Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
3.
J Clin Invest ; 119(11): 3395-407, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19809159

RESUMEN

Rhabdomyosarcoma (RMS) is a childhood cancer originating from skeletal muscle, and patient survival is poor in the presence of metastatic disease. Few determinants that regulate metastasis development have been identified. The receptor tyrosine kinase FGFR4 is highly expressed in RMS tissue, suggesting a role in tumorigenesis, although its functional importance has not been defined. Here, we report the identification of mutations in FGFR4 in human RMS tumors that lead to its activation and present evidence that it functions as an oncogene in RMS. Higher FGFR4 expression in RMS tumors was associated with advanced-stage cancer and poor survival, while FGFR4 knockdown in a human RMS cell line reduced tumor growth and experimental lung metastases when the cells were transplanted into mice. Moreover, 6 FGFR4 tyrosine kinase domain mutations were found among 7 of 94 (7.5%) primary human RMS tumors. The mutants K535 and E550 increased autophosphorylation, Stat3 signaling, tumor proliferation, and metastatic potential when expressed in a murine RMS cell line. These mutants also transformed NIH 3T3 cells and led to an enhanced metastatic phenotype. Finally, murine RMS cell lines expressing the K535 and E550 FGFR4 mutants were substantially more susceptible to apoptosis in the presence of a pharmacologic FGFR inhibitor than the control cell lines expressing the empty vector or wild-type FGFR4. Together, our results demonstrate that mutationally activated FGFR4 acts as an oncogene, and these are what we believe to be the first known mutations in a receptor tyrosine kinase in RMS. These findings support the potential therapeutic targeting of FGFR4 in RMS.


Asunto(s)
Mutación/genética , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/genética , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/metabolismo , Rabdomiosarcoma/fisiopatología , Animales , Ciclo Celular , Línea Celular Transformada , Línea Celular Tumoral , Proliferación Celular , Replicación del ADN , Modelos Animales de Enfermedad , Humanos , Neoplasias Pulmonares/fisiopatología , Neoplasias Pulmonares/secundario , Ratones , Modelos Moleculares , Metástasis de la Neoplasia , Fosforilación , Estructura Terciaria de Proteína , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/química , Rabdomiosarcoma/mortalidad , Factor de Transcripción STAT3/metabolismo , Trasplante Heterólogo
4.
Lab Hematol ; 15(3): 30-3, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19758967

RESUMEN

The diagnosis and management of alpha-thalassemia may be complicated by the variability of the phenotype, which is due to the interaction of coinherited alpha-thalassemia and the variable severity of beta-thalassemia mutations. A well-documented case of complex beta- and alpha-thalassemia coinheritance is described. Laboratory and clinical data for the patient and her family are reviewed. The patient is an asymptomatic girl, one of identical twins. She presented at 1 month of age for follow-up of an abnormal newborn-screening result (hemoglobin F only), which initially suggested homozygosity for beta-thalassemia. Extensive studies on the patient and family revealed that she had coinherited alpha-thalassemia traits and homozygous beta-thalassemia. This case demonstrates the interaction of coinherited alpha- and beta-thalassemia with the resultant amelioration of the clinical phenotype. It also highlights the importance of family studies and close follow-up in diagnosing complex hemoglobinopathies.


Asunto(s)
Enfermedades en Gemelos/genética , Talasemia alfa/genética , Talasemia beta/genética , Negro o Afroamericano , Electroforesis de las Proteínas Sanguíneas , Enfermedades en Gemelos/diagnóstico , Eritrocitos/citología , Salud de la Familia , Femenino , Eliminación de Gen , Tamización de Portadores Genéticos , Hemoglobinas/genética , Humanos , Recién Nacido , Masculino , Tamizaje Neonatal , Fragmentos de Péptidos/genética , Fenotipo , Gemelos Monocigóticos , Talasemia alfa/complicaciones , Talasemia alfa/diagnóstico , Talasemia beta/complicaciones , Talasemia beta/diagnóstico
5.
Cancer Res ; 69(7): 2902-11, 2009 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-19339268

RESUMEN

The highly aggressive muscle cancer alveolar rhabdomyosarcoma (ARMS) is one of the most common soft tissue sarcoma of childhood, yet the outcome for the unresectable and metastatic disease is dismal and unchanged for nearly three decades. To better understand the pathogenesis of this disease and to facilitate novel preclinical approaches, we previously developed a conditional mouse model of ARMS by faithfully recapitulating the genetic mutations observed in the human disease, i.e., activation of Pax3:Fkhr fusion gene with either p53 or Cdkn2a inactivation. In this report, we show that this model recapitulates the immunohistochemical profile and the rapid progression of the human disease. We show that Pax3:Fkhr expression increases during late preneoplasia but tumor cells undergoing metastasis are under apparent selection for Pax3:Fkhr expression. At a whole-genome level, a cross-species gene set enrichment analysis and metagene projection study showed that our mouse model is most similar to human ARMS when compared with other pediatric cancers. We have defined an expression profile conserved between mouse and human ARMS, as well as a Pax3:Fkhr signature, including the target gene, SKP2. We further identified 7 "druggable" kinases overexpressed across species. The data affirm the accuracy of this genetically engineered mouse model.


Asunto(s)
Rabdomiosarcoma Alveolar/genética , Rabdomiosarcoma Alveolar/patología , Alelos , Animales , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Progresión de la Enfermedad , Proteína Forkhead Box O1 , Factores de Transcripción Forkhead/biosíntesis , Factores de Transcripción Forkhead/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , Ratones Noqueados , Proteínas de Fusión Oncogénica/biosíntesis , Proteínas de Fusión Oncogénica/genética , Factor de Transcripción PAX3 , Factores de Transcripción Paired Box/biosíntesis , Factores de Transcripción Paired Box/genética , Penetrancia , Rabdomiosarcoma Alveolar/metabolismo , Activación Transcripcional , Proteína p53 Supresora de Tumor/genética
6.
Pediatr Blood Cancer ; 53(2): 136-44, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19213072

RESUMEN

BACKGROUND: Tumor initiation has been attributed to haploinsufficiency at a single locus for a large number of cancers. Patched1 (Ptc1) was one of the first such loci, and Ptc1 haploinsufficiency has been asserted to lead to medulloblastoma and rhabdomyosarcoma in mice. PROCEDURE: To study the role of Ptc1 in cerebellar tumor development and to create a preclinical therapeutic platform, we have generated a conditional Ptc1 haploinsufficiency model of medulloblastoma by inactivating Ptc1 in Pax7-expressing cells of the cerebellum. RESULTS: These mice developed exclusively medulloblastoma. We show that despite the presence of transcription of Ptc1, Ptc1 protein is nearly undetectable or absent in tumors. Our results suggest that Ptc1 loss of function is complete, but achieved at the protein level rather than by the classic genetic two-hit mechanism or a strict half-dosage genetic haploinsufficiency mechanism. Furthermore, we found that bortezomib, a 26S proteasome inhibitor, had a significant anti-tumor activity in vitro and in vivo, which was accompanied by restoration of Ptc1 protein and downregulation of the hedgehog signaling pathway. The same effect was seen for both human and mouse medulloblastoma tumor cell growth. CONCLUSIONS: These results suggest that proteasome inhibition is a potential new therapeutic approach in medulloblastoma.


Asunto(s)
Antineoplásicos/farmacología , Ácidos Borónicos/farmacología , Neoplasias Cerebelosas/genética , Meduloblastoma/genética , Procesamiento Proteico-Postraduccional/fisiología , Pirazinas/farmacología , Receptores de Superficie Celular/efectos de los fármacos , Animales , Bortezomib , Línea Celular Tumoral , Transformación Celular Neoplásica/efectos de los fármacos , Transformación Celular Neoplásica/genética , Modelos Animales de Enfermedad , Técnicas de Sustitución del Gen , Proteínas Hedgehog/efectos de los fármacos , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Humanos , Immunoblotting , Inmunoprecipitación , Pérdida de Heterocigocidad/efectos de los fármacos , Pérdida de Heterocigocidad/genética , Ratones , Ratones Noqueados , Factor de Transcripción PAX7 , Receptores Patched , Receptor Patched-1 , Complejo de la Endopetidasa Proteasomal/metabolismo , Isoformas de Proteínas , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo
7.
Am J Pathol ; 174(2): 550-64, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19147825

RESUMEN

Rhabdomyosarcoma (RMS) in children occurs as two major histological subtypes, embryonal (ERMS) and alveolar (ARMS). ERMS is associated with an 11p15.5 loss of heterozygosity (LOH) and may be confused with nonmyogenic, non-RMS soft tissue sarcomas. ARMS expresses the product of a genomic translocation that fuses FOXO1 (FKHR) with either PAX3 or PAX7 (P-F); however, at least 25% of cases lack these translocations. Here, we describe a genomic-based classification scheme that is derived from the combined gene expression profiling and LOH analysis of 160 cases of RMS and non-RMS soft tissue sarcomas that is at variance with conventional histopathological schemes. We found that gene expression profiles and patterns of LOH of ARMS cases lacking P-F translocations are indistinguishable from conventional ERMS cases. A subset of tumors that has been histologically classified as RMS lack myogenic gene expression. However, classification based on gene expression is possible using as few as five genes with an estimated error rate of less than 5%. Using immunohistochemistry, we characterized two markers, HMGA2 and TFAP2ss, which facilitate the differential diagnoses of ERMS and P-F RMS, respectively, using clinical material. These objectively derived molecular classes are based solely on genomic analysis at the time of diagnosis and are highly reproducible. Adoption of these molecular criteria may offer a more clinically relevant diagnostic scheme, thus potentially improving patient management and therapeutic RMS outcomes.


Asunto(s)
Biomarcadores de Tumor/análisis , Perfilación de la Expresión Génica , Rabdomiosarcoma/clasificación , Rabdomiosarcoma/diagnóstico , Rabdomiosarcoma/genética , Adolescente , Niño , Preescolar , Diagnóstico Diferencial , Femenino , Genotipo , Proteína HMGA2/genética , Proteína HMGA2/metabolismo , Humanos , Inmunohistoquímica , Lactante , Estimación de Kaplan-Meier , Pérdida de Heterocigocidad , Masculino , Fenotipo , Polimorfismo de Nucleótido Simple , Reproducibilidad de los Resultados , Sarcoma/patología , Sensibilidad y Especificidad , Análisis de Matrices Tisulares , Factor de Transcripción AP-2/genética , Factor de Transcripción AP-2/metabolismo
8.
Pediatr Dev Pathol ; 12(2): 136-42, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-18721005

RESUMEN

Juvenile myelomonocytic leukemia (JMML) is a rare, aggressive, clonal hematopoietic disorder of childhood with features of both myelodysplasia (thrombocytopenia, anemia) and myeloproliferation (leukocytosis, monocytosis). In most cases there is marrow hypercellularity, splenomegaly, and extramedullary involvement. In 1997 an international consensus on terminology was reached and guidelines/criteria for diagnosis were proposed. A recent World Health Organization classification described the current diagnostic criteria of JMML. Although the diagnosis of JMML has been facilitated, it can be challenging, especially in the early stages or when it 1st presents as an extramedullary tumor. We report a series of 7 cases diagnosed over a period of 10 years (from January 1, 1996, to December 31, 2005). Two cases had interesting associated findings that would potentially lead to delay in diagnosis or misdiagnosis. Two other cases had extramedullary involvement with symptoms referable to the organs of involvement at presentation. Clinical and pathologic findings are summarized with a review of relevant literature.


Asunto(s)
Leucemia Mielomonocítica Juvenil/patología , Células de la Médula Ósea/patología , Trasplante de Médula Ósea , Preescolar , Resultado Fatal , Femenino , Humanos , Lactante , Leucemia Mielomonocítica Juvenil/complicaciones , Leucemia Mielomonocítica Juvenil/terapia , Masculino , Monocitos/patología , Esplenomegalia/diagnóstico , Esplenomegalia/etiología
9.
Cancer Res ; 68(23): 9583-8, 2008 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-19047133

RESUMEN

Rhabdomyosarcoma (RMS) is the most common soft tissue sarcoma of childhood and adolescence. Despite advances in therapy, patients with a histologic variant of RMS known as alveolar (aRMS) have a 5-year survival rate of <30%. aRMS tissues exhibit a number of genetic changes, including loss-of-function of the p53 and Rb tumor suppressor pathways, amplification of MYCN, stabilization of telomeres, and most characteristically, reciprocal translocation of loci involving the PAX and FKHR genes, generating the PAX7-FKHR or PAX3-FKHR fusion proteins. We previously showed that PAX3-FKHR expression in primary human myoblasts, cells that can give rise to RMS, cooperated with loss of p16INK4A to promote extended proliferation. To better understand the genetic events required for aRMS formation, we then stepwise converted these cells to their transformed counterpart. PAX3-FKHR, the catalytic unit of telomerase hTERT, and MycN, in cooperation with down-regulation of p16INK4A/p14ARF expression, were necessary and sufficient to convert normal human myoblasts into tumorigenic cells that gave rise to aRMS tumors. However, the order of expression of these transgenes was critical, as only those cells expressing PAX3-FKHR early could form tumors. We therefore suggest that the translocation of PAX3 to FKHR drives proliferation of myoblasts, and a selection for loss of p16INK4A/p14ARF. These early steps, coupled with MycN amplification and telomere stabilization, then drive the cells to a fully tumorigenic state.


Asunto(s)
Transformación Celular Neoplásica/genética , Neoplasias de los Músculos/genética , Rabdomiosarcoma Alveolar/genética , Animales , Línea Celular Tumoral , Inhibidor p16 de la Quinasa Dependiente de Ciclina/biosíntesis , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , Ratones SCID , Neoplasias de los Músculos/metabolismo , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Músculo Esquelético/fisiología , Proteínas de Fusión Oncogénica/biosíntesis , Proteínas de Fusión Oncogénica/genética , Rabdomiosarcoma Alveolar/metabolismo , Trasplante Heterólogo
10.
Cancer Cell ; 14(5): 369-81, 2008 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-18977326

RESUMEN

Studies support the importance of microRNAs in physiological and pathological processes. Here we describe the regulation and function of miR-29 in myogenesis and rhabdomyosarcoma (RMS). Results demonstrate that in myoblasts, miR-29 is repressed by NF-kappaB acting through YY1 and the Polycomb group. During myogenesis, NF-kappaB and YY1 downregulation causes derepression of miR-29, which in turn accelerates differentiation by targeting its repressor YY1. However, in RMS cells and primary tumors that possess impaired differentiation, miR-29 is epigenetically silenced by an activated NF-kappaB-YY1 pathway. Reconstitution of miR-29 in RMS in mice inhibits tumor growth and stimulates differentiation, suggesting that miR-29 acts as a tumor suppressor through its promyogenic function. Together, these results identify a NF-kappaB-YY1-miR-29 regulatory circuit whose disruption may contribute to RMS.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , MicroARNs/metabolismo , Desarrollo de Músculos/fisiología , Mioblastos Esqueléticos/citología , FN-kappa B/metabolismo , Rabdomiosarcoma/metabolismo , Factor de Transcripción YY1/metabolismo , Animales , Western Blotting , Ciclo Celular/fisiología , Diferenciación Celular/fisiología , Proliferación Celular , Células Cultivadas , Inmunoprecipitación de Cromatina , Biología Computacional , Regulación hacia Abajo , Retroalimentación Fisiológica , Fibroblastos , Humanos , Ratones , Ratones Endogámicos C57BL , MicroARNs/antagonistas & inhibidores , MicroARNs/genética , Mioblastos Esqueléticos/metabolismo , FN-kappa B/genética , Conformación de Ácido Nucleico , Regiones Promotoras Genéticas , Rabdomiosarcoma/genética , Rabdomiosarcoma/prevención & control , Transducción de Señal , Factor de Transcripción YY1/genética
11.
Skeletal Radiol ; 37(7): 683-7, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18421456

RESUMEN

We describe herein a rare case of primary rhabdomyosarcoma (RMS) occurring in the sacrum. A 16-year-old woman presented with a 2-month history of pain in bilateral buttocks and posterior thighs. Computed tomography showed a primary tumor with bone destruction in the 2nd sacral vertebra and invasion to the 1st to 3rd vertebrae and retroperitoneal space. Histological examination of the tumor showed proliferation of spindle-shaped cells intermingled with rhabdomyoblasts in a fascicular and storiform growth pattern. Tumor cells showed immunoreactivity for vimentin, desmin, muscle-specific actin, sarcomeric actin, alpha-smooth muscle actin and CD99, and partial immunoreactivity for myoD1, myf-4, myogenin and myoglobin. Reverse transcription polymerase chain reaction demonstrated expression of myoD1. On the basis of the aforementioned findings, a poorly differentiated spindle cell variant of embryonal RMS was diagnosed. The patient underwent combined therapy with chemotherapy and radiotherapy, but died 17 months after incisional biopsy. The present case is instructive in differential diagnosis of primary bone tumors, and the possibility of skeletal RMS needs to be considered.


Asunto(s)
Rabdomiosarcoma/diagnóstico , Sacro/diagnóstico por imagen , Sacro/patología , Neoplasias de la Columna Vertebral/diagnóstico , Adolescente , Femenino , Humanos , Imagen por Resonancia Magnética , Tomografía Computarizada por Rayos X
12.
Cancer Inform ; 6: 127-37, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-19259407

RESUMEN

Tissue resources have become an important component of the infrastructure of institutions as well as companies performing biomedical research. Such tissue resources may be in the model of a bank, collecting a limited type of tissues and processing and storing them following a specific protocol. Such banks or archives may be associated with a clinical study or may function indepedently. An alternative type of tissue resource is utilized by many institutions and cancer centers. In this model, the investigator specifies the methods by which selected tissues are to be collected, processed and stored. In such a "prospective model", initially developed at the University of Alabama at Birmingham and the Ohio State University in the late 1970's and adopted by the Cooperative Human Tissue Network in 1986, specific types of tissues are not collected unless requested by an investigator. At some sites, both a prospective and an archival (bank) model are followed. This article describes an informatics approach needed to support a prospective tissue resource. It is by necessity more complicated than a model which supports a tissue bank but also can be used by a tissue bank. Of great importance is the approach to vocabulary and common data elements needed to support the informatics system of a prospective tissue resource, especially if the informatics system is to be used by a variety of personnel with greatly varying educational backgrounds.

13.
J Clin Oncol ; 25(31): 4909-13, 2007 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-17971587

RESUMEN

PURPOSE: Initial response to induction chemotherapy predicts failure-free survival (FFS) in osteosarcoma and Ewing's sarcoma. For Intergroup Rhabdomyosarcoma Study (IRS) IV patients with group III rhabdomyosarcoma, we assessed whether reported response assessed by anatomic imaging at week 8 predicted FFS. PATIENTS AND METHODS: We studied 444 group III patients who received induction therapy, had response assessed at week 8 by anatomic imaging, and continued with protocol therapy. Induction chemotherapy was generally followed by radiation therapy (RT) starting after week 9. Response to induction therapy was determined at weeks 0 and 8. Local institutions coded response. RESULTS: Response rate for the entire cohort at week 8 was 77% (95% CI, 73% to 81%; complete response [CR], 21%; partial response [PR], 56%) but response had no influence on FFS (P = .57). Two hundred seventy-two patients received standard-timing RT at week 9 and thus only chemotherapy during induction. Response rate was 81% (95% CI, 76% to 86%; CR, 22%; PR, 59%). In these patients, response did not influence FFS except for those with alveolar histology. One hundred thirty-two other patients received chemotherapy and RT during induction (up-front RT). Response rate was 65% (95% CI, 57% to 73%; CR, 12%; PR, 53%), but response had no influence on FFS (P = .69). Forty patients received no RT at all (protocol violation) and response to induction therapy had no effect on FFS. CONCLUSION: In IRS-IV, response rate to induction therapy was 77% in group III patients, was independent of histology, and had no influence on FFS overall.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Terapia Neoadyuvante , Rabdomiosarcoma/tratamiento farmacológico , Neoplasias de los Tejidos Blandos/tratamiento farmacológico , Adolescente , Adulto , Niño , Preescolar , Fraccionamiento de la Dosis de Radiación , Femenino , Humanos , Lactante , Masculino , Rabdomiosarcoma/radioterapia , Neoplasias de los Tejidos Blandos/radioterapia , Análisis de Supervivencia
14.
Pediatr Dev Pathol ; 10(4): 315-9, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17638424

RESUMEN

We describe a case of autoimmune lymphoproliferative syndrome (ALPS), which is very unusual with regard to a clinical onset soon after birth, and a clinical picture dominated by splenomegaly, jaundice, and consumptive peripheral blood cytopenias, with minimal lymphadenopathy. Our documented close follow up demonstrated initial involvement of the spleen, followed by involvement of the bone marrow and the peripheral blood. The patient underwent bone marrow transplant and is alive and well 20 months after diagnosis.


Asunto(s)
Anomalías Múltiples , Enfermedades Autoinmunes/patología , Trastornos Linfoproliferativos/patología , Enfermedades Autoinmunes/congénito , Enfermedades Autoinmunes/terapia , Trasplante de Médula Ósea , Complejo CD3/metabolismo , Enfermedades en Gemelos , Humanos , Recién Nacido , Ictericia/congénito , Ictericia/patología , Trastornos Linfoproliferativos/congénito , Trastornos Linfoproliferativos/terapia , Masculino , Esplenomegalia/congénito , Esplenomegalia/patología , Esplenomegalia/cirugía , Síndrome , Linfocitos T/metabolismo , Linfocitos T/patología , Trombocitopenia/congénito , Trombocitopenia/patología , Resultado del Tratamiento , Gemelos
15.
Cancer Res ; 67(14): 6691-9, 2007 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-17638879

RESUMEN

Rhabdomyosarcoma is the most common soft tissue sarcoma of childhood and adolescence. Despite advances in therapy, patients with a histologic variant of rhabdomyosarcoma known as alveolar rhabdomyosarcoma (ARMS) have a 5-year survival of <30%. ARMS is characterized by a chromosomal translocation generating the PAX3-FKHR fusion gene. However, ectopic expression of PAX3-FKHR often induces inhibition of cell proliferation, or cell death, when expressed in nonmuscle cells. This prompted us to explore the effect of expressing PAX3-FKHR in more relevant cells, specifically primary human skeletal muscle cells because these cells can be converted to a tumorigenic state that mimics rhabdomyosarcoma. PAX3-FKHR expression promoted both fetal and postnatal primary human skeletal muscle cell precursors to bypass the senescence growth arrest checkpoint. This bypass was accompanied by epigenetic DNA methylation of the p16(INK4A) promoter and correspondingly a loss of expression of this tumor suppressor. Knockdown of p16(INK4A) cooperated with PAX3-FKHR to drive proliferation past senescence, whereas reintroduction of wild-type p16(INK4A) in post-senescent cells caused growth arrest. Thus, PAX3-FKHR acts in concert with loss of p16(INK4A) to promote inappropriate proliferation of skeletal muscle cells. This association between PAX3-FKHR expression and p16(INK4A) loss was seen in human ARMS tumor tissue, as both human rhabdomyosarcoma cell lines and tissue microarrays showed a trend toward down-regulation of p16(INK4A) protein in alveolar subsets. We surmise that the generation of the PAX3-FKHR fusion protein may require loss of p16(INK4A) to promote malignant proliferation of skeletal muscle cells as an early step in ARMS tumorigenesis.


Asunto(s)
Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Factores de Transcripción Forkhead/fisiología , Regulación Neoplásica de la Expresión Génica , Neoplasias Pulmonares/metabolismo , Factores de Transcripción Paired Box/fisiología , Rabdomiosarcoma/metabolismo , Línea Celular Tumoral , Proliferación Celular , Senescencia Celular , Metilación de ADN , Proteína Forkhead Box O1 , Humanos , Músculo Esquelético/citología , Análisis de Secuencia por Matrices de Oligonucleótidos , Factor de Transcripción PAX3 , Regiones Promotoras Genéticas , Proteínas Recombinantes de Fusión
16.
BMC Cancer ; 7: 111, 2007 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-17598902

RESUMEN

BACKGROUND: Stat3 has been classified as a proto-oncogene and constitutive Stat3 signaling appears to be involved in oncogenesis of human cancers. However, whether constitutive Stat3 signaling plays a role in the survival and growth of osteosarcomas, rhabdomyosarcomas, and soft-tissue sarcomas is still unclear. METHODS: To examine whether Stat3 is activated in osteosarcomas, rhabdomyosarcomas and other soft-tissue sarcomas we analyzed sarcoma tissue microarray slides and sarcoma cell lines using immunohistochemistry and Western blot analysis, respectively, with a phospho-specific Stat3 antibody. To examine whether the activated Stat3 pathway is important for sarcoma cell growth and survival, adenovirus-mediated expression of a dominant-negative Stat3 (Y705F) and a small molecule inhibitor (termed STA-21) were used to inhibit constitutive Stat3 signaling in human sarcoma cell lines expressing elevated levels of Stat3 phosphorylation. Cell viability was determined by MTT assays and induction of apoptosis was analyzed by western blotting using antibodies that specifically recognize cleaved caspases-3, 8, and 9. RESULTS: Stat3 phosphorylation is elevated in 19% (21/113) of osteosarcoma, 27% (17/64) of rhabdomyosarcoma, and 15% (22/151) of other soft-tissue sarcoma tissues as well as in sarcoma cell lines. Expression of the dominant-negative Stat3 and treatment of STA-21 inhibited cell viability and growth and induced apoptosis through caspases 3, 8 and 9 pathways in human sarcoma cell lines expressing elevated levels of phosphorylated Stat3. CONCLUSION: This study demonstrates that Stat3 phosphorylation is elevated in human rhabdomyosarcoma, osteosarcomas and soft-tissue sarcomas. Furthermore, the activated Stat3 pathway is important for cell growth and survival of human sarcoma cells.


Asunto(s)
Proliferación Celular , Supervivencia Celular/fisiología , Activación Enzimática/fisiología , Osteosarcoma/metabolismo , Rabdomiosarcoma/metabolismo , Factor de Transcripción STAT3/metabolismo , Apoptosis/fisiología , Western Blotting , Caspasas/metabolismo , Línea Celular Tumoral , Técnica del Anticuerpo Fluorescente , Humanos , Inmunohistoquímica , Fosforilación , Proto-Oncogenes Mas , Análisis de Matrices Tisulares
17.
Mod Pathol ; 20(9): 936-46, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17585318

RESUMEN

Rhabdomyosarcoma is the most common pediatric soft-tissue sarcoma, which includes two major subtypes, alveolar and embryonal rhabdomyosarcoma. The mechanism of its oncogenesis is largely unknown. However, the oncogenic process of rhabdomyosarcoma involves multi-stages of signaling protein dysregulation characterized by prolonged activation of tyrosine and serine/threonine kinases. To better understand this protein dysregulation, we evaluated the phosphorylation profiles of multiple tyrosine and serine/threonine kinases to identify whether these protein kinases are activated in rhabdomyosarcoma. We applied immunohistochemistry with phospho-specific antibodies to examine phosphorylation levels of selected receptor and non-receptor tyrosine kinases, mammalian target of rapamycin (mTOR), p70S6K, and protein kinase C (PKC) isozymes on alveolar and embryonal rhabdomyosarcoma tissue microarray slides. Our results showed that the phosphorylation levels of these kinases are elevated in some rhabdomyosarcoma tissues compared to normal tissues. Phosphorylation levels of receptor and non-receptor tyrosine kinases are elevated between 26 and 68% in alveolar rhabdomyosarcoma and between 24 and 71% in embryonal rhabdomyosarcoma, respectively, compared to normal tissues. In addition, phosphorylation levels of mTOR and its downstream targets, p70S6K, S6, and 4EBP1, are increased between 50 and 72% in both subtypes of rhabdomyosarcoma. Further, phosphorylation levels of PKCalpha, PKCdelta, PKCtheta, and PKCzeta/lambda are upregulated between 57 and 69% in alveolar rhabdomyosarcoma and between 43 and 72% in embryonal rhabdomyosarcoma. This is the first report to create a phosphorylation profile of tyrosine and serine/threonine kinases involved in the mTOR and PKC pathways of alveolar and embryonal rhabdomyosarcoma. These protein kinases may play roles in the development or tumor progression of rhabdomyosarcomas and thus may serve as novel targets for therapeutic intervention.


Asunto(s)
Proteínas Serina-Treonina Quinasas/análisis , Proteínas Tirosina Quinasas/análisis , Rabdomiosarcoma Alveolar/enzimología , Rabdomiosarcoma Embrionario/enzimología , Proteínas Adaptadoras Transductoras de Señales/análisis , Adolescente , Proteínas de Ciclo Celular , Niño , Preescolar , Femenino , Humanos , Inmunohistoquímica , Lactante , Recién Nacido , Isoenzimas/análisis , Masculino , Estadificación de Neoplasias , Fosfoproteínas/análisis , Fosforilación , Proyectos Piloto , Proteína Quinasa C/análisis , Proteínas Quinasas/análisis , Proteínas Tirosina Quinasas Receptoras/análisis , Rabdomiosarcoma Alveolar/patología , Rabdomiosarcoma Embrionario/patología , Proteína S6 Ribosómica/análisis , Proteínas Quinasas S6 Ribosómicas 70-kDa/análisis , Serina-Treonina Quinasas TOR , Análisis de Matrices Tisulares
18.
Am J Surg Pathol ; 31(6): 895-901, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17527077

RESUMEN

At the molecular level, alveolar rhabdomyosarcomas (ARMS) are characterized by 3 mutually exclusive PAX/FKHR conditions: PAX3/FKHR fusion (present in 60% of cases), PAX7/FKHR fusion (present in 20%), and PAX/FKHR fusion-negativity (present in 20%). The possibility of morphologic variation among these molecular subtypes has not been investigated. We undertook a blinded retrospective study of 65 cases of ARMS (16 PAX/FKHR fusion-negative, 36 PAX3/FKHR-positive, and 13 PAX7/FKHR-positive by routine reverse transcription-polymerase chain reaction). We evaluated cytohistologic parameters such as microcyst formation, solid foci, differentiation, giant cell formation, anaplasia, nuclear grade, mitosis/karyorrhexis index, rosette formation, geographic necrosis, presence and amount of rhabdomyoblastic differentiation, and the presence of foci resembling embryonal rhabdomyosarcoma. We analyzed the results using a simple chi formula. Of these features, only totally solid alveolar architecture reached significance (P=0.00014), with 7 of 16 PAX/FKHR-negative cases lacking this feature, compared with 0 of 36 PAX3/FKHR cases and 2/13 PAX7/FKHR cases. These preliminary results indicate that in general, only totally solid alveolar architecture in ARMS may predict the absence of a PAX/FKHR fusion. No features seemed to predict the presence of a particular fusion type. Our results suggest that histologic assessment of ARMS has limited correlation with PAX/FKHR fusion status.


Asunto(s)
Proteínas de Fusión Oncogénica/genética , Rabdomiosarcoma Alveolar/genética , Rabdomiosarcoma Alveolar/patología , Niño , Proteína Forkhead Box O1 , Factores de Transcripción Forkhead/genética , Humanos , Factor de Transcripción PAX3 , Factor de Transcripción PAX7/genética , Factores de Transcripción Paired Box/genética , Estudios Retrospectivos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
20.
Cancer Genet Cytogenet ; 174(1): 68-73, 2007 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-17350470

RESUMEN

Embryonal rhabdomyosarcoma (ERMS) is the most common subtype of RMS that predominantly involves the genitourinary tract and the head and neck regions in children younger than 10 years of age. Cytogenetically, ERMS is most frequently hyperdiploid, with extra copies of chromosomes 2, 7, 8, 11, 12, 13, and 20. No consistent structural chromosomal alteration has been identified in ERMS. In contrast, a t(2;13)(q35;q14) or t(1;13)(q36;q14) corresponding to PAX3-FOXO1A (previously FKHR) and PAX7-FOXO1A gene fusions are considered tumor-specific anomalies for alveolar RMS (ARMS). Occasionally, a recurrent secondary structural rearrangement involving chromosomes 1 and 16 is seen in translocation-positive ARMS, a der(16)t(1;16) resulting in an imbalance of 1q and 16q material. Conventional cytogenetic analysis of an ERMS arising in the urinary bladder of a 22-month-old male child revealed this nonrandom secondary chromosomal aberration, der(16)(1;16)(q22;q24), in a hyperdiploid complement with extra copies of chromosomes 2, 7, 8, 10, 12, 13, 19, and 20. Subsequent analyses showed tumor cells to be negative for FOXO1A, PAX3, or PAX7 gene locus rearrangements (by fluorescence in situ hybridization) and also negative for PAX3-FOXO1A and PAX7-FOXO1A fusion transcripts (by reverse transcriptase-polymerase chain reaction). These results suggest that the unbalanced t(1;16) translocation may be seen in RMSs lacking a primary genetic rearrangement.


Asunto(s)
Cromosomas Humanos Par 16/genética , Cromosomas Humanos Par 1/genética , Rabdomiosarcoma Embrionario/genética , Translocación Genética , Adolescente , Adulto , Niño , Preescolar , Bandeo Cromosómico , Femenino , Humanos , Lactante , Cariotipificación , Masculino , Rabdomiosarcoma Embrionario/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA