metabolismo , Péptidos/metabolismo , Receptores de Citocinas/metabolismo , Animales , Antineoplásicos/metabolismo , Hepatocitos/citología , Hepatocitos/metabolismo , Humanos , Mediadores de Inflamación/metabolismo , Queratina-7 , Queratinas/metabolismo , Subunidad alfa del Receptor del Factor Inhibidor de Leucemia , Hígado/citología , Hígado/metabolismo , Cirrosis Hepática/patología , Oncostatina M , Subunidades de Proteína/metabolismo , Receptores OSM-LIF , Receptores de Oncostatina M
18.
Anat Rec A Discov Mol Cell Evol Biol ; 283(1): 182-6, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15674824

RESUMEN

Oncostatin M (OSM), a member of the interleukin-6 family of cytokines, is thought to be expressed mostly by activated T-lymphocytes and monocytes in adult animals. However, here we report specific constitutive tissue expression of OSM in the pancreas, kidney, testes, spleen, stomach, and brain, but not liver or lung, of three adult rodent species.


Asunto(s)
Citocinas/metabolismo , Péptidos/metabolismo , Roedores/metabolismo , Animales , Femenino , Gerbillinae , Edad Gestacional , Técnicas para Inmunoenzimas , Masculino , Ratones , Oncostatina M , Ratas , Ratas Wistar , Especificidad de la Especie
19.
J Hepatol ; 37(2): 214-21, 2002 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12127426

RESUMEN

BACKGROUND: Liver injury transforms hepatic stellate cells into myofibroblast (MFB)-like cells. With recovery from injury, MFBs undergo apoptosis, but it is unknown whether they can also revert to quiescence. AIM: To determine whether human (h)MFBs become quiescent if cultured on a basement membrane-like substrate (Matrigel). METHODS: hMFBs obtained from cirrhotic liver were re-cultured on plastic or Matrigel. Expression of genes of collagen metabolism was assayed before and after transforming growth factor beta (TGFbeta) and Oncostatin M (OSM) stimulation. RESULTS: hMFBs had typical MFB-like morphology, with abundant alpha-smooth muscle actin (SMA) but no cytoplasmic lipid droplets. hMFBs re-cultured on Matrigel reverted to alphaSMA-negative, lipid droplet-positive quiescent morphology. alphaSMA, collagen alpha1(1) (COL1A1) and collagen alpha2(1) (COL1A2) messages were upregulated in hMFBs cultured on plastic, but suppressed by Matrigel. The opposite was true for metalloproteinase-1 mRNA. OSM but not TGFbeta reduced alphaSMA mRNA by 30% while TGFbeta but not OSM upregulated COL1A1 mRNA by 48%, in hMFBs on plastic. TGFbeta and OSM stimulated COL1A1 gene expression in Matrigel by 50 and 60%, respectively. CONCLUSIONS: Matrigel culture de-activates hMFBs yet collagen gene expression still responds to fibrogenic cytokines. The responses of hMFB gene expression to TGFbeta and OSM, are regulated differently by the extracellular matrix.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Colágeno Tipo I , Fibroblastos/citología , Fibroblastos/fisiología , Hepatocitos/citología , Hepatocitos/fisiología , Actinas/genética , Antineoplásicos/farmacología , Membrana Basal , Materiales Biocompatibles/farmacología , Células Cultivadas , Colágeno/genética , Colágeno/farmacología , Cadena alfa 1 del Colágeno Tipo I , Combinación de Medicamentos , Expresión Génica , Humanos , Laminina/farmacología , Metaloproteinasa 1 de la Matriz/genética , Oncostatina M , Péptidos/farmacología , Plásticos , Proteoglicanos/farmacología , ARN Mensajero/análisis , Inhibidor Tisular de Metaloproteinasa-1/genética , Factor de Crecimiento Transformador beta/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...