Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Z Rheumatol ; 83(5): 393-400, 2024 Jun.
Artículo en Alemán | MEDLINE | ID: mdl-38802503

RESUMEN

Lysosomal storage diseases are a group of rare hereditary metabolic diseases. Due to a deficiency of lysosomal enzymes, complex substrates accumulate in the lysosomes of various organs. Depending on the affected enzyme, this results in clinically variable and chronic progressive multiorgan diseases. Diagnosis is often delayed. As clinical symptoms include the musculoskeletal system, an awareness of lysosomal storage diseases is of relevance to (pediatric) rheumatologists. This article is focused on Mucopolysaccharidosis type I­S, Mucolipidosis type III, Gaucher disease and Fabry disease. When suspecting a lysosomal storage disease, enzyme activity should be determined in dried blood spots or leukocytes. For some diseases, specific biomarkers can additionally be analyzed. Diagnosis should be confirmed by genetic testing. As causal treatment options are available for three of the presented diseases, a timely diagnosis is very important.


Asunto(s)
Enfermedades por Almacenamiento Lisosomal , Enfermedades Reumáticas , Humanos , Enfermedades por Almacenamiento Lisosomal/diagnóstico , Enfermedades por Almacenamiento Lisosomal/genética , Enfermedades Reumáticas/diagnóstico , Enfermedades Reumáticas/genética , Enfermedades Reumáticas/sangre , Reumatología , Diagnóstico Diferencial , Medicina Basada en la Evidencia
3.
PLoS Genet ; 11(5): e1005239, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-26000857

RESUMEN

In vitro disease modeling based on induced pluripotent stem cells (iPSCs) provides a powerful system to study cellular pathophysiology, especially in combination with targeted genome editing and protocols to differentiate iPSCs into affected cell types. In this study, we established zinc-finger nuclease-mediated genome editing in primary fibroblasts and iPSCs generated from a mouse model for radiosensitive severe combined immunodeficiency (RS-SCID), a rare disorder characterized by cellular sensitivity to radiation and the absence of lymphocytes due to impaired DNA-dependent protein kinase (DNA-PK) activity. Our results demonstrate that gene editing in RS-SCID fibroblasts rescued DNA-PK dependent signaling to overcome radiosensitivity. Furthermore, in vitro T-cell differentiation from iPSCs was employed to model the stage-specific T-cell maturation block induced by the disease causing mutation. Genetic correction of the RS-SCID iPSCs restored T-lymphocyte maturation, polyclonal V(D)J recombination of the T-cell receptor followed by successful beta-selection. In conclusion, we provide proof that iPSC-based in vitro T-cell differentiation is a valuable paradigm for SCID disease modeling, which can be utilized to investigate disorders of T-cell development and to validate gene therapy strategies for T-cell deficiencies. Moreover, this study emphasizes the significance of designer nucleases as a tool for generating isogenic disease models and their future role in producing autologous, genetically corrected transplants for various clinical applications.


Asunto(s)
Diferenciación Celular , Proteína Quinasa Activada por ADN/metabolismo , Proteínas de Unión al ADN/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Quinasas/metabolismo , Transducción de Señal , Linfocitos T/citología , Animales , Proteína Quinasa Activada por ADN/deficiencia , Proteína Quinasa Activada por ADN/genética , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Modelos Animales de Enfermedad , Fibroblastos/citología , Fibroblastos/metabolismo , Genoma , Técnicas de Genotipaje , Células HEK293 , Humanos , Células Madre Pluripotentes Inducidas/citología , Masculino , Ratones , Células 3T3 NIH , Proteínas Nucleares/deficiencia , Proteínas Nucleares/genética , Fenotipo , Proteínas Quinasas/genética , Linfocitos T/metabolismo , Dedos de Zinc
4.
Blood ; 121(5): 812-21, 2013 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-23118218

RESUMEN

UNLABELLED: Survival of chronic lymphocytic leukemia (CLL) cells depends on stimuli provided by a suitable microenvironment. The factors and mechanisms providing this growth support for CLL cells are not fully understood. We found that plasma levels of macrophage migration inhibitory factor (MIF), a proinflammatory and immunoregulatory chemokine, were elevated in CLL patients. Therefore, we characterized the functional role of MIF in a CLL mouse model. For this purpose, we crossed Eµ-TCL1 mice with MIF knockout (MIF-/-) mice. The resulting TCL1+/wtMIF/ mice showed a delayed onset of leukemia, reduced splenomegaly and hepatomegaly, and a longer survival than TCL1+/wtMIFwt/wt controls. Immunohistochemical examination of the lymphoid organs showed that the numbers of macrophages were significantly reduced in the spleen and bone marrow of TCL1+/wtMIF/ mice compared with TCL1+/wtMIFwt/wt controls. Mechanistic studies in vitro revealed that the absence of MIF rendered CLL cells more susceptible to apoptosis. Accordingly, incubation with an anti-MIF antibody reduced the survival of CLL cells on a macrophage feeder layer. In addition, the migratory activity of TCL1+/wtMIF/ macrophages was decreased compared with TCL1+/wtMIFwt/wt macrophages. Taken together, our results provide evidence that MIF supports the development of CLL by enhancing the interaction of CLL cells with macrophages. KEY POINTS: Targeted deletion of the gene for macrophage migration inhibitory factor (MIF) delays development of chronic lymphocytic leukemia and prolongs survival in mice. MIF recruits leukemia-associated macrophages to spleen or liver.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Comunicación Celular/inmunología , Oxidorreductasas Intramoleculares/inmunología , Leucemia Linfocítica Crónica de Células B/inmunología , Factores Inhibidores de la Migración de Macrófagos/inmunología , Macrófagos/inmunología , Animales , Linfocitos T CD8-positivos/patología , Supervivencia Celular , Células Nutrientes , Humanos , Oxidorreductasas Intramoleculares/genética , Leucemia Linfocítica Crónica de Células B/genética , Leucemia Linfocítica Crónica de Células B/patología , Factores Inhibidores de la Migración de Macrófagos/genética , Macrófagos/patología , Ratones , Ratones Noqueados , Neoplasias Experimentales/genética , Neoplasias Experimentales/inmunología , Neoplasias Experimentales/patología , Células Tumorales Cultivadas
5.
PLoS Biol ; 9(7): e1001099, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21765802

RESUMEN

Using the murine model of tyrosinemia type 1 (fumarylacetoacetate hydrolase [FAH] deficiency; FAH⁻/⁻ mice) as a paradigm for orphan disorders, such as hereditary metabolic liver diseases, we evaluated fibroblast-derived FAH⁻/⁻-induced pluripotent stem cells (iPS cells) as targets for gene correction in combination with the tetraploid embryo complementation method. First, after characterizing the FAH⁻/⁻ iPS cell lines, we aggregated FAH⁻/⁻-iPS cells with tetraploid embryos and obtained entirely FAH⁻/⁻-iPS cell-derived mice that were viable and exhibited the phenotype of the founding FAH⁻/⁻ mice. Then, we transduced FAH cDNA into the FAH⁻/⁻-iPS cells using a third-generation lentiviral vector to generate gene-corrected iPS cells. We could not detect any chromosomal alterations in these cells by high-resolution array CGH analysis, and after their aggregation with tetraploid embryos, we obtained fully iPS cell-derived healthy mice with an astonishing high efficiency for full-term development of up to 63.3%. The gene correction was validated functionally by the long-term survival and expansion of FAH-positive cells of these mice after withdrawal of the rescuing drug NTBC (2-(2-nitro-4-fluoromethylbenzoyl)-1,3-cyclohexanedione). Furthermore, our results demonstrate that both a liver-specific promoter (transthyretin, TTR)-driven FAH transgene and a strong viral promoter (from spleen focus-forming virus, SFFV)-driven FAH transgene rescued the FAH-deficiency phenotypes in the mice derived from the respective gene-corrected iPS cells. In conclusion, our data demonstrate that a lentiviral gene repair strategy does not abrogate the full pluripotent potential of fibroblast-derived iPS cells, and genetic manipulation of iPS cells in combination with tetraploid embryo aggregation provides a practical and rapid approach to evaluate the efficacy of gene correction of human diseases in mouse models.


Asunto(s)
Fibroblastos/efectos de los fármacos , Prueba de Complementación Genética/métodos , Terapia Genética/métodos , Vectores Genéticos/farmacología , Hidrolasas , Células Madre Pluripotentes Inducidas , Lentivirus , Tirosinemias , Animales , Supervivencia Celular , Células Cultivadas , Cromosomas/química , Ciclohexanonas/farmacología , Modelos Animales de Enfermedad , Femenino , Feto , Fibroblastos/citología , Humanos , Hidrolasas/deficiencia , Hidrolasas/genética , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/fisiología , Hígado/metabolismo , Hígado/patología , Ratones , Ratones Noqueados , Nitrobenzoatos/farmacología , Embarazo , Regiones Promotoras Genéticas , Virus Formadores de Foco en el Bazo/química , Virus Formadores de Foco en el Bazo/genética , Tetraploidía , Tirosinemias/genética , Tirosinemias/metabolismo , Tirosinemias/patología , Tirosinemias/terapia
6.
Genes Chromosomes Cancer ; 52(4): 423-30, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23340989

RESUMEN

Suv39h1 mediates heterochromatin formation in pericentric and telomeric regions by trimethylation of lysine 9 of histone 3 (H3K9me3). Yet, its role in the induction of chromosomal instability is poorly understood. We established a leukemia model by retrovirally expressing Myc in wild-type and histone methyltransferase Suv39h1-deficient hematopoietic cells and characterized the resulting leukemias for chromosomal instability. All mice that received cells overexpressing Myc developed myeloid leukemia with a median survival of 44 days posttransplantation. Myc-overexpressing wild-type leukemias demonstrated clones with numerical chromosomal aberrations (5/16). In secondary transplantations of these leukemic cells, structural changes, mostly end-to-end fusions of chromosomes, appeared (10/12). In contrast, leukemic cells overexpressing Myc with reduced or no Suv39h1 expression had a normal karyotype in primary, secondary, and tertiary transplantations (16/16). Myc-transduced Suv39h1-deficient cells showed less critically short telomeres (P < 0.05) compared with Myc-transduced wild-type bone marrow cells. Gene expression analysis showed upregulation of genes involved in the alternative lengthening of telomeres (ALT) mechanism. Thus, we hypothesize that loss of Suv39h1 implies activation of the ALT mechanism, in turn ensuring telomere length and stability. Our data show for the first time that Suv39h1 deficiency may prevent chromosomal instability by more efficient telomere stabilization in hematopoietic bone marrow cells overexpressing Myc.


Asunto(s)
Inestabilidad Cromosómica , Leucemia Mieloide/genética , Metiltransferasas/genética , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Represoras/genética , Animales , Células de la Médula Ósea/metabolismo , Trasplante de Médula Ósea , Femenino , Perfilación de la Expresión Génica , Regulación Leucémica de la Expresión Génica , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Hibridación Fluorescente in Situ , Leucemia Mieloide/metabolismo , Leucemia Mieloide/patología , Masculino , Metiltransferasas/deficiencia , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Proteínas Proto-Oncogénicas c-myc/metabolismo , Proteínas Represoras/deficiencia , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Cariotipificación Espectral , Telómero/genética , Homeostasis del Telómero/genética , Acortamiento del Telómero/genética
7.
Mol Ther ; 20(6): 1187-95, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22472950

RESUMEN

Integrating vectors developed on the basis of various retroviruses have demonstrated therapeutic potential following genetic modification of long-lived hematopoietic stem and progenitor cells. Lentiviral vectors (LV) are assumed to circumvent genotoxic events previously observed with γ-retroviral vectors, due to their integration bias to transcription units in comparison to the γ-retroviral preference for promoter regions and CpG islands. However, recently several studies have revealed the potential for gene activation by LV insertions. Here, we report a murine acute B-lymphoblastic leukemia (B-ALL) triggered by insertional gene inactivation. LV integration occurred into the 8th intron of Ebf1, a major regulator of B-lymphopoiesis. Various aberrant splice variants could be detected that involved splice donor and acceptor sites of the lentiviral construct, inducing downregulation of Ebf1 full-length message. The transcriptome signature was compatible with loss of this major determinant of B-cell differentiation, with partial acquisition of myeloid markers, including Csf1r (macrophage colony-stimulating factor (M-CSF) receptor). This was accompanied by receptor phosphorylation and STAT5 activation, both most likely contributing to leukemic progression. Our results highlight the risk of intragenic vector integration to initiate leukemia by inducing haploinsufficiency of a tumor suppressor gene. We propose to address this risk in future vector design.


Asunto(s)
Vectores Genéticos , Haploinsuficiencia , Lentivirus/genética , Leucemia/genética , Transactivadores/genética , Integración Viral , Animales , Análisis por Conglomerados , Regulación hacia Abajo/genética , Femenino , Perfilación de la Expresión Génica , Regulación Leucémica de la Expresión Génica , Orden Génico , Inestabilidad Genómica , Células Madre Hematopoyéticas/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Mutagénesis Insercional , Leucemia-Linfoma Linfoblástico de Células Precursoras B/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras B/metabolismo , Procesamiento Postranscripcional del ARN , Factor de Transcripción STAT5/metabolismo , Transactivadores/metabolismo , Transcripción Genética , Transducción Genética
8.
Chromosome Res ; 19(7): 857-68, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22009222

RESUMEN

Induced pluripotent stem (iPS) cells have greatly provoked people's interest due to their enormous potential of clinical applications. Increasing care is taken with the genetic safety of iPS cells. However, up to now, the chromosomal integrity of murine iPS (miPS) cells has been largely unknown. We have observed recurrent trisomy and/or Robertsonian translocation (Rb) of chromosome 14 in six out of nine independent miPS cell lines from three laboratories by G-banding, fluorescence in situ hybridization (FISH) and spectral karyotyping (SKY) analyses, while all the miPS cell lines were derived from mouse embryonic fibroblasts (MEFs) or neural precursor cells (NPCs) with a normal karyotype. The miPS cells with trisomy and/or Rb of chromosome 14 showed growth advantage over the miPS cells with a normal karyotype. We found a significantly higher frequency of Rbs in the miPS cell lines induced with c-Myc than those without c-Myc. Our findings demonstrate that miPS cell lines have the propensity for chromosomal aberrations and there is an obvious correlation between the extent of chromosomal aberrations in miPS cells and the transcriptional factors used for their reprogramming. Therefore, our study raises awareness of the need for improvements of the induction conditions of miPS cells in order to avoid the chromosomal aberrations and ensure future safe applications.


Asunto(s)
Cromosomas/genética , Hibridación Fluorescente in Situ/métodos , Células Madre Pluripotentes Inducidas/citología , Cariotipificación/métodos , Translocación Genética , Trisomía , Animales , Diferenciación Celular , Línea Celular , Bandeo Cromosómico , Cromosomas/química , Fibroblastos/citología , Fibroblastos/metabolismo , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Células Madre Pluripotentes Inducidas/metabolismo , Cariotipo , Ratones , Neuronas/citología , Neuronas/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Proteínas Proto-Oncogénicas c-myc/farmacología
9.
Exp Cell Res ; 317(13): 1885-94, 2011 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-21570390

RESUMEN

Pluripotent cells referred to as embryonic germ cells (EGCs) can be derived from the embryonic precursors of the mature gametes: the primordial germ cells (PGCs). A homozygous mutation (ter) of the dead-end homolog 1 gene (Dnd1) in the rat causes gonadal teratocarcinogenesis and sterility due to neoplastic transformation and loss of germ cells. We mated heterozygous ter/+ WKY-Dnd1(ter)/Ztm rats and were able to cultivate the first genital ridge-derived EGCs of the rat embryo at day 14.5 post coitum (pc). Genotyping revealed that ten EGC lines were Dnd1 deficient, while only one wild type cell line had survived in culture. This suggests that the inactivation of the putative tumor suppressor gene Dnd1 facilitates the immortalization of late EGCs in vitro. Injection of the wild type EGCs into blastocysts resulted in the first germ-line competent chimeras. These new pluripotent rat EGCs offer an innovative approach for studies on germ cell tumor development as well as a new tool for genetic manipulations in rats.


Asunto(s)
Células Germinativas/citología , Proteínas de Unión al ARN/metabolismo , Proteínas Supresoras de Tumor/deficiencia , Animales , Células Cultivadas , Femenino , Genes Supresores de Tumor , Células Germinativas/metabolismo , Masculino , Proteínas de Unión al ARN/genética , Ratas , Ratas Endogámicas , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo
10.
Front Neurol ; 13: 907317, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35734474

RESUMEN

Gaucher Disease (GD) 2 is a rare inherited lysosomal disorder. Early-onset and rapid progression of neurovisceral symptoms lead to fatal outcome in early childhood. Treatment is symptomatic, a curative therapy is currently not available. This prospective study describes the clinical and biochemical outcome of a GD 2 patient treated with high dose ambroxol from the age of 4 months. Due to progressive hepatosplenomegaly additional enzyme replacement therapy was required 1 year after ambroxol monotherapy was initiated. Detailed clinical follow-up data demonstrated an age-appropriate neurocognitive and motor development but no clear benefit on peripheral organs. Glucosylsphingosine (Lyso-GL1) in cerebrospinal fluid decreased remarkably compared to pre-treatment, whereas Lyso-GL1 and chitotriosidase in blood increased. Ambroxol treatment of patient fibroblasts revealed a significant increase in ß-glucocerebrosidase activity in vitro. To our knowledge, this is the first report of a GD 2 patient with age-appropriate cognitive and motor development at 3 years of age. Combination of high dose ambroxol with ERT proved to be a successful approach to manage both visceral and neurological manifestations.

11.
Blood ; 113(19): 4690-701, 2009 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-19228926

RESUMEN

Loss of neurofibromin or interferon consensus sequence binding protein (Icsbp) leads to a myeloproliferative disorder. Transcription of NF1 is directly controlled by ICSBP. It has been postulated that loss of NF1 expression resulting from loss of transcriptional activation by ICSBP contributes to human hematologic malignancies. To investigate the functional cooperation of these 2 proteins, we have established Icsbp-deficient mice with Nf1 haploinsufficiency. We here demonstrate that loss of Icsbp and Nf1 haploinsufficiency synergize to induce a forced myeloproliferation in Icsbp-deficient mice because of an expansion of a mature myeloid progenitor cell. Furthermore, Nf1 haploinsufficiency and loss of Icsbp contribute synergistically to progression of the myeloproliferative disorder toward transplantable leukemias. Leukemias are characterized by distinct phenotypes, which correlate with progressive genetic abnormalities. Loss of Nf1 heterozygosity is not mandatory for disease progression, but its occurrence with other genetic abnormalities indicates progressive genetic alterations in a defined subset of leukemias. These data show that loss of the 2 tumor suppressor genes Nf1 and Icsbp synergize in the induction of leukemias.


Asunto(s)
Regulación Leucémica de la Expresión Génica , Factores Reguladores del Interferón/fisiología , Leucemia/etiología , Trastornos Mieloproliferativos/etiología , Neurofibromina 1/fisiología , Animales , Células de la Médula Ósea , Ensayo de Unidades Formadoras de Colonias , Sinergismo Farmacológico , Femenino , Citometría de Flujo , Células Madre Hematopoyéticas , Leucemia/metabolismo , Leucemia/patología , Pérdida de Heterocigocidad , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mielopoyesis , Trastornos Mieloproliferativos/metabolismo , Trastornos Mieloproliferativos/patología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
12.
Nature ; 436(7051): 660-5, 2005 Aug 04.
Artículo en Inglés | MEDLINE | ID: mdl-16079837

RESUMEN

Acute induction of oncogenic Ras provokes cellular senescence involving the retinoblastoma (Rb) pathway, but the tumour suppressive potential of senescence in vivo remains elusive. Recently, Rb-mediated silencing of growth-promoting genes by heterochromatin formation associated with methylation of histone H3 lysine 9 (H3K9me) was identified as a critical feature of cellular senescence, which may depend on the histone methyltransferase Suv39h1. Here we show that Emicro-N-Ras transgenic mice harbouring targeted heterozygous lesions at the Suv39h1, or the p53 locus for comparison, succumb to invasive T-cell lymphomas that lack expression of Suv39h1 or p53, respectively. By contrast, most N-Ras-transgenic wild-type ('control') animals develop a non-lymphoid neoplasia significantly later. Proliferation of primary lymphocytes is directly stalled by a Suv39h1-dependent, H3K9me-related senescent growth arrest in response to oncogenic Ras, thereby cancelling lymphomagenesis at an initial step. Suv39h1-deficient lymphoma cells grow rapidly but, unlike p53-deficient cells, remain highly susceptible to adriamycin-induced apoptosis. In contrast, only control, but not Suv39h1-deficient or p53-deficient, lymphomas senesce after drug therapy when apoptosis is blocked. These results identify H3K9me-mediated senescence as a novel Suv39h1-dependent tumour suppressor mechanism whose inactivation permits the formation of aggressive but apoptosis-competent lymphomas in response to oncogenic Ras.


Asunto(s)
Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/patología , Senescencia Celular , Genes ras/genética , Linfoma/genética , Linfoma/patología , Factores de Ribosilacion-ADP/metabolismo , Animales , Apoptosis/efectos de los fármacos , Biomarcadores de Tumor/análisis , Biomarcadores de Tumor/metabolismo , Transformación Celular Neoplásica/metabolismo , Senescencia Celular/efectos de los fármacos , Senescencia Celular/genética , Inestabilidad Cromosómica/genética , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Regulación Neoplásica de la Expresión Génica , Heterocromatina/genética , Heterocromatina/metabolismo , Linfoma/metabolismo , Metilación , Metiltransferasas/deficiencia , Metiltransferasas/genética , Metiltransferasas/metabolismo , Ratones , Ratones Transgénicos , Lesiones Precancerosas/genética , Lesiones Precancerosas/metabolismo , Lesiones Precancerosas/patología , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Transgenes/genética , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
13.
J Pathol ; 217(5): 620-32, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19191266

RESUMEN

Infiltrating lobular breast cancer (ILBC) is a clinically and biologically distinct tumour entity defined by a characteristic linear cord invasion pattern and inactivation of the CDH1 tumour suppressor gene encoding for E-cadherin. ILBCs also lack beta-catenin expression and show aberrant cytoplasmic localization of the E-cadherin binding protein p120-catenin. The lack of a well-characterized ILBC cell line has hampered the functional characterization of ILBC cells in vitro. We report the establishment of a permanent ILBC cell line, named IPH-926, which was derived from a patient with metastatic ILBC. The DNA fingerprint of IPH-926 verified genetic identity with the patient and had no match among the human cell line collections of several international biological resource banks. IPH-926 expressed various epithelial cell markers but lacked expression of E-cadherin due to a previously unreported, homozygous CDH1 241ins4 frameshift mutation. Detection of the same CDH1 241ins4 mutation in archival tumour tissue of the corresponding primary ILBC proved the clonal origin of IPH-926 from this particular tumour. IPH-926 also lacked beta-catenin expression and showed aberrant cytoplasmic localization of p120-catenin. Array-CGH analysis of IPH-926 revealed a profile of genomic imbalances that included many distinct alterations previously observed in primary ILBCs. Spectral karyotyping of IPH-926 showed a hyperdiploid chromosome complement and numerous clonal, structural aberrations. IPH-926 cells were anti-cancer drug-resistant, clonogenic in soft agar, and tumourigenic in SCID mice. In xenograft tumours, IPH-926 cells recapitulated the linear cord invasion pattern that defines ILBCs. In summary, IPH-926 significantly extends the biological spectrum of the established breast cancer cell lines and will facilitate functional analyses of genuine human ILBC cells in vitro and in vivo.


Asunto(s)
Neoplasias de la Mama/genética , Cadherinas/genética , Carcinoma Lobular/genética , Anciano , Desequilibrio Alélico , Animales , Antígenos CD , Secuencia de Bases , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Cadherinas/deficiencia , Cadherinas/metabolismo , Carcinoma Lobular/metabolismo , Carcinoma Lobular/patología , Carcinoma Lobular/secundario , Línea Celular Tumoral , Femenino , Humanos , Cariotipificación , Ratones , Ratones Endogámicos NOD , Mutación , Proteínas de Neoplasias/metabolismo , Trasplante de Neoplasias , Trasplante Heterólogo
14.
Mol Genet Metab Rep ; 23: 100578, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32226768

RESUMEN

BACKGROUND: Mucopolysaccharidosis type IIIA (MPS IIIA, Sanfilippo A syndrome) is a chronic progressive neurodegenerative storage disorder caused by a deficiency of lysosomal sulfamidase. The clinical hallmarks are sleep disturbances, behavioral abnormalities and loss of cognitive, speech and motor abilities. Affected children show developmental slowing from the second year of life, dementia occurs by the age of 5 years followed by death in the second decade of life. Only a few studies concerning HSCT in MPS IIIA have been published and do not document a clear benefit of treatment. METHODS: The present study summarizes the clinical outcome of a girl with MPS IIIA who received HSCT at the age of 2.5 years. Her clinical course was compared with the natural history of six untreated MPS IIIA patients carrying the same mutations (p.R74C and p. R245H) in the SGSH-gene. RESULTS: Eight years after successful HSCT, the patient showed a global developmental delay. However, cognitive abilities continued to develop, albeit very slowly. There was no sign of regression. She could talk in short sentences, had good motor abilities and performed basic daily living activities by herself. She did not present with sleeping problems, but behavioral abnormalities were profound. In contrast, the six untreated patients with identical mutations in the SGSH-gene showed the typical progressive course of disease with early and continuous loss of abilities. CONCLUSIONS: The present data suggest a beneficial effect of HSCT performed at an early stage of MPS IIIA on cognitive skills, motor function and quality of life.

15.
Methods Mol Biol ; 506: 453-66, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19110644

RESUMEN

Cytogenetic characterization of murine chromosomes using banding techniques like R- or G-banding is technically demanding due to the similar size and the acrocentric structure of all chromosomes. The molecular cytogenetic technique of spectral karyotyping (SKY) overcomes that difficulty by karyotyping metaphase chromosomes after different and simultaneous fluorescence labeling of the whole genome. SKY allows the detection and identification of numerical as well as structural chromosome aberrations with a resolution of approximately 2 Mb. The technique is applicable to all fast-proliferating cells, e.g., cells of the hematopoietic system like stem cells or T- and B-lymphocytes. It is also applicable to murine embryonic fibroblast or cells isolated from tissues with increased proliferation-especially tumor tissues. Furthermore, SKY is recommended for the cytogenetic characterization of newly established cell lines.


Asunto(s)
Inestabilidad Cromosómica , Animales , Hibridación Fluorescente in Situ , Cariotipificación , Ratones , Conformación de Ácido Nucleico
16.
Exp Hematol ; 36(2): 204-15, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18206728

RESUMEN

OBJECTIVE: Hematopoietic stem cells (HSCs) are characterized by their ability to give rise to all mature blood lineages and to renew themselves. In the present study, we show that beta-catenin plays an essential role in the immortalization of hematopoietic progenitor cells (HPCs). MATERIALS AND METHODS: Cellular, molecular, and cytogenetic properties of the immortalized HPCs were determined using flow cytometry (immunophenotyping), microscopy (telomere length, spectral karyotyping), telomere repeat amplification protocol assay (telomerase activity), real-time polymerase chain reaction (expression studies), and in vitro and in vivo differentiation assays. RESULTS: Retroviral-mediated overexpression of human beta-catenin in lin(-)- and lin(-)Sca-1+c-kit+ hematopoietic cells led to generation of novel, murine interleukin-3-, and stem cell factor-dependent hematopoietic multipotent progenitor cell lines (DK1mix and DK2mix) with stable surface antigen expression of the stem cell markers Sca-1 and c-kit (>92%) in long-term culture. Further, immunophenotypic characterization revealed a CD244+CD150(-)CD48(-) phenotype of DKmix cells, which is consistent with the expression profile of multipotential hematopoietic progenitors. Upon exposure to selective hematopoietic cytokines in vitro, and upon transplantation into irradiated congenic recipients, DKmix cells generated progenies of lymphoid and myeloid lineages. Continuous in vitro proliferation and expansion of DKmix cells were associated with stabilized telomere length and consistent telomerase activity. Interestingly, constitutive expression of beta-catenin was not required for sustained long-term viability and proliferation of immortalized DKmix cells. CONCLUSION: In summary, our findings define beta-catenin as an important regulator in HPC self-renewal and function. Further, our results distinguish the importance of beta-catenin in the immortalization process of HPCs, from its dispensable role in their maintenance.


Asunto(s)
Células Madre Hematopoyéticas/metabolismo , Células Madre Multipotentes/metabolismo , Retroviridae , Transducción Genética , beta Catenina/biosíntesis , Animales , Antígenos de Diferenciación/biosíntesis , Antígenos de Diferenciación/genética , Línea Celular Transformada , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/genética , Citometría de Flujo , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Células Madre Hematopoyéticas/citología , Humanos , Interleucina-3/metabolismo , Interleucina-3/farmacología , Ratones , Células Madre Multipotentes/citología , Factor de Células Madre/metabolismo , Factor de Células Madre/farmacología , Telomerasa/genética , Telomerasa/metabolismo , Telómero/genética , Telómero/metabolismo , beta Catenina/genética
17.
Cancer Res ; 66(14): 6913-8, 2006 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-16849534

RESUMEN

AIMP3 (previously known as p18) was shown to up-regulate p53 in response to DNA damage. Here, we show that AIMP3 couples oncogenic stresses to p53 activation to prevent cell transformation. Growth factor- or Ras-dependent induction of p53 was blocked by single allelic loss of AIMP3 as well as by suppression of AIMP3. AIMP3 heterozygous cells became susceptible to cell transformation induced by oncogenes such as Ras or Myc alone. The transformed AIMP3+/- cells showed severe abnormality in cell division and chromosomal structure. Thus, AIMP3 plays crucial roles in p53-mediated tumor-suppressive response against oncogenic stresses via differential activation of ATM and ATR, and in the maintenance of genomic stability.


Asunto(s)
Transformación Celular Neoplásica/genética , Inhibidor p18 de las Quinasas Dependientes de la Ciclina/genética , Regulación de la Expresión Génica/genética , Genes ras , Inestabilidad Genómica/genética , Proteína p53 Supresora de Tumor/genética , Alelos , Animales , Inhibidor p18 de las Quinasas Dependientes de la Ciclina/metabolismo , Embrión de Mamíferos , Fibroblastos , Ratones , ARN sin Sentido/genética , Transfección , Proteína p53 Supresora de Tumor/biosíntesis
18.
Haematologica ; 92(4): 460-8, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17488656

RESUMEN

BACKGROUND AND OBJECTIVES: Mantle cell lymphoma (MCL), a mature B-cell neoplasm, is genetically characterized by the translocation t(11;14)(q13;q32). However, secondary alterations are required for malignant transformation. The identification of inactivated tumor suppressor genes contributing to the development of MCL may lead to further elucidation of the biology of this disease and help to identify novel targets for therapy. DESIGN AND METHODS: Whole genome microarray-based gene expression profiling on treated versus untreated MCL cell lines was used to identify genes induced by 5-aza-2'-deoxycytidine. The degree of promoter methylation and transcriptional silencing of selected genes was then proven in MCL cell lines and primary cases by methylation-specific polymerase chain reaction (PCR) techniques, real-time PCR and gene expression profiling. RESULTS: After 5-aza-2'-deoxycytidine treatment, we identified more than 1000 upregulated genes, 16 of which were upregulated > or =3-fold. Most of them were not known to be silenced by methylation in MCL. A low expression of ING1, RUNX3 and BNIP3L was observed in three of the five the MCL cell lines. In addition, the expression of PARG1, which is located in the frequently deleted region 1p22.1, was substantially reduced and displayed at least partial promoter methylation in all investigated MCL cell lines as well as in 31 primary MCL cases. INTERPRETATION AND CONCLUSIONS: In summary, we identified interesting novel candidate genes that probably contribute to the progression of MCL and suggest that PARG1 is a strong candidate tumor suppressor gene in MCL.


Asunto(s)
Metilación de ADN , ADN de Neoplasias/química , Proteínas Activadoras de GTPasa/genética , Genes Supresores de Tumor , Linfoma de Células del Manto/genética , Regiones Promotoras Genéticas/genética , Azacitidina/análogos & derivados , Azacitidina/farmacología , Línea Celular Tumoral/efectos de los fármacos , Línea Celular Tumoral/metabolismo , Cromosomas Humanos Par 1/genética , Cromosomas Humanos Par 11/ultraestructura , Cromosomas Humanos Par 14/ultraestructura , ADN de Neoplasias/genética , Decitabina , Progresión de la Enfermedad , Proteínas Activadoras de GTPasa/biosíntesis , Proteínas Activadoras de GTPasa/fisiología , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Silenciador del Gen/efectos de los fármacos , Humanos , Linfoma de Células del Manto/patología , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena de la Polimerasa/métodos , Transcripción Genética/efectos de los fármacos , Translocación Genética
19.
Cancer Res ; 65(7): 2662-7, 2005 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-15805263

RESUMEN

We have identified a t(8;9)(p21-23;p23-24) in seven male patients (mean age 50, range 32-74) with diverse hematologic malignancies and clinical outcomes: atypical chronic myeloid leukemia/chronic eosinophilic leukemia (n = 5), secondary acute myeloid leukemia (n = 1), and pre-B-cell acute lymphoblastic leukemia (n = 1). Initial fluorescence in situ hybridization studies of one patient indicated that the nonreceptor tyrosine kinase Janus-activated kinase 2 (JAK2) at 9p24 was disrupted. Rapid amplification of cDNA ends-PCR identified the 8p22 partner gene as human autoantigen pericentriolar material (PCM1), a gene encoding a large centrosomal protein with multiple coiled-coil domains. Reverse transcription-PCR and fluorescence in situ hybridization confirmed the fusion in this case and also identified PCM1-JAK2 in the six other t(8;9) patients. The breakpoints were variable in both genes, but in all cases the chimeric mRNA is predicted to encode a protein that retains several of the predicted coiled-coil domains from PCM1 and the entire tyrosine kinase domain of JAK2. Reciprocal JAK2-PCM1 mRNA was not detected in any patient. We conclude that human autoantigen pericentriolar material (PCM1)-JAK2 is a novel, recurrent fusion gene in hematologic malignancies. Patients with PCM1-JAK2 disease are attractive candidates for targeted signal transduction therapy.


Asunto(s)
Proteínas de Ciclo Celular/genética , Cromosomas Humanos Par 8/genética , Cromosomas Humanos Par 9/genética , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Leucemia Mieloide/genética , Proteínas de Fusión Oncogénica/genética , Proteínas Tirosina Quinasas/genética , Proteínas Proto-Oncogénicas/genética , Translocación Genética , Enfermedad Aguda , Adulto , Anciano , Secuencia de Aminoácidos , Autoantígenos , Secuencia de Bases , Humanos , Janus Quinasa 2 , Masculino , Persona de Mediana Edad , Datos de Secuencia Molecular , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
20.
Cancer Genet Cytogenet ; 161(1): 51-6, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16080957

RESUMEN

Most patients with Philadelphia (Ph)-positive acute lymphoblastic leukemia (ALL) show evidence of secondary chromosome aberrations that may influence the course of disease and response to treatment. To better understand how these secondary chromosomal aberrations occur and to investigate whether the p185/p190 BCR-ABL fusion protein may directly induce an increased chromosomal instability and subsequently the appearance of clonal chromosome aberrations, three BRC-ABL (p185/ p190)-transduced mouse pre-B cell lines were analyzed by spectral karyotyping and fluorescence in situ hybridization. The human wild-type BCR-ABL gene was expressed at a level comparable with that in human Ph-positive leukemias at diagnosis. All BCR-ABL-transduced cell lines acquired similar clonal chromosomal aberrations. Trisomy 5 was always present, followed by loss of the Y chromosome, trisomy of chromosomes 12 and 18, and an unbalanced translocation between chromosomes X and 12. Thus, ectopic p185/p190 BCR-ABL expression, such as p210 BCR-ABL, PML-RARA, or C-MYC transduction, may induce an increased chromosomal instability leading to clonal karyotypic evolution, which may mimic secondary chromosome aberrations in human Ph-positive ALL.


Asunto(s)
Aberraciones Cromosómicas , Proteínas de Fusión bcr-abl/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras B/genética , Animales , Línea Celular Transformada , ADN de Neoplasias/análisis , Humanos , Hibridación Fluorescente in Situ , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Ratones , Ratones Endogámicos C57BL , Cromosoma Filadelfia , Leucemia-Linfoma Linfoblástico de Células Precursoras B/metabolismo , Retroviridae/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Cariotipificación Espectral , Transfección , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA