Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros








Intervalo de ano de publicação
1.
Oncotarget ; 4(10): 1737-47, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24077805

RESUMO

Somatic mutations in Isocitrate Dehydrogenase 1 (IDH1) are frequent in low grade and progressive gliomas and are characterized by the production of 2-hydroxyglutarate (2-HG) from α-ketoglutarate by the mutant enzyme. 2-HG is an "oncometabolite" that competitively inhibits α-KG dependent dioxygenases resulting in various widespread cellular changes including abnormal hypermethylation of genomic DNA and suppression of cellular differentiation. Despite the growing understanding of IDH mutant gliomas, the development of effective therapies has proved challenging in part due to the scarcity of endogenous mutant in vivo models. Here we report the generation of an endogenous IDH1 anaplastic astrocytoma model which rapidly grows in vivo, produces 2-HG and exhibits DNA hypermethylation. Using this model, we have demonstrated the preclinical efficacy and mechanism of action of the FDA approved demethylating drug 5-azacytidine in vivo. Long term administration of 5-azacytidine resulted in reduction of DNA methylation of promoter loci, induction of glial differentiation, reduction of cell proliferation and a significant reduction in tumor growth. Tumor regression was observed at 14 weeks and subsequently showed no signs of re-growth at 7 weeks despite discontinuation of therapy. These results have implications for clinical trials of demethylating agents for patients with IDH mutated gliomas.


Assuntos
Antimetabólitos Antineoplásicos/farmacologia , Azacitidina/farmacologia , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/enzimologia , Glioma/tratamento farmacológico , Glioma/enzimologia , Isocitrato Desidrogenase/genética , Animais , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patologia , Diferenciação Celular/efeitos dos fármacos , Processos de Crescimento Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Metilação de DNA/efeitos dos fármacos , Feminino , Glioma/genética , Glioma/patologia , Humanos , Imuno-Histoquímica , Isocitrato Desidrogenase/metabolismo , Masculino , Camundongos , Camundongos Nus , Mutação , Ensaios Antitumorais Modelo de Xenoenxerto
2.
Cancer Immunol Res ; 1(5): 296-302, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24777967

RESUMO

Meningiomas are the most common primary intracranial tumors. Surgical resection remains the treatment of choice for these tumors. However, a significant number of tumors are not surgically accessible, recur, or become malignant, necessitating the repetition of surgery and sometimes radiation. Chemotherapy is rarely used and is generally not recognized as an effective treatment. Cancer/testis (CT) genes represent a unique class of genes, which are expressed by germ cells, normally silenced in somatic cells, but activated in various cancers. CT proteins can elicit spontaneous immune responses in patients with cancer and this feature makes them attractive targets for immunotherapy-based approaches. We analyzed mRNA expression of 37 testis-restricted CT genes in a discovery set of 18 meningiomas by reverse transcription PCR. The overall frequency of expression of CT genes ranged from 5.6% to 27.8%. The most frequently expressed was NY-ESO-1, in 5 patients (27.8%). We subsequently analyzed NY-ESO-1 protein expression in a larger set of meningiomas by immunohistochemistry and found expression in 108 of 110 cases. In some cases, NY-ESO-1 expression was diffused and homogenous, but in most instances it was heterogeneous. Importantly, NY-ESO-1 expression was positively correlated with higher grade and patients presenting with higher levels of NY-ESO-1 staining had significantly worse disease-free and overall survival. We have also shown that NY-ESO-1 expression may lead to humoral immune response in patients with meningioma. Considering the limited treatment options for patients with meningioma, the potential of NY-ESO-1-based immunotherapy should be explored.


Assuntos
Antígenos de Neoplasias/biossíntese , Proteínas de Membrana/biossíntese , Neoplasias Meníngeas/imunologia , Neoplasias Meníngeas/terapia , Meningioma/imunologia , Meningioma/terapia , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Anticorpos Antineoplásicos/biossíntese , Anticorpos Antineoplásicos/genética , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/imunologia , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Imuno-Histoquímica , Imunoterapia/métodos , Masculino , Proteínas de Membrana/genética , Proteínas de Membrana/imunologia , Neoplasias Meníngeas/genética , Meningioma/genética , Pessoa de Meia-Idade , Recidiva Local de Neoplasia/imunologia , Recidiva Local de Neoplasia/terapia , Adulto Jovem
3.
Mol Cancer Res ; 10(7): 904-13, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22618028

RESUMO

The Hippo signaling pathway is functionally conserved in Drosophila melanogaster and mammals, and its proposed function is to control tissue homeostasis by regulating cell proliferation and apoptosis. The core components are composed of a kinase cascade that culminates with the phosphorylation and inhibition of Yes-associated protein 1 (YAP1). Phospho-YAP1 is retained in the cytoplasm. In the absence of Hippo signaling, YAP1 translocates to the nucleus, associates with co-activators TEAD1-4, and functions as a transcriptional factor promoting the expression of key target genes. Components of the Hippo pathway are mutated in human cancers, and deregulation of this pathway plays a role in tumorigenesis. Loss of the NF2 tumor suppressor gene is the most common genetic alteration in meningiomas, and the NF2 gene product, Merlin, acts upstream of the Hippo pathway. Here, we show that primary meningioma tumors have high nuclear expression of YAP1. In meningioma cells, Merlin expression is associated with phosphorylation of YAP1. Using an siRNA transient knockdown of YAP1 in NF2-mutant meningioma cells, we show that suppression of YAP1 impaired cell proliferation and migration. Conversely, YAP1 overexpression led to a strong augment of cell proliferation and anchorage-independent growth and restriction of cisplatin-induced apoptosis. In addition, expression of YAP1 in nontransformed arachnoidal cells led to the development of tumors in nude mice. Together, these findings suggest that in meningiomas, deregulation of the Hippo pathway is largely observed in primary tumors and that YAP1 functions as an oncogene promoting meningioma tumorigenesis.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Neoplasias Encefálicas , Meningioma , Fosfoproteínas , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Apoptose/efeitos dos fármacos , Apoptose/genética , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Cisplatino/farmacologia , Regulação Neoplásica da Expressão Gênica , Técnicas de Silenciamento de Genes , Humanos , Meningioma/genética , Meningioma/metabolismo , Camundongos , Neoplasias Experimentais/metabolismo , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , RNA Interferente Pequeno , Análise Serial de Tecidos , Fatores de Transcrição , Cicatrização , Proteínas de Sinalização YAP
5.
Biochemistry ; 48(28): 6633-43, 2009 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-19530647

RESUMO

The eukaryotic single-stranded DNA-binding protein, replication protein A (RPA), is essential in DNA metabolism and is phosphorylated in response to DNA-damaging agents. Rad52 and RPA participate in the repair of double-stranded DNA breaks (DSBs). It is known that human RPA and Rad52 form a complex, but the molecular mass, stoichiometry, and exact role of this complex in DSB repair are unclear. In this study, absolute molecular masses of individual proteins and complexes were measured in solution using analytical size-exclusion chromatography coupled with multiangle light scattering, the protein species present in each purified fraction were verified via sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE)/Western analyses, and the presence of biotinylated ssDNA in the complexes was verified by chemiluminescence detection. Then, employing UV cross-linking, the protein partner holding the ssDNA was identified. These data show that phosphorylated RPA promoted formation of a complex with monomeric Rad52 and caused the transfer of ssDNA from RPA to Rad52. This suggests that RPA phosphorylation may regulate the first steps of DSB repair and is necessary for the mediator function of Rad52.


Assuntos
Reparo do DNA , DNA de Cadeia Simples/metabolismo , Proteína Rad52 de Recombinação e Reparo de DNA/metabolismo , Proteína de Replicação A/metabolismo , Cromatografia em Gel , DNA de Cadeia Simples/ultraestrutura , Humanos , Luz , Modelos Biológicos , Fosforilação , Proteína Rad52 de Recombinação e Reparo de DNA/ultraestrutura , Proteína de Replicação A/ultraestrutura , Espalhamento de Radiação
6.
Neoplasia ; 10(11): 1204-12, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18953429

RESUMO

Neurofibromatosis type 2 (NF2) is an autosomal dominant disorder characterized by the occurrence of schwannomas and meningiomas. Several studies have examined the ability of the NF2 gene product, merlin, to function as a tumor suppressor in diverse cell types; however, little is known about merlin growth regulation in meningiomas. In Drosophila, merlin controls cell proliferation and apoptosis by signaling through the Hippo pathway to inhibit the function of the transcriptional coactivator Yorkie. The Hippo pathway is conserved in mammals. On the basis of these observations, we developed human meningioma cell lines matched for merlin expression to evaluate merlin growth regulation and investigate the relationship between NF2 status and Yes-associated protein (YAP), the mammalian homolog of Yorkie. NF2 loss in meningioma cells was associated with loss of contact-dependent growth inhibition, enhanced anchorage-independent growth and increased cell proliferation due to increased S-phase entry. In addition, merlin loss in both meningioma cell lines and primary tumors resulted in increased YAP expression and nuclear localization. Finally, siRNA-mediated reduction of YAP in NF2-deficient meningioma cells rescued the effects of merlin loss on cell proliferation and S-phase entry. Collectively, these results represent the first demonstration that merlin regulates cell growth in human cancer cells by suppressing YAP.


Assuntos
Meningioma/metabolismo , Neurofibromina 2/metabolismo , Proteínas Nucleares/metabolismo , Transdução de Sinais , Fatores de Transcrição/metabolismo , Aracnoide-Máter/citologia , Aracnoide-Máter/metabolismo , Proteínas de Ciclo Celular , Linhagem Celular , Linhagem Celular Tumoral , Proliferação de Células , Clonagem Molecular , Ciclina E/metabolismo , Genes da Neurofibromatose 2 , Humanos , Meningioma/genética , Meningioma/patologia , Neurofibromina 2/genética , Proteínas Nucleares/genética , Proteínas Oncogênicas/metabolismo , Reação em Cadeia da Polimerase , Interferência de RNA , Fatores de Transcrição/genética
7.
Neoplasia ; 10(6): 604-12, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18516297

RESUMO

The Notch signaling cascade is deregulated in diverse cancer types. Specific Notch function in cancer is dependent on the cellular context, the particular homologs expressed, and cross-talk with other signaling pathways. We have previously shown that components of the Notch signaling pathway are deregulated in meningiomas. However, the functional consequence of abnormal Notch signaling to meningiomas is unknown. Here, we report that exogenous expression of the Notch pathway effector, HES1, is associated with tetraploid cells in meningioma cell lines. Activated Notch1 and Notch2 receptors induced endogenous HES1 expression and were associated with tetraploidy in meningiomas. Tetraploid meningioma cells exhibited nuclear features of chromosomal instability and increased frequency of nuclear atypia, such as multipolar mitotic spindles and accumulation of cells with large nuclei. FACS-sorted tetraploid cells are viable but have higher rates of spontaneous apoptosis when compared with diploid cells. We have used spectral karyotyping to show that, in contrast to diploid cells, tetraploid cells develop a higher number of both numerical and structural chromosomal abnormalities. Our findings identify a novel function for the Notch signaling pathway in generating tetraploidy and contributing to chromosomal instability. We speculate that abnormal Notch signaling pathway is an initiating genetic mechanism for meningioma and potentially promotes tumor development.


Assuntos
Instabilidade Cromossômica , Meningioma/genética , Receptores Notch/metabolismo , Apoptose , Linhagem Celular Tumoral , Separação Celular , Sobrevivência Celular , Aberrações Cromossômicas , Citometria de Fluxo , Humanos , Cariotipagem , Ploidias , Regiões Promotoras Genéticas , Transdução de Sinais
8.
Brain Pathol ; 18(2): 172-9, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18093250

RESUMO

Meningioma tumor growth involves the subarachnoid space that contains the cerebrospinal fluid. Modeling tumor growth in this microenvironment has been associated with widespread leptomeningeal dissemination, which is uncharacteristic of human meningiomas. Consequently, survival times and tumor properties are varied, limiting their utility in testing experimental therapies. We report the development and characterization of a reproducible orthotopic skull-base meningioma model in athymic mice using the IOMM-Lee cell line. Localized tumor growth was obtained by using optimal cell densities and matrigel as the implantation medium. Survival times were within a narrow range of 17-21 days. The xenografts grew locally compressing surrounding brain tissue. These tumors had histopathologic characteristics of anaplastic meningiomas including high cellularity, nuclear pleomorphism, cellular pattern loss, necrosis and conspicuous mitosis. Similar to human meningiomas, considerable invasion of the dura and skull and some invasion of adjacent brain along perivascular tracts were observed. The pattern of hypoxia was also similar to human malignant meningiomas. We use bioluminescent imaging to non-invasively monitor the growth of the xenografts and determine the survival benefit from temozolomide treatment. Thus, we describe a malignant meningioma model system that will be useful for investigating the biology of meningiomas and for preclinical assessment of therapeutic agents.


Assuntos
Neoplasias Meníngeas/patologia , Meningioma/patologia , Transplante de Neoplasias/métodos , Neoplasias da Base do Crânio/patologia , Animais , Antineoplásicos Alquilantes/uso terapêutico , Linhagem Celular Tumoral/fisiologia , Metilases de Modificação do DNA/metabolismo , Enzimas Reparadoras do DNA/metabolismo , Dacarbazina/análogos & derivados , Dacarbazina/uso terapêutico , Modelos Animais de Doenças , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/fisiologia , Proteínas de Fluorescência Verde/metabolismo , Neoplasias Meníngeas/tratamento farmacológico , Neoplasias Meníngeas/etiologia , Meningioma/tratamento farmacológico , Meningioma/etiologia , Camundongos , Camundongos Nus , Neoplasias da Base do Crânio/tratamento farmacológico , Neoplasias da Base do Crânio/etiologia , Temozolomida , Sais de Tetrazólio , Tiazóis , Fatores de Tempo , Proteínas Supressoras de Tumor/metabolismo
9.
Mol Cancer ; 6: 64, 2007 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-17937814

RESUMO

BACKGROUND: Meningiomas are common brain tumors that are classified into three World Health Organization grades (benign, atypical and malignant) and are molecularly ill-defined tumors. The purpose of this study was identify molecular signatures unique to the different grades of meningiomas and to unravel underlying molecular mechanisms driving meningioma tumorigenesis. RESULTS: We have used a combination of gene expression microarrays and array comparative genomic hybridization (aCGH) to show that meningiomas of all three grades fall into two main molecular groups designated 'low-proliferative' and 'high-proliferative' meningiomas. While all benign meningiomas fall into the low-proliferative group and all malignant meningiomas fall into the high-proliferative group, atypical meningiomas distribute into either one of these groups. High-proliferative atypical meningiomas had an elevated median MIB-1 labeling index and a greater frequency of copy number aberrations (CNAs) compared to low-proliferative atypical meningiomas. Additionally, losses on chromosome 6q, 9p, 13 and 14 were found exclusively in the high-proliferative meningiomas. We have identified genes that distinguish benign low-proliferative meningiomas from malignant high-proliferative meningiomas and have found that gain of cell-proliferation markers and loss of components of the transforming growth factor-beta signaling pathway were the major molecular mechanisms that distinguish these two groups. CONCLUSION: Collectively, our data suggests that atypical meningiomas are not a molecularly distinct group but are similar to either benign or malignant meningiomas. It is anticipated that identified molecular and CNA markers will potentially be more accurate prognostic markers of meningiomas.


Assuntos
Aberrações Cromossômicas , Neoplasias Meníngeas/classificação , Neoplasias Meníngeas/genética , Meningioma/classificação , Meningioma/genética , Hibridização de Ácido Nucleico/métodos , Adulto , Idoso , Idoso de 80 Anos ou mais , Deleção Cromossômica , Feminino , Perfilação da Expressão Gênica/métodos , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , Neoplasias Meníngeas/metabolismo , Meningioma/metabolismo , Pessoa de Meia-Idade
10.
Clin Cancer Res ; 13(1): 68-75, 2007 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-17200340

RESUMO

PURPOSE: Hypoxia in the tumor microenvironment triggers a variety of genetic and adaptive responses that regulate tumor growth. Tumor hypoxia is often associated with more malignant phenotypes, resistance to therapy, and poor survival. The purpose of this study was to evaluate the prevalence of hypoxia in meningiomas using the endogenous hypoxia marker carbonic anhydrase 9 (CA9) and to relate the expression of CA9 to tumor vascularity, histopathologic grade, and clinical variables, such as recurrent tumor status. EXPERIMENTAL DESIGN: Expression of CA9 and CD34, an endothelial cell marker, was examined in serial paraffin-embedded sections by immunohistochemistry in 25 grade 1, 17 grade 2, and 20 grade 3 meningiomas. Areas of immunoreactivity were semiquantitatively scored and correlated to clinical variables using Statistical Analysis System statistical software. RESULTS: Approximately 50% (29 of 62) of all meningiomas contained regions of hypoxia as judged by expression of CA9, and this expression was significantly associated with higher-grade histology (P = 0.001). In contrast, vascularity, as assessed by the percentage of vascular hotspots, was inversely associated with tumor grade (P = 0.023) and was not associated with CA9 expression. Among lower-grade meningiomas, CA9 expression tended to be more common in recurrent tumors. CONCLUSIONS: Tumor hypoxia is an endogenous feature of meningiomas, and therapeutic regimens should include strategies to target the subpopulation of hypoxic as well as the normoxic cells within the tumor. Hypoxia in meningiomas is associated with an aggressive phenotype. Further studies to define the contribution of hypoxia to meningioma pathophysiology are warranted.


Assuntos
Antígenos de Neoplasias/biossíntese , Anidrases Carbônicas/biossíntese , Hipóxia , Meningioma/tratamento farmacológico , Meningioma/patologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Antígenos CD34/biossíntese , Antineoplásicos/farmacologia , Anidrase Carbônica IX , Feminino , Humanos , Imuno-Histoquímica , Masculino , Microcirculação , Pessoa de Meia-Idade , Fenótipo , Fatores de Tempo , Resultado do Tratamento
11.
J Neurooncol ; 78(2): 113-21, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16554968

RESUMO

Even though meningiomas are the second most common brain tumor in adults, little is known about the molecular basis of their growth and development. The lack of suitable cell culture model systems is an impediment to this understanding. Most studies on meningiomas rely on primary, early passage cell lines that eventually senesce or a few established cell lines that have been derived from aggressive variants of meningiomas. We have isolated three primary meningioma cell lines that are negative for telomerase activity. We can overcome the senescence of a Grade III derived meningioma cell line by expressing the telomerase catalytic subunit (hTERT), whereas Grade I meningioma cell lines require the expression of the human papillomavirus E6 and E7 oncogenes in conjunction with hTERT. Meningioma cell lines, immortalized in this manner, maintain their pre-transfection morphology and form colonies in vitro. We have confirmed the meningothelial origin of these cell lines by assessing expression of vimentin and desmoplakin, characteristic markers for meningiomas. Additionally, we have karyotyped these cell lines using array CGH and shown that they represent a spectrum of the genetic diversity seen in primary meningiomas. Thus, these cell lines represent novel cellular reagents for investigating the molecular oncogenesis of meningiomas.


Assuntos
Biomarcadores Tumorais/metabolismo , Linhagem Celular Tumoral/metabolismo , Senescência Celular/genética , Proteínas de Ligação a DNA/genética , Regulação Neoplásica da Expressão Gênica/fisiologia , Neoplasias Meníngeas/genética , Meningioma/genética , Proteínas Oncogênicas Virais/metabolismo , Telomerase/genética , Transformação Genética/genética , Técnicas de Cultura de Células/métodos , Senescência Celular/fisiologia , Proteínas de Ligação a DNA/metabolismo , Desmoplaquinas/metabolismo , Regulação Neoplásica da Expressão Gênica/genética , Técnicas de Transferência de Genes , Humanos , Cariotipagem , Neoplasias Meníngeas/metabolismo , Meningioma/metabolismo , Papillomaviridae , Proteínas E7 de Papillomavirus/metabolismo , Telomerase/metabolismo , Transfecção/métodos , Vimentina/metabolismo
12.
Genome Res ; 14(7): 1413-23, 2004 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15197164

RESUMO

We report the results of a transcript finishing initiative, undertaken for the purpose of identifying and characterizing novel human transcripts, in which RT-PCR was used to bridge gaps between paired EST clusters, mapped against the genomic sequence. Each pair of EST clusters selected for experimental validation was designated a transcript finishing unit (TFU). A total of 489 TFUs were selected for validation, and an overall efficiency of 43.1% was achieved. We generated a total of 59,975 bp of transcribed sequences organized into 432 exons, contributing to the definition of the structure of 211 human transcripts. The structure of several transcripts reported here was confirmed during the course of this project, through the generation of their corresponding full-length cDNA sequences. Nevertheless, for 21% of the validated TFUs, a full-length cDNA sequence is not yet available in public databases, and the structure of 69.2% of these TFUs was not correctly predicted by computer programs. The TF strategy provides a significant contribution to the definition of the complete catalog of human genes and transcripts, because it appears to be particularly useful for identification of low abundance transcripts expressed in a restricted set of tissues as well as for the delineation of gene boundaries and alternatively spliced isoforms.


Assuntos
Software , Transcrição Gênica/genética , Processamento Alternativo/genética , Linhagem Celular , Linhagem Celular Tumoral , Biologia Computacional/métodos , Biologia Computacional/estatística & dados numéricos , Sequência Consenso/genética , DNA de Neoplasias , Bases de Dados Genéticas/classificação , Etiquetas de Sequências Expressas , Genes/genética , Genoma Humano , Células HeLa/patologia , Humanos , Dados de Sequência Molecular , Fases de Leitura Aberta/genética , Design de Software , Validação de Programas de Computador , Células U937/patologia
13.
Arq. bras. endocrinol. metab ; 46(4): 325-329, ago. 2002.
Artigo em Português | LILACS | ID: lil-322172

RESUMO

A contribuiçäo maior da ciência brasileira ao genoma humano foi trazida pelo Projeto Genoma Humano do Câncer (Human Genome Cancer Project- HCGP) uma parceria da FAPESP e do Ludwig Institute for Cancer Research e desenvolvido por 29 diferentes laboratórios de seqüenciamento e um centro de bioinformática. Foram seqüenciados mais de 1 milhäo de fragmentos génicos expressos (expressed sequences tags, ESTs), provenientes de diferentes tumores humanos. Grande parte destes dados é de acesso público através da website do Gene Bank (www.ncbi.nim.nig.gov), mantido pelo NCBI - National Center for Biotechnology Information. Atualmente, diversos projetos estäo em desenvolvimento utilizando informações geradas no HCGP e abrangem observar a expressäo diferenciada dos genes em diferentes tumores, caracterizaçäo completa de genes específicos, assim como o estudo funcional e estrutural dos produtos protéicos. É promissora a perspectiva de que num futuro próximo, diferentes resultados provenientes destas investigações possam trazer benefícios preventivos, prognósticos e clínicos em câncer e outras doenças.


Assuntos
Genoma Humano , Projeto Genoma Humano , Neoplasias , Brasil , Doenças das Plantas/parasitologia , Expressão Gênica , Genoma , Polimorfismo Genético/fisiologia , Apoio à Pesquisa como Assunto
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA