Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Oncol Rep ; 38(1): 545-550, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28560387

RESUMO

Neuroblastoma is a childhood cancer originating from embryonic neural crest cells. Amplification of the proto­oncogene N-myc, seen in ~30% of neuroblastoma tumors, is a marker for poor prognosis. Recently discovered small regulatory RNAs, microRNAs (miRNAs), are implicated in cancers, including neuroblastoma. miRNAs downregulate the expression of genes by binding to the 3'-untranslated regions (3'-UTRs), thereby inhibiting translation or inducing degradation of cognate mRNAs. Our study sought to identify miRNAs that regulate N-myc expression and thereby malignancy in neuroblastoma. miRNAs whose expression negatively correlates with N-myc expression were identified from a miRNA microarray of 4 N-myc-amplified neuroblastoma cell lines. Three of these miRNAs (miR-17, miR-20a and miR-18a) belong to the miR-17-92 cluster, previously shown to be upregulated by N-myc. qPCR validation of these miRNAs in a larger panel of cell lines revealed that levels of miR-17 were inversely proportional to N-myc mRNA amounts in the N-myc-amplified cell lines. Notably, miR-17 also downregulated N-myc protein synthesis in the N-myc-amplified cells, thereby generating a negative feedback regulatory loop between the proto-oncogene and this miRNA. Moreover, the neuronal-specific RNA-binding protein HuD (ELAVL4), which regulates the processing/stability of N-myc mRNA, competes with miR-17 for a binding site in the 3'-UTR of N-myc. Thus, N-myc levels appear to be modulated by the antagonistic interactions of both miR-17, as a negative regulator, and HuD, as a positive regulator, providing further evidence of the complex cellular control mechanisms of this oncogene in N-myc-amplified neuroblastoma cells.


Assuntos
Proteína Semelhante a ELAV 4/metabolismo , Regulação Neoplásica da Expressão Gênica , MicroRNAs/metabolismo , Proteína Proto-Oncogênica N-Myc/genética , Neuroblastoma/genética , Regiões 3' não Traduzidas/genética , Diferenciação Celular/genética , Linhagem Celular Tumoral , Regulação para Baixo , Proteína Semelhante a ELAV 4/genética , Retroalimentação Fisiológica , Humanos , MicroRNAs/genética , Análise em Microsséries , Proteína Proto-Oncogênica N-Myc/metabolismo , Células-Tronco Neurais , Neurônios/metabolismo , Proto-Oncogene Mas , Proto-Oncogenes/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
2.
Stem Cell Res ; 15(2): 419-26, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26342562

RESUMO

Neuroblastoma, a malignancy of multipotent embryonic neural crest cells, is the most common extracranial solid cancer in childhood and most common cancer in infancy. Cellular phenotype has been shown to be an important determinant of the malignant potential in human neuroblastoma cells and tumors. Whereas neuroblastic (N-type) are moderately malignant and nonneuronal (S-type) cells are nonmalignant, I-type stem cells are highly tumorigenic, irrespective of N-myc amplification status. In the present study, we sought to determine which genes were overexpressed in the I-type cells which might characterize and maintain the stem cell state and/or malignancy of human neuroblastoma cancer stem cells. We used a microarray platform to compare the steady-state expression levels of mRNAs from 13 human neuroblastoma cell lines representing the three cellular phenotypes. Using qRT-PCR and Western blot analyses, we identified seven genes whose expression is consistently elevated exclusively in neuroblastoma cancer stem cells: CD133, KIT, NOTCH1, GPRC5C, PIGF2, TRKB, and LNGFR. Moreover, we show that the genes are phenotype specific, as differentiation of I-type BE(2)-C cells to either an N- or S-type morphology results in significantly reduced mRNA expression. Finally, we show that NOTCH1 plays an important role in maintaining the stem cell phenotype. The identification and characterization of these genes, elevated in highly malignant neuroblastoma stem cells, could provide the basis for developing novel therapies for treatment of this lethal childhood cancer.


Assuntos
Regulação Neoplásica da Expressão Gênica , Células-Tronco Neoplásicas/metabolismo , Antígeno AC133 , Antígenos CD/metabolismo , Western Blotting , Linhagem Celular Tumoral , Glicoproteínas/metabolismo , Humanos , Células-Tronco Neoplásicas/citologia , Neuroblastoma/metabolismo , Neuroblastoma/patologia , Peptídeos/metabolismo , Fenótipo , Fator de Crescimento Placentário , Proteínas da Gravidez/metabolismo , Proteínas Proto-Oncogênicas c-kit/metabolismo , Interferência de RNA , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Receptor Notch1/antagonistas & inibidores , Receptor Notch1/genética , Receptor Notch1/metabolismo , Receptores Acoplados a Proteínas G/metabolismo
3.
BMC Cancer ; 14: 309, 2014 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-24885481

RESUMO

BACKGROUND: Neuroblastoma (NB) is the most common extracranial solid tumor in children. NB tumors and derived cell lines are phenotypically heterogeneous. Cell lines are classified by phenotype, each having distinct differentiation and tumorigenic properties. The neuroblastic phenotype is tumorigenic, has neuronal features and includes stem cells (I-cells) and neuronal cells (N-cells). The non-neuronal phenotype (S-cell) comprises cells that are non-tumorigenic with features of glial/smooth muscle precursor cells. This study identified miRNAs associated with each distinct cell phenotypes and investigated their role in regulating associated differentiation and tumorigenic properties. METHODS: A miRNA microarray was performed on the three cell phenotypes and expression verified by qRT-PCR. miRNAs specific for certain cell phenotypes were modulated using miRNA inhibitors or stable transfection. Neuronal differentiation was induced by RA; non-neuronal differentiation by BrdU. Changes in tumorigenicity were assayed by soft agar colony forming ability. N-myc binding to miR-375 promoter was assayed by chromatin-immunoprecipitation. RESULTS: Unsupervised hierarchical clustering of miRNA microarray data segregated neuroblastic and non-neuronal cell lines and showed that specific miRNAs define each phenotype. qRT-PCR validation confirmed that increased levels of miR-21, miR-221 and miR-335 are associated with the non-neuronal phenotype, whereas increased levels of miR-124 and miR-375 are exclusive to neuroblastic cells. Downregulation of miR-335 in non-neuronal cells modulates expression levels of HAND1 and JAG1, known modulators of neuronal differentiation. Overexpression of miR-124 in stem cells induces terminal neuronal differentiation with reduced malignancy. Expression of miR-375 is exclusive for N-myc-expressing neuroblastic cells and is regulated by N-myc. Moreover, miR-375 downregulates expression of the neuronal-specific RNA binding protein HuD. CONCLUSIONS: Thus, miRNAs define distinct NB cell phenotypes. Increased levels of miR-21, miR-221 and miR-335 characterize the non-neuronal, non-malignant phenotype and miR-335 maintains the non-neuronal features possibly by blocking neuronal differentiation. miR-124 induces terminal neuronal differentiation with reduction in malignancy. Data suggest N-myc inhibits neuronal differentiation of neuroblastic cells possibly by upregulating miR-375 which, in turn, suppresses HuD. As tumor differentiation state is highly predictive of patient survival, the involvement of these miRNAs with NB differentiation and tumorigenic state could be exploited in the development of novel therapeutic strategies for this enigmatic childhood cancer.


Assuntos
Neoplasias Encefálicas/genética , Carcinogênese/genética , MicroRNAs/biossíntese , Neuroblastoma/genética , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Diferenciação Celular/genética , Linhagem Celular Tumoral , Criança , Proteínas ELAV/antagonistas & inibidores , Proteínas ELAV/genética , Proteína Semelhante a ELAV 4 , Regulação Neoplásica da Expressão Gênica , Humanos , Análise em Microsséries , Células-Tronco Neurais/metabolismo , Células-Tronco Neurais/patologia , Neuroblastoma/metabolismo , Neuroblastoma/patologia , Fenótipo , Proteínas de Ligação a RNA/genética
4.
Neurogastroenterol Motil ; 23(10): 912-e396, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21806741

RESUMO

BACKGROUND: Our primary goals were to investigate the effects of two-channel gastric pacing on gastric myoelectrical activity, and energy consumption with the secondary intent to monitor gastric emptying and symptoms in patients with severe diabetic gastroparesis. METHODS: Four pairs of temporary pacing wires were inserted on the serosa of the stomach at the time of laparotomy to place the Enterra™ System in 19 patients with severe gastroparesis not responding to standard medical therapies. Two of the pairs were for electrical stimulation and the other two for recording. Five days after surgery the optimal pacing parameters for the entrainment of gastric slow waves in each patient were identified by serosal recordings. Two-channel gastric pacing was then initiated for 6 weeks using a newly developed external multi-channel pulse generator. Electrogastrogram (EGG), Total Symptom Score (TSS), and a 4-h gastric emptying test were assessed at baseline and after 6 weeks of active gastric pacing. Enterra™ device was turned OFF during the duration of this study. KEY RESULTS: Two-channel gastric pacing at 1.1 times the intrinsic frequency entrained gastric slow waves and normalized gastric dysrhythmia. After 6 weeks of gastric pacing, tachygastria was decreased from 15 ± 3 to 5 ± 1% in the fasting state and from 10 ± 2 to 5 ± 1% postprandially (P < 0.05), mean TSS was reduced from 21.3 ± 1.1 to 7.0 ± 1.5 (P < 0.05) and mean 4-h gastric retention improved from 42 to 28% (P = 0.05). CONCLUSIONS & INFERENCES: Two-channel gastric pacing is a novel treatment approach which is able to normalize and enhance gastric slow wave activity as well as accelerate gastric emptying in patients with diabetic gastroparesis with a goal safety profile.


Assuntos
Complicações do Diabetes/terapia , Terapia por Estimulação Elétrica/métodos , Gastroparesia/terapia , Adulto , Terapia por Estimulação Elétrica/instrumentação , Eletrodos Implantados , Feminino , Gastroparesia/etiologia , Humanos , Masculino , Pessoa de Meia-Idade
5.
Genes Cancer ; 2(11): 1034-43, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22737269

RESUMO

Cellular heterogeneity is a well-known feature of human neuroblastoma tumors and cell lines. Of the 3 phenotypes (N-, I-, and S-type) isolated and characterized, the I-type cancer stem cell of neuroblastoma is the most malignant. Here, we report that, although wild-type N-Ras protein is expressed at the same level in all 3 neuroblastoma cell phenotypes, activated N-Ras-GTP level is significantly higher in I-type cancer stem cells. When activated N-Ras levels were decreased by transfection of a dominant-negative N-Ras construct, the malignant potential of I-type cancer stem cells decreased significantly. Conversely, when weakly malignant N-type cells were transfected with a constitutively active N-Ras construct, activated N-Ras levels, and malignant potential, were significantly increased. Thus, high levels of N-Ras-GTP are required for the increased malignancy of I-type neuroblastoma cancer stem cells. Moreover, increased activation of N-Ras results from significant down-regulation of neurofibromin (NF1), an important RasGAP. This specific down-regulation is mediated by an ubiquitin-proteasome-dependent pathway. Thus, decreased expression of NF1 in I-type neuroblastoma cancer stem cells causes a high level of activated N-Ras that is, at least in part, responsible for their higher tumorigenic potential.

6.
Am J Surg ; 195(1): 122-9, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18082553

RESUMO

BACKGROUND: The aim of the current study was to investigate the efficacy of 2-channel gastric electrical stimulation (GES) with a custom-made implantable pacemaker on delayed gastric emptying and gastric dysrhythmia induced by glucagon in dogs. METHODS: Six dogs were studied in 4 randomized session (saline, glucagon, glucagon with single-channel or 2-channel GES). GES was applied via the first pair of electrodes for single-channel GES or the first and third pairs of electrodes for 2-channel GES. Gastric emptying was assessed for 90 minutes and gastric slow waves were recorded at the same time. RESULTS: Both single-channel and 2-channel GES improved gastric dysrhythmia (P < .05 vs glucagon session). Two-channel GES but not single-channel GES improved glucagon-induced delayed gastric emptying at 30 minutes, 45 minutes, 60 minutes, 75 minutes, and 90 minutes. CONCLUSION: Two-channel GES with a novel implantable pacemaker is more efficient and effective than single-channel GES in improving delayed gastric emptying induced by glucagon. This implantable multipoint pacemaker may provide a new option for treatment of gastric motility disorders.


Assuntos
Terapia por Estimulação Elétrica , Esvaziamento Gástrico , Gastroparesia/terapia , Animais , Glicemia/análise , Cães , Estimulação Elétrica , Eletrodos Implantados , Feminino , Gastroparesia/induzido quimicamente , Glucagon/efeitos adversos , Resultado do Tratamento
7.
Semin Cancer Biol ; 17(3): 241-7, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16839774

RESUMO

Human neuroblastoma is an embryonic cancer of the neural crest. Cellular heterogeneity is a characteristic feature of both tumors and derived cell lines. Recent studies have revealed that both cell lines and tumors contain cancer stem cells. In culture, these cells are self-renewing, multipotent, and highly malignant; in tumors their frequency correlates with a worse prognosis. Their identification and characterization should now permit a targeted approach to more effective treatment of this often fatal childhood cancer.


Assuntos
Células-Tronco Neoplásicas/patologia , Neuroblastoma/patologia , Humanos , Crista Neural/patologia , Neuroblastoma/genética
8.
J Mol Neurosci ; 26(1): 39-55, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-15968085

RESUMO

Transcription of the gene encoding the epinephrine-synthesizing enzyme phenylethanolamine N-methyltransferase (PNMT, E.C. 2.1.1.28) accelerates in response to hormonal and neural stimuli. Cholinergic stimulation through neuronal nicotinic receptors constitutes the primary means for neural regulation of PNMT expression in the adrenal medulla (AM). Therefore, the regulatory sequence conveying responsiveness of the PNMT gene to nicotinic stimuli has been characterized in the 5' upstream region of the rat PNMT promoter. Functional analyses using nested deletion and substitution mutations of the PNMT promoter map the nicotine responsive region to a sequence spanning -633 to -595 bp, designated the PNMT nicotine-responsive element (NicRE). Sequences at the 5' (-633 to -620) and 3' (-599 to -595) ends of this region are essential to convey nicotine responsiveness to PNMT promoter constructs expressed in primary bovine chromaffin cells and in selected lines derived from mouse pheochromocytomas and human neuroblastomas. Profiles of nuclear proteins associating with PNMT promoter sequences also change following nicotine treatment of these cells. Electrophoretic mobility shift and DNase I footprinting analyses distinguish multiple sites of DNA-protein interactions within the NicRE region. Because the PNMT promoter does not contain a cAMP responsive element (the site through which nicotine stimulation is mediated for other catecholamine-synthesizing and AM genes), the NicRE of the PNMT gene must therefore be distinct. Thus, nicotinic cholinergic stimuli appear to regulate expression of the epinephrine-synthesizing gene PNMT through a previously uncharacterized regulatory element.


Assuntos
Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Metiltransferases/genética , Nicotina/farmacologia , Regiões Promotoras Genéticas/efeitos dos fármacos , Glândulas Suprarrenais/enzimologia , Animais , Sequência de Bases , Bovinos , Linhagem Celular Tumoral , Células Cultivadas , Pegada de DNA , Desoxirribonuclease I , Humanos , Metiltransferases/metabolismo , Neurônios/fisiologia , Fosfatidiletanolamina N-Metiltransferase , Receptores Nicotínicos/fisiologia , Proteínas Recombinantes/metabolismo , Deleção de Sequência , Transcrição Gênica/efeitos dos fármacos , Transfecção
10.
J Biol Chem ; 279(27): 27994-9, 2004 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-15117961

RESUMO

Amplification of the N-myc proto-oncogene signifies aggressive behavior in human neuroblastoma. Likewise, overexpression of the intermediate filament nestin, a neuroectodermal stem cell marker, is linked to increased aggressiveness in several nervous system tumors. We investigated the interaction of these two proteins in human neuroblastoma cells. Neuroblastic cell variants with high levels of N-Myc protein have significantly higher nestin protein levels than non-amplified cell lines, suggesting that the transcription factor N-Myc may regulate nestin expression. Stable transfection of a nestin antisense sequence into neuroblastic, N-myc-amplified, LA1-55n cells results in a 2-fold reduction in nestin protein without altering N-Myc expression. However, cell functions attributed to N-Myc (growth rate, anchorage-independent growth, and motility) all decrease significantly. Transfection studies that modulate N-Myc levels also result in commensurate changes in nestin mRNA and protein amounts as well as in cell proliferation and motility. Thus, nestin appears to be downstream of and regulated by N-Myc. Gel mobility shift assays show that N-Myc binds specifically to E-box sequences in the regulatory second intron of the nestin gene and nuclear run-off studies show that increases in N-Myc protein up-regulate nestin transcription rate. Subcellular fractionation and immunoblot studies indicate that nestin is present in the nucleus as well as in the cytoplasm of neuroblastoma cell lines. Finally, DNA cross-linking experiments show that nestin binds DNA in N-myc-amplified N-type cell lines. Thus, nestin may be one mediator of N-myc-associated tumor aggressiveness of human neuroblastoma.


Assuntos
Proteínas de Filamentos Intermediários/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Neuroblastoma/metabolismo , Northern Blotting , Western Blotting , Linhagem Celular Tumoral , Movimento Celular , Núcleo Celular/metabolismo , Reagentes de Ligações Cruzadas/farmacologia , Citoplasma/metabolismo , DNA/metabolismo , Genes myc/genética , Humanos , Immunoblotting , Proteínas de Filamentos Intermediários/metabolismo , Invasividade Neoplásica , Proteínas do Tecido Nervoso/metabolismo , Nestina , Neuroblastoma/patologia , Oligonucleotídeos Antissenso/metabolismo , Oligonucleotídeos Antissenso/farmacologia , Proto-Oncogene Mas , Proteínas Proto-Oncogênicas c-myc/biossíntese , RNA Mensageiro/metabolismo , Fase S , Transfecção
11.
Neoplasia ; 6(6): 838-45, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15720811

RESUMO

Cellular heterogeneity is a hallmark of human neuroblastoma tumors and cell lines. Within a single neuroblastoma are cells from distinct neural crest lineages whose relative abundance is significant for prognosis. We postulate that a self-renewing multipotent tumor stem cell, which gives rise to diverse cell lineages, is the malignant progenitor of this cancer. To test this hypothesis, we have established 22 cloned, phenotypically homogeneous populations of the three major cell types from 17 neuroblastoma cell lines. In vitro, malignant neuroblastoma stem cells, termed I-type (intermediate type), have distinct morphologic, biochemical, differentiative, and tumorigenic properties. I-type cells express features of both neuroblastic (N) cells (scant cytoplasm, neuritic processes, neurofilaments, pseudoganglia, and granin and neurotransmitter enzyme expression) and substrate-adherent (S) cells (extensive cytoplasm and vimentin and CD44 expression). Moreover, they show bidirectional differentiation to either N or S cells when induced by specific agents. I-type cells are significantly more malignant than N- or S-type cells, with four- to five-fold greater plating efficiencies in soft agar and six-fold higher tumorigenicity in athymic mice. Differences in malignant potential are unrelated to N-myc amplification/overexpression or the ability to digest and migrate through the extracellular matrix. Immunocytochemical analyses of a small series of tumors reveal that frequency of cells coexpressing N and S cell markers correlates with poor prognosis. Thus, I-type stem cells may be instrumental in the genesis and growth of tumors in the patient. Their unique biology deserves attention and further investigation.


Assuntos
Linhagem Celular Tumoral/patologia , Neoplasias do Sistema Nervoso/fisiopatologia , Neuroblastoma/fisiopatologia , Células-Tronco/patologia , Células-Tronco/fisiologia , Animais , Western Blotting , Diferenciação Celular/fisiologia , Linhagem da Célula , Movimento Celular/fisiologia , Transformação Celular Neoplásica/patologia , Humanos , Imuno-Histoquímica , Camundongos , Proteínas Proto-Oncogênicas c-myc/biossíntese , Reação em Cadeia da Polimerase Via Transcriptase Reversa
12.
Cancer Lett ; 197(1-2): 35-9, 2003 Jul 18.
Artigo em Inglês | MEDLINE | ID: mdl-12880957

RESUMO

Human neuroblastoma arises from the developing neural crest. Tumors are categorized clinically by their location, age at diagnosis, spread/metastasis, and degree of cellular maturation and heterogeneity. Our long-term studies have shown the presence in human neuroblastoma cell lines of three distinct cell types: I-type stem cells, N-type neuroblastic/neuroendocrine precursors, and S-type Schwannian/melanoblastic precursors. These distinct cell types can differentiate predictably along specific neural crest lineages in response to particular morphogens. As assessed by tumor formation in nude mice and anchorage-independent growth in soft agar, I-type stem cells are significantly more malignant than either N- or S-type cells. Recent research shows that three similar cell types are also present in human neuroblastoma tumors. Using immunocytochemical, laser-capture microdissection, or short-term culture methods to identify cell types in tumors of different stages and/or different outcomes, these studies have shown that (1) all tumors contain neuroblasts in various differentiation states; (2) presumptive I-type stem cells are present in tumors of all stages; and (3) stromal cells may be tumor-derived, i.e. S-type cells, as well as of normal origin. More importantly, there is a higher incidence of I-type cells in tumors that progress, consistent with the high malignant potential of this cell type in vitro. A better understanding of the cause and consequences of cellular heterogeneity of human neuroblastoma tumors is an important prerequisite to the development of more effective therapies for this often fatal disease.


Assuntos
Neoplasias do Sistema Nervoso/patologia , Neuroblastoma/patologia , Animais , Diferenciação Celular , Linhagem da Célula , Transformação Celular Neoplásica , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Incidência , Proteínas de Neoplasias/metabolismo , Neoplasias do Sistema Nervoso/metabolismo , Neuroblastoma/metabolismo , Células Tumorais Cultivadas
14.
Oncol Res ; 13(2): 87-94, 2002.
Artigo em Inglês | MEDLINE | ID: mdl-12392156

RESUMO

Human neuroblastoma cell lines comprise cellular counterparts of normal differentiation phenotypes arising from the developing neural crest Three distinct cell types have been isolated from cell lines: N-type cells with properties of embryonic sympathoadrenoblasts, S-type cells resembling nonneuronal Schwannian/glial/melanoblastic precursors, and I-type stem cells that can differentiate into either N- or S-type cells. Sympathoadrenoblasts from the normal neural crest further differentiate into neuronal or neuroendocrine cells. In this study, we show that malignant N-type neuroblasts likewise can differentiate futher along these same pathways. Retinoic acid and forskolin induce a neuronalphenotype, denoted morphologically by cell aggregation and increased neurite formation and biochemically by increases in neurofilament proteins, tyrosine hydroxylase, and secretogranin II and decrease inchromogranin A. By contrast, dexamethasone, a synthetic glucocorticoid, induces a chromaffin cell phenotype characterized by increased cell flattening, loss of neuritic processes, increased chromogranin A and tyrosine hydroxylase proteins, and decreased amounts of secretogranin II and neurofilaments. N-myc gene expression is upregulated by glucocorticoids; dexamethasone-treated N-type cells show significant (2.3- to 7.8-fold) increases in N-myc mRNA and protein steady-state levels. This effect is specific for glucocorticosteroids, is blocked by addition of the steroid receptor antagonist RU486, and involves direct activation of the N-myc promoter. These findings are the first to show that glucocorticoids upregulate N-myc expression in human neuroblastoma cells.


Assuntos
Dexametasona/farmacologia , Neuroblastoma/patologia , Sistemas Neurossecretores/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-myc/biossíntese , Diferenciação Celular/efeitos dos fármacos , Células Cromafins/citologia , Células Cromafins/efeitos dos fármacos , Genes myc , Humanos , Mifepristona/farmacologia , Sistemas Neurossecretores/citologia , Regiões Promotoras Genéticas , Proteínas Proto-Oncogênicas c-myc/análise , Proteínas Proto-Oncogênicas c-myc/genética , RNA Mensageiro/análise
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA