Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Biochem J ; 420(3): 391-402, 2009 May 27.
Artigo em Inglês | MEDLINE | ID: mdl-19260824

RESUMO

L1-CAM (L1 cell-adhesion molecule), or more simply L1, plays an important role in the progression of human carcinoma. Overexpression promotes tumour-cell invasion and motility, growth in nude mice and tumour metastasis. It is feasible that L1-dependent signalling contributes to these effects. However, little is known about its mechanism in tumour cells. We reported previously that L1 is cleaved by ADAM (a disintegrin and metalloprotease) and that the cytoplasmic part is essential for L1 function. Here we analysed more closely the role of proteolytic cleavage in L1-mediated nuclear signalling. Using OVMz carcinoma cells and L1-transfected cells as a model, we found that ADAM10-mediated cleavage of L1 proceeds in lipid raft and non-raft domains. The cleavage product, L1-32, is further processed by PS (presenilin)/gamma-secretase to release L1-ICD, an L1 intracellular domain of 28 kDa. Overexpression of dominant-negative PS1 or use of a specific gamma-secretase inhibitor leads to an accumulation of L1-32. Fluorescence and biochemical analysis revealed a nuclear localization for L1-ICD. Moreover, inhibition of ADAM10 and/or gamma-secretase blocks nuclear translocation of L1-ICD and L1-dependent gene regulation. Overexpression of recombinant L1-ICD mediates gene regulation in a similar manner to full-length L1. Our results establish for the first time that regulated proteolytic processing by ADAM10 and PS/gamma-secretase is essential for the nuclear signalling of L1 in human carcinoma cell lines.


Assuntos
Proteínas ADAM/metabolismo , Secretases da Proteína Precursora do Amiloide/metabolismo , Núcleo Celular/metabolismo , Proteínas de Membrana/metabolismo , Molécula L1 de Adesão de Célula Nervosa/metabolismo , Presenilinas/metabolismo , Transdução de Sinais , Proteínas ADAM/genética , Proteína ADAM10 , Transporte Ativo do Núcleo Celular , Secretases da Proteína Precursora do Amiloide/genética , Animais , Sítios de Ligação/genética , Células CHO , Linhagem Celular , Linhagem Celular Tumoral , Cricetinae , Cricetulus , Feminino , Citometria de Fluxo , Humanos , Proteínas de Membrana Lisossomal/metabolismo , Microdomínios da Membrana/metabolismo , Proteínas de Membrana/genética , Microscopia de Fluorescência , Molécula L1 de Adesão de Célula Nervosa/genética , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Presenilinas/genética , RNA Interferente Pequeno/genética , Receptores do Ácido Retinoico/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transcrição Gênica , Transfecção
2.
Exp Cell Res ; 314(13): 2411-8, 2008 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-18555990

RESUMO

L1 cell adhesion molecule (L1-CAM) is a transmembrane cell adhesion molecule initially defined as a promigratory molecule in the developing nervous system. L1 is also overexpressed in a variety of human carcinomas and is associated with bad prognosis. In carcinoma cell lines L1 augments cell motility and metastasis, tumor growth in nude mice and induces expression of L1-dependent genes. It is not known whether L1-signaling requires ligand binding. The RGD motif in the sixth Ig domain of L1 is a binding site for integrins. In the present study we analyzed the role of RGDs in L1-signaling using site-directed mutagenesis combined with antibody blocking studies. We observed that L1-RGE expressing HEK293 cells showed reduced cell-cell binding, cell motility, invasiveness and tumor growth in NOD/SCID mice. The RGE-mutation impaired L1-dependent gene regulation and antibodies to alphavbeta5 integrin had similar effects. Mutant L1 was unable to translocate to the nucleus. Our findings highlight the importance of the RGD site in L1 for human tumors and suggest that nuclear signaling of L1 is dependent on integrins.


Assuntos
Núcleo Celular/metabolismo , Integrinas/metabolismo , Molécula L1 de Adesão de Célula Nervosa/química , Oligopeptídeos/fisiologia , Transporte Ativo do Núcleo Celular/genética , Animais , Sítios de Ligação/genética , Sítios de Ligação/fisiologia , Células CHO , Adesão Celular/genética , Comunicação Celular/genética , Movimento Celular/genética , Células Cultivadas , Cricetinae , Cricetulus , Feminino , Deleção de Genes , Regulação da Expressão Gênica , Humanos , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Molécula L1 de Adesão de Célula Nervosa/genética , Molécula L1 de Adesão de Célula Nervosa/metabolismo , Molécula L1 de Adesão de Célula Nervosa/fisiologia , Oligopeptídeos/genética , Transdução de Sinais , Transplante Heterólogo
3.
J Cell Biochem ; 103(6): 1783-97, 2008 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-17910038

RESUMO

Exosomes are small membrane vesicles derived from intracellular multivescicular bodies (MVBs) that can undergo constitutive and regulated secretion from cells. Exosomes can also secrete soluble proteins through metalloprotease-dependent ectodomain shedding. In this study, we sought to determine whether ErbB1 receptors are present within exosomes isolated from the human keratinocyte cell line, HaCaT, and whether exosome-associated ErbB1 receptors can undergo further proteolytic processing. We show that full-length transmembrane ErbB1 is secreted in HaCaT exosomes. EGF treatment and calcium flux stimulated the release of phosphorylated ErbB1 in exosomes but only ligand-stimulated release was blocked by the ErbB1 kinase inhibitor, AG1478, indicating that ligand-dependent ErbB1 receptor activation can initiate ErbB1 secretion into exosomes. In addition, other immunoreactive but truncated ErbB1 isoforms were detected in exosomes suggestive of additional proteolytic processing. We demonstrate that cellular and exosomal ErbB1 receptors can undergo ectodomain shedding to generate soluble N-terminal ectodomains and membrane-associated C-terminal remnant fragments (CTFs). ErbB1 shedding was activated by calcium flux and the metalloprotease activator APMA (4-aminophenylmercuric acetate) and was blocked by a metalloprotease inhibitor (GM6001). Soluble ErbB1 ectodomains shed into conditioned medium retained the ability to bind exogenous ligand. Our results provide new insights into the proteolysis, trafficking and fate of ErbB1 receptors and suggest that the novel ErbB1 isoforms may have functions distinct from the plasma membrane receptor.


Assuntos
Membrana Celular/metabolismo , Receptores ErbB/biossíntese , Metaloproteases/metabolismo , Transporte Biológico , Linhagem Celular , Dipeptídeos/farmacologia , Receptores ErbB/antagonistas & inibidores , Receptores ErbB/isolamento & purificação , Exocitose , Humanos , Membranas Intracelulares/metabolismo , Metaloproteases/antagonistas & inibidores , Acetato de Fenilmercúrio/análogos & derivados , Acetato de Fenilmercúrio/farmacologia , Isoformas de Proteínas/biossíntese , Isoformas de Proteínas/isolamento & purificação , Estrutura Terciária de Proteína , Quinazolinas , Transdução de Sinais , Tirfostinas/farmacologia
4.
Biochem J ; 393(Pt 3): 609-18, 2006 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-16229685

RESUMO

Ectodomain shedding is a proteolytic mechanism by which transmembrane molecules are converted into a soluble form. Cleavage is mediated by metalloproteases and proceeds in a constitutive or inducible fashion. Although believed to be a cell-surface event, there is increasing evidence that cleavage can take place in intracellular compartments. However, it is unknown how cleaved soluble molecules get access to the extracellular space. By analysing L1 (CD171) and CD44 in ovarian carcinoma cells, we show in the present paper that the cleavage induced by ionomycin, APMA (4-aminophenylmercuric acetate) or MCD (methyl-beta-cyclodextrin) is initiated in an endosomal compartment that is subsequently released in the form of exosomes. Calcium influx augmented the release of exosomes containing functionally active forms of ADAM10 (a disintegrin and metalloprotease 10) and ADAM17 [TACE (tumour necrosis factor a-converting enzyme)] as well as CD44 and L1 cytoplasmic cleavage fragments. Cleavage could also proceed in released exosomes, but only depletion of ADAM10 by small interfering RNA blocked cleavage under constitutive and induced conditions. In contrast, cleavage of L1 in response to PMA occurred at the cell surface and was mediated by ADAM17. We conclude that different ADAMs are involved in distinct cellular compartments and that ADAM10 is responsible for shedding in vesicles. Our findings open up the possibility that exosomes serve as a platform for ectodomain shedding and as a vehicle for the cellular export of soluble molecules.


Assuntos
Vesículas Citoplasmáticas/metabolismo , Exocitose , Receptores de Hialuronatos/metabolismo , Molécula L1 de Adesão de Célula Nervosa/metabolismo , Proteínas ADAM/genética , Proteínas ADAM/metabolismo , Proteína ADAM10 , Proteína ADAM17 , Secretases da Proteína Precursora do Amiloide , Linhagem Celular Tumoral , Regulação da Expressão Gênica , Humanos , Receptores de Hialuronatos/química , Receptores de Hialuronatos/genética , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Molécula L1 de Adesão de Célula Nervosa/química , Molécula L1 de Adesão de Célula Nervosa/genética , Estrutura Terciária de Proteína , Interferência de RNA
5.
Clin Cancer Res ; 11(7): 2492-501, 2005 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-15814625

RESUMO

PURPOSE: The L1 adhesion molecule (CD171) is overexpressed in human ovarian and endometrial carcinomas and is associated with bad prognosis. Although expressed as a transmembrane molecule, L1 is released from carcinoma cells in a soluble form. Soluble L1 is present in serum and ascites of ovarian carcinoma patients. We investigated the mode of L1 cleavage and the function of soluble L1. EXPERIMENTAL DESIGN: We used ovarian carcinoma cell lines and ascites from ovarian carcinoma patients to analyze soluble L1 and L1 cleavage by Western blot analysis and ELISA. RESULTS: We find that in ovarian carcinoma cells the constitutive cleavage of L1 proceeds in secretory vesicles. We show that apoptotic stimuli like C2-ceramide, staurosporine, UV irradiation, and hypoxic conditions enhance L1-vesicle release resulting in elevated levels of soluble L1. Constitutive cleavage of L1 is mediated by a disintegrin and metalloproteinase 10, but under apoptotic conditions multiple metalloproteinases are involved. L1 cleavage occurs in two types of vesicles with distinct density features: constitutively released vesicles with similarity to exosomes and apoptotic vesicles. Both types of L1-containing vesicles are present in the ascites fluids of ovarian carcinoma patients. Soluble L1 from ascites is a potent inducer of cell migration and can trigger extracellular signal-regulated kinase phosphorylation. CONCLUSIONS: We suggest that tumor-derived vesicles may be an important source for soluble L1 that could regulate tumor cell function in an autocrine/paracrine fashion.


Assuntos
Apoptose , Vesículas Citoplasmáticas/metabolismo , Molécula L1 de Adesão de Célula Nervosa/metabolismo , Neoplasias Ovarianas/metabolismo , Esfingosina/análogos & derivados , Proteínas ADAM , Proteína ADAM17 , Secretases da Proteína Precursora do Amiloide , Animais , Líquido Ascítico/química , Ácido Aspártico Endopeptidases/metabolismo , Ligação Competitiva , Células CHO , Hipóxia Celular , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Cricetinae , Cricetulus , Vesículas Citoplasmáticas/efeitos dos fármacos , Endopeptidases , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Feminino , Células HeLa , Humanos , Metaloendopeptidases/metabolismo , Molécula L1 de Adesão de Célula Nervosa/farmacologia , Neoplasias Ovarianas/patologia , Fosforilação/efeitos dos fármacos , Solubilidade , Esfingosina/farmacologia
6.
Lancet ; 362(9387): 869-75, 2003 Sep 13.
Artigo em Inglês | MEDLINE | ID: mdl-13678974

RESUMO

BACKGROUND: Ovarian and uterine carcinomas are the most common cause of cancer-related deaths in gynecological malignant diseases. We aimed to assess whether the L1 adhesion molecule, an important mediator for cell migration for neural and tumour cells, is expressed in these carcinomas. METHODS: We investigated L1 expression by immunohistochemistry, RT-PCR, and Western blot analysis of tumour samples. Soluble L1 in the serum was detected by ELISA and immunoprecipitation. FINDINGS: We detected the L1 adhesion molecule in ovarian and uterine tumours in a stage-dependent manner. In a retrospective study L1 was found in 46 of 58 ovarian carcinomas and 20 of 72 uterine adenocarcinomas. L1 expression was an excellent predictor of poor outlook (p<0.00001). Patients with L1 positive uterine tumours were at high risk for progression even in the endometrioid-type tumours, which usually have a favourable prognosis. In uterine tumours, expression of L1 in curettage samples enabled us to identify aggressive tumours before the operation. Soluble L1 was specifically detected in serum samples from patients with ovarian and uterine tumours. ADAM10, which was implicated in previous studies as L1 sheddase, was expressed in tumours in which soluble L1 was present in the serum. INTERPRETATION: L1 is overexpressed in ovarian and uterine carcinomas and is associated with short survival. L1 can serve as a new marker for prediction of clinical outcome and could be helpful to identify patients with uterine tumours who are at high risk for recurrent disease. L1 expression and cleavage could promote dissemination of tumours by facilitating cell migration.


Assuntos
Complexo Antígeno L1 Leucocitário/análise , Neoplasias Ovarianas/metabolismo , Neoplasias Uterinas/metabolismo , Antígenos de Neoplasias/análise , Antígenos de Neoplasias/genética , Biomarcadores Tumorais/genética , Western Blotting , Movimento Celular/genética , Progressão da Doença , Feminino , Humanos , Imuno-Histoquímica , Complexo Antígeno L1 Leucocitário/genética , Pessoa de Meia-Idade , Neoplasias Ovarianas/diagnóstico , Neoplasias Ovarianas/genética , Prognóstico , Estudos Retrospectivos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Análise de Sobrevida , Taxa de Sobrevida , Neoplasias Uterinas/diagnóstico , Neoplasias Uterinas/genética
7.
FASEB J ; 17(2): 292-4, 2003 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-12475894

RESUMO

Cells can release membrane components in a soluble form and as membrane vesicles. L1, an important molecule for cell migration of neural and tumor cells, is released by membrane-proximal cleavage, and soluble L1 promotes cell migration. Release of L1 is enhanced by shedding inducers such as phorbol ester and pervanadate, but it is also enhanced by depletion of cellular cholesterol with methyl-beta-cyclodextrin (MCD). How such different compounds can induce shedding is presently unknown. We show here that ADAM10 is involved in L1 cleavage, which occurs at the cell surface and in the Golgi apparatus. MCD and pervanadate treatment induced the release of microvesicles containing full-length L1 and the active form of ADAM10. L1 cleavage occurred in isolated vesicles. L1-containing microvesicles could trigger haptotactic cell migration. Only the neural L1 form carrying the RSLE signal for clathrin-dependent endocytosis was recruited and cleaved in vesicles. Phorbol ester treatment activated L1 cleavage predominantly at the cell surface. Our results provide evidence for two pathways of L1 cleavage, based on ADAM10 localization, that can be activated differentially: 1) direct cleavage at the cell surface, and 2) release and cleavage in secretory vesicles most likely derived from the Golgi apparatus. The findings establish a novel role for ADAM10 as a vesicle-based protease.


Assuntos
Membrana Celular/metabolismo , Vesículas Citoplasmáticas/metabolismo , Endopeptidases/metabolismo , Molécula L1 de Adesão de Célula Nervosa/metabolismo , Motivos de Aminoácidos/genética , Sequência de Aminoácidos , Secretases da Proteína Precursora do Amiloide , Animais , Ácido Aspártico Endopeptidases , Células CHO , Linhagem Celular , Movimento Celular/efeitos dos fármacos , Cricetinae , Endopeptidases/genética , Éxons/genética , Complexo de Golgi/metabolismo , Humanos , Molécula L1 de Adesão de Célula Nervosa/genética , Molécula L1 de Adesão de Célula Nervosa/farmacologia , Transfecção
8.
Cell Adh Migr ; 6(4): 374-84, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22796939

RESUMO

The L1 cell adhesion molecule (L1CAM) plays a major role in the development of the nervous system and in the malignancy of human tumors. In terms of biological function, L1CAM comes along in two different flavors: (1) a static function as a cell adhesion molecule that acts as a glue between cells; (2) a motility promoting function that drives cell migration during neural development and supports metastasis of human cancers. Important factors that contribute to the switch in the functional mode of L1CAM are: (1) the cleavage from the cell surface by membrane proximal proteolysis and (2) the ability to change binding partners and engage in L1CAM-integrin binding. Recent studies have shown that the cleavage of L1CAM by metalloproteinases and the binding of L1CAM to integrins via its RGD-motif in the sixth Ig-domain activate signaling pathways distinct from the ones elicited by homophilic binding. Here we highlight important features of L1CAM proteolysis and the signaling of L1CAM via integrin engagement. The novel insights into L1CAM downstream signaling and its regulation during tumor progression and epithelial-mesenchymal transition (EMT) will lead to a better understanding of the dualistic role of L1CAM as a cell adhesion and/or motility promoting cell surface molecule.


Assuntos
Movimento Celular , Neoplasias/metabolismo , Neoplasias/patologia , Molécula L1 de Adesão de Célula Nervosa/fisiologia , Sequência de Aminoácidos , Animais , Comunicação Celular , Transição Epitelial-Mesenquimal , Humanos , Modelos Moleculares , Dados de Sequência Molecular , Invasividade Neoplásica , Molécula L1 de Adesão de Célula Nervosa/química , Molécula L1 de Adesão de Célula Nervosa/metabolismo , Estrutura Terciária de Proteína , Transdução de Sinais
9.
Cancer Res ; 70(11): 4613-23, 2010 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-20460525

RESUMO

We describe for the first time a new integral molecular pathway, linking transcription factor E2F3a to epidermal growth factor receptor (EGFR) activation in ovarian cancer cells. Investigations on the role of E2F family members in EGFR-mediated mitogenic signaling revealed that E2F3a was selectively upregulated following EGFR activation, whereas all other E2F family members remained unaffected. In contrast, EGF treatment of healthy ovarian surface epithelial and mesothelial cells yielded a selective upregulation of proliferation-promoting E2F1 and E2F2 without influencing E2F3a expression. In ovarian cancer cell lines, the extent of EGF-induced proliferative stimulus was closely related to the magnitude of E2F3a increase, and proliferation inhibition by E2F3a knockdown was not overcome by EGF exposure. Furthermore, the EGFR-E2F3a axis was found to be signal transducer and activator of transcription 1/3 dependent and the ratio of IFN-regulatory factor (IRF)-1 to IRF-2 was shown to be determinative for E2F3a control. In a pilot study on 32 primary ovarian cancer specimens, a highly significant correlation between activated EGFR and E2F3a expression was disclosed. This new integral pathway in the EGFR-driven mitogenic cell response, which through its key player E2F3a was found to be essential in triggering proliferation in ovarian cancer cells, provides new insights into EGFR signaling and could represent the basis for appealing new therapeutic approaches in ovarian cancer.


Assuntos
Fator de Transcrição E2F3/metabolismo , Receptores ErbB/metabolismo , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Processos de Crescimento Celular/fisiologia , Linhagem Celular Tumoral , Fator de Transcrição E2F3/biossíntese , Fator de Transcrição E2F3/genética , Fator de Crescimento Epidérmico/farmacologia , Receptores ErbB/antagonistas & inibidores , Feminino , Humanos , Fator Regulador 1 de Interferon/metabolismo , Fator Regulador 2 de Interferon/metabolismo , Neoplasias Ovarianas/genética , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , RNA Interferente Pequeno/administração & dosagem , RNA Interferente Pequeno/genética , Transdução de Sinais , Transfecção , Regulação para Cima/efeitos dos fármacos
10.
Int J Cancer ; 119(3): 549-55, 2006 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-16506207

RESUMO

The adhesion molecule L1 is expressed in primary melanomas and cutaneous metastases in contrast to melanocytic nevi and melanocytes, and is significantly associated with metastatic spread. Recent studies have demonstrated that in carcinomas L1 expression is associated with sustained activation of the extracellular signal-regulated kinase (ERK) pathway and upregulation of ERK-dependent, motility- and invasion-associated gene products including alphavbeta3 integrin. The objective of this study was to further investigate the role of the adhesion molecule L1 in melanoma progression, and to evaluate whether targeting the L1 adhesion molecule would have therapeutic effects against invasive melanoma growth. Using human melanoma cells from different stages of progression in monolayer and organotypic human skin culture mimicking the pathophysiological environment of cutaneous melanoma, we found that (1) L1 expression mostly correlates with melanoma progression and alphavbeta3 integrin expression, (2) overexpression of L1 in early radial growth phase melanoma cells promotes conversion from radial to vertical growth phase melanoma without upregulation of alphavbeta3 integrin expression, and (3) suppression of L1 function significantly reduces migration and invasion of melanoma cells, but does not completely block invasive melanoma growth. Altogether, L1 plays a critical role in melanoma invasion and progression and offers therapeutic potential in combination with conventional anticancer agents.


Assuntos
Melanoma/patologia , Molécula L1 de Adesão de Célula Nervosa/metabolismo , Molécula L1 de Adesão de Célula Nervosa/fisiologia , Animais , Anticorpos/farmacologia , Western Blotting , Células CHO , Divisão Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Cricetinae , Cricetulus , Progressão da Doença , Humanos , Imuno-Histoquímica , Integrina alfaVbeta3/análise , Masculino , Melanócitos/metabolismo , Melanócitos/patologia , Melanoma/metabolismo , Invasividade Neoplásica , Metástase Neoplásica , Molécula L1 de Adesão de Célula Nervosa/análise , Molécula L1 de Adesão de Célula Nervosa/imunologia , Pele/metabolismo , Pele/patologia , Técnicas de Cultura de Tecidos/métodos
11.
Int J Cancer ; 115(4): 658-65, 2005 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-15704102

RESUMO

L1 is a neural cell adhesion molecule involved in cell migration, axon growth and guidance. Recent data have shown that L1 is overexpressed in ovarian and endometrial tumors and is associated with bad prognosis. How L1 promotes tumor progression is presently unknown. Here we show that L1 expression is predominantly confined to the invasive front of ovarian carcinomas. Overexpression of L1 in carcinoma cell lines by adenovirus-mediated gene transfer enhanced the haptotactic cell migration on extracellular matrix proteins. Expression of L1 augmented tumor growth of carcinomas xenografted in nonobese diabetic/severe combined immunodeficient mice (NOD/SCID). A recent report has demonstrated L1-dependent upregulation of beta3 integrin involving activation of the extracellular signal-regulated kinase (erk) pathway. We find that L1 and beta3 integrin are not coexpressed in ovarian carcinoma tissues. Overexpression of L1 did not upregulate beta3 integrin in ovarian carcinoma cell lines but could do so in HEK293 cells. Our results suggest that L1 could drive progression by enhancing cell migration and tumor growth but that L1 dependent and erk-regulated gene expression requires cell-type specific elements.


Assuntos
Integrina beta3/genética , Molécula L1 de Adesão de Célula Nervosa/genética , Neoplasias Ovarianas/genética , Animais , Divisão Celular , Linhagem Celular , Linhagem Celular Tumoral , Movimento Celular , Feminino , Humanos , Rim , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Molécula L1 de Adesão de Célula Nervosa/metabolismo , Neoplasias Ovarianas/patologia , RNA Interferente Pequeno/genética , Transfecção , Transplante Heterólogo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA