Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Genes Dev ; 30(22): 2459-2474, 2016 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-27940961

RESUMO

IMPs, also known as insulin-like growth factor 2 (IGF2) messenger RNA (mRNA)-binding proteins (IGF2BPs), are highly conserved oncofetal RNA-binding proteins (RBPs) that regulate RNA processing at several levels, including localization, translation, and stability. Three mammalian IMP paralogs (IMP1-3) have been identified that are expressed in most organs during embryogenesis, where they are believed to play an important role in cell migration, metabolism, and stem cell renewal. Whereas some IMP2 expression is retained in several adult mouse organs, IMP1 and IMP3 are either absent or expressed at very low levels in most tissues after birth. However, all three paralogs can be re-expressed upon malignant transformation and are found in a broad range of cancer types where their expression often correlates with poor prognosis. IMPs appear to resume their physiological functions in malignant cells, which not only contribute to tumor progression but participate in the establishment and maintenance of tumor cell hierarchies. This review summarizes our current understanding of the functions of IMPs during normal development and focuses on a series of recent observations that have provided new insight into how their physiological functions enable IMPs to play a potentially key role in cancer stem cell maintenance and tumor growth.


Assuntos
Desenvolvimento Embrionário/genética , Neoplasias/genética , Neoplasias/fisiopatologia , Proteínas de Ligação a RNA/metabolismo , Células-Tronco/fisiologia , Animais , Regulação da Expressão Gênica no Desenvolvimento , Regulação Neoplásica da Expressão Gênica , Humanos , Processamento Pós-Transcricional do RNA/genética , Proteínas de Ligação a RNA/genética , Células-Tronco/metabolismo
2.
Swiss Med Wkly ; 144: w14018, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25356909

RESUMO

Gliomas are the most common primary intrinsic brain tumours. Their classification is based on phenotypic resemblance to normal glial cells (astrocytomas, oligodendrogliomas, mixed oligoastrocytomas) and pathological grading. Whereas this system is clinically relevant and has been the basis for our understanding of gliomas, systematic use of next-generation sequencing has transformed our knowledge of their pathogenesis and has uncovered genetic changes in an unanticipated number of genes and regulatory elements. In the past few years, in-depth analysis of low-grade astrocytomas and glioblastomas in both paediatric and adult populations has clarified our molecular understanding of these diseases, with distinct molecular events occurring in different age groups. In oligodendrogliomas, recent studies have highlighted mutations in candidate tumour suppressor genes located on 1p/19q, chromosome arms frequently deleted in this tumour. In this review, we discuss recent discoveries in the genetics of adult and paediatric gliomas, and highlight how some of the founding genetic mutations reshape the cancer epigenome. These studies provide an in-depth view of the molecular routes leading to brain tumour development and will be key for refining classification systems and improving clinical care.


Assuntos
Neoplasias Encefálicas/genética , Epigênese Genética , Glioma/genética , Proteínas Adaptadoras de Transdução de Sinal/genética , Adulto , Fatores Etários , Astrocitoma/genética , Criança , Proteínas Correpressoras , DNA Helicases/genética , Histona-Lisina N-Metiltransferase/genética , Histonas/genética , Humanos , Isocitrato Desidrogenase/genética , Chaperonas Moleculares , Gradação de Tumores , Proteínas Nucleares/genética , Oligodendroglioma/genética , Fenótipo , Receptores Proteína Tirosina Quinases/genética , Proteína Nuclear Ligada ao X
3.
Cancer Cell ; 21(6): 807-21, 2012 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-22698405

RESUMO

We have recently demonstrated that human pediatric mesenchymal stem cells can be reprogrammed toward a Ewing sarcoma family tumor (ESFT) cancer stem cell (CSC) phenotype by mechanisms that implicate microRNAs (miRNAs). Here, we show that the miRNA profile of ESFT CSCs is shared by embryonic stem cells and CSCs from divergent tumor types. We also provide evidence that the miRNA profile of ESFT CSCs is the result of reversible disruption of TARBP2-dependent miRNA maturation. Restoration of TARBP2 activity and systemic delivery of synthetic forms of either of two of its targets, miRNA-143 or miRNA-145, inhibited ESFT CSC clonogenicity and tumor growth in vivo. Our observations suggest that CSC self-renewal and tumor maintenance may depend on deregulation of TARBP2-dependent miRNA expression.


Assuntos
Perfilação da Expressão Gênica , MicroRNAs/genética , Células-Tronco Neoplásicas/metabolismo , Proteínas de Ligação a RNA/genética , Animais , Linhagem Celular Tumoral , Células Cultivadas , Criança , Pré-Escolar , Células-Tronco Embrionárias/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Masculino , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos Endogâmicos NOD , Camundongos Knockout , Camundongos SCID , Mutação , Neoplasias Experimentais/genética , Neoplasias Experimentais/metabolismo , Neoplasias Experimentais/patologia , Células-Tronco Neoplásicas/patologia , Proteínas de Ligação a RNA/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sarcoma de Ewing/genética , Sarcoma de Ewing/patologia , Sarcoma de Ewing/terapia , Transplante Heterólogo
4.
PLoS One ; 6(8): e23592, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21853155

RESUMO

Ewing's sarcoma family tumors (ESFT) are the second most common bone malignancy in children and young adults, characterized by unique chromosomal translocations that in 85% of cases lead to expression of the EWS-FLI-1 fusion protein. EWS-FLI-1 functions as an aberrant transcription factor that can both induce and suppress members of its target gene repertoire. We have recently demonstrated that EWS-FLI-1 can alter microRNA (miRNA) expression and that miRNA145 is a direct EWS-FLI-1 target whose suppression is implicated in ESFT development. Here, we use miRNA arrays to compare the global miRNA expression profile of human mesenchymal stem cells (MSC) and ESFT cell lines, and show that ESFT display a distinct miRNA signature that includes induction of the oncogenic miRNA 17-92 cluster and repression of the tumor suppressor let-7 family. We demonstrate that direct repression of let-7a by EWS-FLI-1 participates in the tumorigenic potential of ESFT cells in vivo. The mechanism whereby let-7a expression regulates ESFT growth is shown to be mediated by its target gene HMGA2, as let-7a overexpression and HMGA2 repression both block ESFT cell tumorigenicity. Consistent with these observations, systemic delivery of synthetic let-7a into ESFT-bearing mice restored its expression in tumor cells, decreased HMGA2 expression levels and resulted in ESFT growth inhibition in vivo. Our observations provide evidence that deregulation of let-7a target gene expression participates in ESFT development and identify let-7a as promising new therapeutic target for one of the most aggressive pediatric malignancies.


Assuntos
MicroRNAs/metabolismo , Proteínas de Fusão Oncogênica/metabolismo , Lesões Pré-Cancerosas/patologia , Proteína Proto-Oncogênica c-fli-1/metabolismo , Proteína EWS de Ligação a RNA/metabolismo , Sarcoma de Ewing/patologia , Linhagem Celular Tumoral , Proliferação de Células , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/patologia , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Proteína HMGA2/genética , Proteína HMGA2/metabolismo , Humanos , Células-Tronco Mesenquimais/metabolismo , MicroRNAs/genética , Lesões Pré-Cancerosas/genética , Sarcoma de Ewing/genética
6.
Genes Dev ; 24(9): 916-32, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20382729

RESUMO

Cancer stem cells (CSCs) display plasticity and self-renewal properties reminiscent of normal tissue stem cells, but the events responsible for their emergence remain obscure. We recently identified CSCs in Ewing sarcoma family tumors (ESFTs) and showed that they retain mesenchymal stem cell (MSC) plasticity. In the present study, we addressed the mechanisms that underlie ESFT CSC development. We show that the EWS-FLI-1 fusion gene, associated with 85%-90% of ESFTs and believed to initiate their pathogenesis, induces expression of the embryonic stem cell (ESC) genes OCT4, SOX2, and NANOG in human pediatric MSCs (hpMSCs) but not in their adult counterparts. Moreover, under appropriate culture conditions, hpMSCs expressing EWS-FLI-1 generate a cell subpopulation displaying ESFT CSC features in vitro. We further demonstrate that induction of the ESFT CSC phenotype is the result of the combined effect of EWS-FLI-1 on its target gene expression and repression of microRNA-145 (miRNA145) promoter activity. Finally, we provide evidence that EWS-FLI-1 and miRNA-145 function in a mutually repressive feedback loop and identify their common target gene, SOX2, in addition to miRNA145 itself, as key players in ESFT cell differentiation and tumorigenicity. Our observations provide insight for the first time into the mechanisms whereby a single oncogene can reprogram primary cells to display a CSC phenotype.


Assuntos
Reprogramação Celular , Regulação Neoplásica da Expressão Gênica , Células-Tronco Mesenquimais/citologia , MicroRNAs/metabolismo , Proteína Proto-Oncogênica c-fli-1/metabolismo , Proteína EWS de Ligação a RNA/metabolismo , Fatores de Transcrição SOXB1/metabolismo , Adolescente , Adulto , Diferenciação Celular , Linhagem Celular Tumoral , Criança , Proteínas de Homeodomínio/metabolismo , Humanos , Proteína Homeobox Nanog , Células-Tronco Neoplásicas/citologia , Células-Tronco Neoplásicas/metabolismo , Fator 3 de Transcrição de Octâmero/metabolismo , Fenótipo , Sarcoma de Ewing/fisiopatologia , Células Tumorais Cultivadas
7.
Cancer Res ; 69(24): 9211-8, 2009 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-19934320

RESUMO

Overexpression of the polycomb group protein enhancer of zeste homologue 2 (EZH2) occurs in diverse malignancies, including prostate cancer, breast cancer, and glioblastoma multiforme (GBM). Based on its ability to modulate transcription of key genes implicated in cell cycle control, DNA repair, and cell differentiation, EZH2 is believed to play a crucial role in tissue-specific stem cell maintenance and tumor development. Here, we show that targeted pharmacologic disruption of EZH2 by the S-adenosylhomocysteine hydrolase inhibitor 3-deazaneplanocin A (DZNep), or its specific downregulation by short hairpin RNA (shRNA), strongly impairs GBM cancer stem cell (CSC) self-renewal in vitro and tumor-initiating capacity in vivo. Using genome-wide expression analysis of DZNep-treated GBM CSCs, we found the expression of c-myc, recently reported to be essential for GBM CSCs, to be strongly repressed upon EZH2 depletion. Specific shRNA-mediated downregulation of EZH2 in combination with chromatin immunoprecipitation experiments revealed that c-myc is a direct target of EZH2 in GBM CSCs. Taken together, our observations provide evidence that direct transcriptional regulation of c-myc by EZH2 may constitute a novel mechanism underlying GBM CSC maintenance and suggest that EZH2 may be a valuable new therapeutic target for GBM management.


Assuntos
Proteínas de Ligação a DNA/biossíntese , Glioblastoma/patologia , Células-Tronco Neoplásicas/patologia , Fatores de Transcrição/biossíntese , Animais , Linhagem Celular Tumoral , Proteínas de Ligação a DNA/antagonistas & inibidores , Proteínas de Ligação a DNA/genética , Regulação para Baixo , Proteína Potenciadora do Homólogo 2 de Zeste , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Genes myc , Glioblastoma/genética , Glioblastoma/metabolismo , Humanos , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Células-Tronco Neoplásicas/metabolismo , Complexo Repressor Polycomb 2 , RNA Interferente Pequeno/genética , Fatores de Transcrição/antagonistas & inibidores , Fatores de Transcrição/genética
8.
PLoS One ; 4(11): e7904, 2009 Nov 19.
Artigo em Inglês | MEDLINE | ID: mdl-19936258

RESUMO

BACKGROUND: A characteristic SYT-SSX fusion gene resulting from the chromosomal translocation t(X;18)(p11;q11) is detectable in almost all synovial sarcomas, a malignant soft tissue tumor widely believed to originate from as yet unidentified pluripotent stem cells. The resulting fusion protein has no DNA binding motifs but possesses protein-protein interaction domains that are believed to mediate association with chromatin remodeling complexes. Despite recent advances in the identification of molecules that interact with SYT-SSX and with the corresponding wild type SYT and SSX proteins, the mechanisms whereby the SYT-SSX might contribute to neoplastic transformation remain unclear. Epigenetic deregulation has been suggested to be one possible mechanism. METHODOLOGY/PRINCIPAL FINDINGS: We addressed the effect of SYT/SSX expression on the transcriptome of four independent isolates of primary human bone marrow mesenchymal stem cells (hMSC). We observed transcriptional changes similar to the gene expression signature of synovial sarcoma, principally involving genes whose regulation is linked to epigenetic factors, including imprinted genes, genes with transcription start sites within a CpG island and chromatin related genes. Single population analysis revealed hMSC isolate-specific transcriptional changes involving genes that are important for biological functions of stem cells as well as genes that are considered to be molecular markers of synovial sarcoma including IGF2, EPHRINS, and BCL2. Methylation status analysis of sequences at the H19/IGF2 imprinted locus indicated that distinct epigenetic features characterize hMSC populations and condition the transcriptional effects of SYT-SSX expression. CONCLUSIONS/SIGNIFICANCE: Our observations suggest that epigenetic features may define the cellular microenvironment in which SYT-SSX displays its functional effects.


Assuntos
Epigênese Genética , Regulação da Expressão Gênica , Células-Tronco Mesenquimais/citologia , Proteínas de Fusão Oncogênica/genética , Proteínas de Fusão Oncogênica/fisiologia , Adolescente , Alelos , Criança , Cromatina/metabolismo , Ilhas de CpG , DNA/genética , Perfilação da Expressão Gênica , Humanos , Sarcoma Sinovial/metabolismo , Transcrição Gênica , Translocação Genética
10.
PLoS One ; 4(8): e6510, 2009 Aug 04.
Artigo em Inglês | MEDLINE | ID: mdl-19652710

RESUMO

BACKGROUND: Characteristic symptoms of malaria include recurrent fever attacks and neurodegeneration, signs that are also found in patients with a hyperactive Nalp3 inflammasome. Plasmodium species produce a crystal called hemozoin that is generated by detoxification of heme after hemoglobin degradation in infected red blood cells. Thus, we hypothesized that hemozoin could activate the Nalp3 inflammasome, due to its particulate nature reminiscent of other inflammasome-activating agents. METHODOLOGY/PRINCIPAL FINDINGS: We found that hemozoin acts as a proinflammatory danger signal that activates the Nalp3 inflammasome, causing the release of IL-1beta. Similar to other Nalp3-activating particles, hemozoin activity is blocked by inhibiting phagocytosis, K(+) efflux and NADPH oxidase. In vivo, intraperitoneal injection of hemozoin results in acute peritonitis, which is impaired in Nalp3-, caspase-1- and IL-1R-deficient mice. Likewise, the pathogenesis of cerebral malaria is dampened in Nalp3-deficient mice infected with Plasmodium berghei sporozoites, while parasitemia remains unchanged. SIGNIFICANCE/CONCLUSIONS: The potent pro-inflammatory effect of hemozoin through inflammasome activation may possibly be implicated in plasmodium-associated pathologies such as cerebral malaria.


Assuntos
Hemeproteínas/fisiologia , Plasmodium berghei/fisiologia , Proteínas de Protozoários/fisiologia , Animais , Interleucina-1beta/metabolismo , Camundongos , Fagocitose
11.
Cancer Res ; 69(5): 1776-81, 2009 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-19208848

RESUMO

Cancer stem cells that display tumor-initiating properties have recently been identified in several distinct types of malignancies, holding promise for more effective therapeutic strategies. However, evidence of such cells in sarcomas, which include some of the most aggressive and therapy-resistant tumors, has not been shown to date. Here, we identify and characterize cancer stem cells in Ewing's sarcoma family tumors (ESFT), a highly aggressive pediatric malignancy believed to be of mesenchymal stem cell (MSC) origin. Using magnetic bead cell separation of primary ESFT, we have isolated a subpopulation of CD133+ tumor cells that display the capacity to initiate and sustain tumor growth through serial transplantation in nonobese diabetic/severe combined immunodeficiency mice, re-establishing at each in vivo passage the parental tumor phenotype and hierarchical cell organization. Consistent with the plasticity of MSCs, in vitro differentiation assays showed that the CD133+ cell population retained the ability to differentiate along adipogenic, osteogenic, and chondrogenic lineages. Quantitative real-time PCR analysis of genes implicated in stem cell maintenance revealed that CD133+ ESFT cells express significantly higher levels of OCT4 and NANOG than their CD133- counterparts. Taken together, our observations provide the first identification of ESFT cancer stem cells and demonstration of their MSC properties, a critical step towards a better biological understanding and rational therapeutic targeting of these tumors.


Assuntos
Neoplasias Ósseas/patologia , Separação Imunomagnética/métodos , Células-Tronco Neoplásicas/patologia , Sarcoma de Ewing/patologia , Antígeno AC133 , Animais , Antígenos CD/análise , Linhagem Celular Tumoral , Glicoproteínas/análise , Humanos , Camundongos , Peptídeos/análise
12.
PLoS One ; 3(7): e2634, 2008 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-18648544

RESUMO

BACKGROUND: The EWS-FLI-1 fusion protein is associated with 85-90% of Ewing's sarcoma family tumors (ESFT), the remaining 10-15% of cases expressing chimeric genes encoding EWS or FUS fused to one of several ets transcription factor family members, including ERG-1, FEV, ETV1 and ETV6. ESFT are dependent on insulin-like growth factor-1 (IGF-1) for growth and survival and recent evidence suggests that mesenchymal progenitor/stem cells constitute a candidate ESFT origin. METHODOLOGY/PRINCIPAL FINDINGS: To address the functional relatedness between ESFT-associated fusion proteins, we compared mouse progenitor cell (MPC) permissiveness for EWS-FLI-1, EWS-ERG and FUS-ERG expression and assessed the corresponding expression profile changes. Whereas all MPC isolates tested could stably express EWS-FLI-1, only some sustained stable EWS-ERG expression and none could express FUS-ERG for more than 3-5 days. Only 14% and 4% of the total number of genes that were respectively induced and repressed in MPCs by the three fusion proteins were shared. However, all three fusion proteins, but neither FLI-1 nor ERG-1 alone, activated the IGF1 promoter and induced IGF1 expression. CONCLUSION/SIGNIFICANCE: Whereas expression of different ESFT-associated fusion proteins may require distinct cellular microenvironments and induce transcriptome changes of limited similarity, IGF1 induction may provide one common mechanism for their implication in ESFT pathogenesis.


Assuntos
Regulação da Expressão Gênica , Fator de Crescimento Insulin-Like I/metabolismo , Células-Tronco Mesenquimais/citologia , Proteínas de Fusão Oncogênica/química , Proteínas de Fusão Oncogênica/metabolismo , Proteína Proto-Oncogênica c-fli-1/química , Proteína FUS de Ligação a RNA/metabolismo , Fatores de Transcrição/metabolismo , Animais , Perfilação da Expressão Gênica , Humanos , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Modelos Biológicos , Fenótipo , Regiões Promotoras Genéticas , Proteína Proto-Oncogênica c-fli-1/metabolismo , Proteína EWS de Ligação a RNA
13.
Cancer Res ; 68(7): 2176-85, 2008 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-18381423

RESUMO

Ewing's sarcoma family tumors (ESFT) express the EWS-FLI-1 fusion gene generated by the chromosomal translocation t(11;22)(q24;q12). Expression of the EWS-FLI-1 fusion protein in a permissive cellular environment is believed to play a key role in ESFT pathogenesis. However, EWS-FLI-1 induces growth arrest or apoptosis in differentiated primary cells, and the identity of permissive primary human cells that can support its expression and function has until now remained elusive. Here we show that expression of EWS-FLI-1 in human mesenchymal stem cells (hMSC) is not only stably maintained without inhibiting proliferation but also induces a gene expression profile bearing striking similarity to that of ESFT, including genes that are among the highest ESFT discriminators. Expression of EWS-FLI-1 in hMSCs may recapitulate the initial steps of Ewing's sarcoma development, allowing identification of genes that play an important role early in its pathogenesis. Among relevant candidate transcripts induced by EWS-FLI-1 in hMSCs, we found the polycomb group gene EZH2, which we show to play a critical role in Ewing's sarcoma growth. These observations are consistent with our recent findings using mouse mesenchymal progenitor cells and provide compelling evidence that hMSCs are candidate cells of origin of ESFT.


Assuntos
Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/patologia , Proteínas de Fusão Oncogênica/biossíntese , Proteína Proto-Oncogênica c-fli-1/biossíntese , Sarcoma de Ewing/metabolismo , Sarcoma de Ewing/patologia , Animais , Neoplasias Ósseas/genética , Neoplasias Ósseas/metabolismo , Neoplasias Ósseas/patologia , Diferenciação Celular/fisiologia , Proteína Potenciadora do Homólogo 2 de Zeste , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica/genética , Histona-Lisina N-Metiltransferase , Humanos , Hospedeiro Imunocomprometido , Camundongos , Proteínas de Fusão Oncogênica/genética , Fenótipo , Complexo Repressor Polycomb 2 , Proteínas/genética , Proteína Proto-Oncogênica c-fli-1/genética , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Proteína EWS de Ligação a RNA , Sarcoma de Ewing/genética , Neoplasias de Tecidos Moles/genética , Neoplasias de Tecidos Moles/metabolismo , Neoplasias de Tecidos Moles/patologia
14.
Cancer Res ; 66(14): 7016-23, 2006 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-16849546

RESUMO

A subset of sarcomas is associated with specific chromosomal translocations that give rise to fusion genes believed to participate in transformation and oncogenesis. Identification of the primary cell environment that provides permissiveness for the oncogenic potential of these fusion genes is essential to understand sarcoma pathogenesis. We have recently shown that expression of the EWS-FLI-1 fusion protein in primary mesenchymal progenitor cells (MPCs) suffices to develop Ewing's sarcoma-like tumors in mice. Because most sarcomas bearing unique chromosomal translocations are believed to originate from common progenitor cells, and because MPCs populate most organs, we expressed the sarcoma-associated fusion proteins FUS/TLS-CHOP, EWS-ATF1, and SYT-SSX1 in MPCs and tested the tumorigenic potential of these cells in vivo. Whereas expression of EWS-ATF1 and SYT-SSX1 failed to transform MPCs, FUS-CHOP-expressing cells formed tumors resembling human myxoid liposarcoma. Transcription profile analysis of these tumors revealed induction of transcripts known to be associated with myxoid liposarcoma and novel candidate genes, including PDGFA, whose expression was confirmed in human tumor samples. MPC(FUS-CHOP) and the previously described MPC(EWS-FLI-1) tumors displayed distinct transcription profiles, consistent with the different target gene repertoires of their respective fusion proteins. Unexpectedly, a set of genes implicated in cell survival and adhesion displayed similar behavior in the two tumors, suggesting events that may be common to primary MPC transformation. Taken together, our observations suggest that expression of FUS-CHOP may be the initiating event in myxoid liposarcoma pathogenesis, and that MPCs may constitute one cell type from which these tumors originate.


Assuntos
Transformação Celular Neoplásica/genética , Lipossarcoma Mixoide/metabolismo , Lipossarcoma Mixoide/patologia , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/patologia , Proteínas de Fusão Oncogênica/biossíntese , Proteína FUS de Ligação a RNA/biossíntese , Fator de Transcrição CHOP/biossíntese , Animais , Células da Medula Óssea/metabolismo , Células da Medula Óssea/patologia , Células da Medula Óssea/fisiologia , Transformação Celular Neoplásica/metabolismo , Transformação Celular Neoplásica/patologia , Humanos , Lipossarcoma Mixoide/genética , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos SCID , Proteínas de Fusão Oncogênica/genética , Proteína FUS de Ligação a RNA/genética , Fator de Transcrição CHOP/genética , Transfecção
15.
Cancer Res ; 65(24): 11459-68, 2005 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-16357154

RESUMO

Ewing's sarcoma is a member of Ewing's family tumors (EFTs) and the second most common solid bone and soft tissue malignancy of children and young adults. It is associated in 85% of cases with the t(11;22)(q24:q12) chromosomal translocation that generates fusion of the 5' segment of the EWS gene with the 3' segment of the ETS family gene FLI-1. The EWS-FLI-1 fusion protein behaves as an aberrant transcriptional activator and is believed to contribute to EFT development. However, EWS-FLI-1 induces growth arrest and apoptosis in normal fibroblasts, and primary cells that are permissive for its putative oncogenic properties have not been discovered, hampering basic understanding of EFT biology. Here, we show that EWS-FLI-1 alone can transform primary bone marrow-derived mesenchymal progenitor cells and generate tumors that display hallmarks of Ewing's sarcoma, including a small round cell phenotype, expression of EFT-associated markers, insulin like growth factor-I dependence, and induction or repression of numerous EWS-FLI-1 target genes. These observations provide the first identification of candidate primary cells from which EFTs originate and suggest that EWS-FLI-1 expression may constitute the initiating event in EFT pathogenesis.


Assuntos
Células da Medula Óssea/patologia , Transformação Celular Neoplásica , Células-Tronco Mesenquimais/patologia , Sarcoma de Ewing/patologia , Animais , Biomarcadores Tumorais/metabolismo , Inibidor de Quinase Dependente de Ciclina p19/metabolismo , Fibroblastos/metabolismo , Fibroblastos/patologia , Perfilação da Expressão Gênica , Humanos , Fator de Crescimento Insulin-Like I/metabolismo , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos SCID , Análise de Sequência com Séries de Oligonucleotídeos , Proteínas de Fusão Oncogênica/metabolismo , Fenótipo , Proteína Proto-Oncogênica c-fli-1/metabolismo , Proteína EWS de Ligação a RNA , Sarcoma de Ewing/etiologia , Sarcoma de Ewing/metabolismo , Células-Tronco/citologia , Células-Tronco/metabolismo , Proteína Supressora de Tumor p53/metabolismo
16.
Neurocase ; 11(3): 216-26, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16006343

RESUMO

A 79-year-old patient with neuropathologically confirmed Alzheimer's disease (AD) presented with a selective environmental reduplicative paramnesia (RP), the belief that one or more environments exist simultaneously in two or more physical locations. Clinical presentation and neuropathological examination revealed an atypical form of AD. High neurofibrillary tangle densities were observed in the frontal and temporal association cortex, whereas the parietal and entorhinal cortex, as well as the hippocampus, were nearly spared. These findings are compared to those reported in frontal and frontotemporal variants of AD and discussed in the light of current anatomoclinical models for environmental RP.


Assuntos
Doença de Alzheimer/complicações , Amnésia/etiologia , Esquizofrenia Paranoide/etiologia , Atividades Cotidianas , Idoso , Doença de Alzheimer/patologia , Doença de Alzheimer/fisiopatologia , Atenção/fisiologia , Córtex Cerebral/patologia , Cognição/fisiologia , Emoções , Meio Ambiente , Seguimentos , Lateralidade Funcional , Humanos , Inteligência/fisiologia , Idioma , Masculino , Emaranhados Neurofibrilares/patologia , Orientação/fisiologia , Desempenho Psicomotor/fisiologia , Tomografia Computadorizada por Raios X/métodos , Percepção Visual/fisiologia
17.
Cancer Res ; 64(24): 8945-53, 2004 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-15604257

RESUMO

Endothelin receptor B (ETRB or EDNRB) is overexpressed in most human melanomas and is proposed to provide a marker of melanoma progression. We have shown previously that inhibition of ETRB leads to increased human melanoma cell death in vitro and in vivo, resulting in shrinkage of tumors grown in immunocompromised mice. In the present work, we analyzed the effects of ETRB inhibition on 10 human melanoma cell lines derived from tumors at distinct stages of progression. Our observations suggest that the ETRB antagonist BQ788 induces apoptosis most effectively in metastatic melanoma cells. Microarray analysis shows that BQ788 treatment leads to a reduction in the expression of the survival factor BCL-2A1 and the DNA repair factor poly(ADP-ribose) polymerase 3 that is more pronounced in cells derived from metastatic than primary melanoma. Decreased cell viability was observed to correlate with reduction in ETRB expression, and reduction in ETRB protein levels by small interfering RNA led to an increase in cell death. Interestingly, reduction of ETRB expression by BQ788 was accompanied by a strong induction of VEGF expression and repression of the angiogenic suppressor gravin. These changes in gene expression correlated with increased angiogenesis in tumors injected with ETRB antagonist in vivo. Taken together, our observations suggest that ETRB may provide a potential therapeutic target in high-grade melanomas and identify candidate pathways that may be implicated in the regulation of cell survival and tumor progression associated with ETRB signaling.


Assuntos
Apoptose/efeitos dos fármacos , Antagonistas do Receptor de Endotelina B , Melanoma/irrigação sanguínea , Melanoma/patologia , Oligopeptídeos/farmacologia , Piperidinas/farmacologia , Apoptose/fisiologia , Caspase 6 , Caspases/metabolismo , Proteínas de Ciclo Celular/biossíntese , Linhagem Celular Tumoral , Progressão da Doença , Ativação Enzimática , Humanos , Metástase Linfática , Melanoma/tratamento farmacológico , Melanoma/secundário , Neovascularização Patológica/metabolismo , Neovascularização Patológica/patologia , Análise de Sequência com Séries de Oligonucleotídeos , Poli(ADP-Ribose) Polimerases/biossíntese , Neoplasias Cutâneas/secundário
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...