Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Cancer Med ; 12(9): 10768-10780, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36880347

RESUMEN

Multidrug resistance (MDR) is a primary limitation of breast cancer chemotherapy. The common mechanism of MDR is various anticancer drugs can be effluxed by the cell membrane protein P-glycoprotein (P-gp). Here, we found that ectopic overexpression of Shc3 was detected specifically in drug-resistant breast cancer cells, consequently reducing sensitivity to chemotherapy and promoting cell migration by mediating P-gp expression. However, the molecular mechanism underlying the interplay between P-gp and Shc3 in breast cancer is unknown. We reported an additional resistance mechanism involving an increase in the active form of P-gp after Shc3 upregulation. MCF-7/ADR and SK-BR-3 cells can be sensitive to doxorubicin after knockdown of Shc3. Our results indicated that the interaction between ErbB2 and EphA2 is indirect and regulated by Shc3, and also, this complex is essential for activation of the MAPK and AKT pathways. Meanwhile, Shc3 promotes ErbB2 nuclear translocation, followed by a subsequent increase of the COX2 expression through ErbB2 binding to the COX2 promoter. We further demonstrated that COX2 expression was positively correlated with P-gp expression and the Shc3/ErbB2/COX2 axis upregulates P-gp activity in vivo. Our results show the crucial roles of Shc3 and ErbB2 in modulating P-gp efficacy in breast cancer cells and suggest that Shc3 inhibition may enhance the sensitivity to chemotherapeutic drugs that target oncogene addiction pathways.


Asunto(s)
Antineoplásicos , Neoplasias de la Mama , Humanos , Femenino , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Ciclooxigenasa 2/metabolismo , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/genética , Antineoplásicos/uso terapéutico , Doxorrubicina/farmacología , Doxorrubicina/uso terapéutico , Resistencia a Múltiples Medicamentos , Resistencia a Antineoplásicos/genética , Regulación Neoplásica de la Expresión Génica , Línea Celular Tumoral , Proteína Transformadora 3 que Contiene Dominios de Homología 2 de Src/metabolismo , Receptor ErbB-2/genética , Receptor ErbB-2/metabolismo
2.
Int J Mol Sci ; 22(15)2021 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-34360699

RESUMEN

Reactive astrocytes are a hallmark of neurodegenerative disease including multiple sclerosis. It is widely accepted that astrocytes may adopt alternative phenotypes depending on a combination of environmental cues and intrinsic features in a highly plastic and heterogeneous manner. However, we still lack a full understanding of signals and associated signaling pathways driving astrocyte reaction and of the mechanisms by which they drive disease. We have previously shown in the experimental autoimmune encephalomyelitis mouse model that deficiency of the molecular adaptor Rai reduces disease severity and demyelination. Moreover, using primary mouse astrocytes, we showed that Rai contributes to the generation of a pro-inflammatory central nervous system (CNS) microenvironment through the production of nitric oxide and IL-6 and by impairing CD39 activity in response to soluble factors released by encephalitogenic T cells. Here, we investigated the impact of Rai expression on astrocyte function both under basal conditions and in response to IL-17 treatment using a proteomic approach. We found that astrocytes and astrocyte-derived extracellular vesicles contain a set of proteins, to which Rai contributes, that are involved in the regulation of oligodendrocyte differentiation and myelination, nitrogen metabolism, and oxidative stress. The HIF-1α pathway and cellular energetic metabolism were the most statistically relevant molecular pathways and were related to ENOA and HSP70 dysregulation.


Asunto(s)
Astrocitos/metabolismo , Encefalomielitis Autoinmune Experimental/metabolismo , Vesículas Extracelulares/metabolismo , Interleucina-17/farmacología , Neuroprotección , Oligodendroglía/fisiología , Proteína Transformadora 3 que Contiene Dominios de Homología 2 de Src/genética , Animales , Diferenciación Celular , Encefalomielitis Autoinmune Experimental/genética , Encefalomielitis Autoinmune Experimental/fisiopatología , Regulación de la Expresión Génica , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Vaina de Mielina , Proteómica , Proteína Transformadora 3 que Contiene Dominios de Homología 2 de Src/metabolismo
3.
Cell Death Dis ; 12(3): 278, 2021 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-33723262

RESUMEN

Hepatocellular carcinoma (HCC) is one of the most common cancers with an insidious onset, strong invasiveness, insensitivity to chemotherapy, and poor prognosis, thus makes clinical treatment challenging. The mechanisms require further elucidation for developing novel therapies and targeting drug resistance. Here, we observed high Shc3 expression in patients with chemoresistant and recurrent HCCs. Shc3 overexpression induced a significant increase in MDR1/P-glycoprotein expression, whereas Shc3 knockdown impaired this expression. Further, Shc3 inhibition significantly restored HCC cell sensitivity to doxorubicin and sorafenib. Mechanistically, Shc3 interacted with ß-catenin, inhibited destruction complex stability, promoted ß-catenin release, and dampened ß-catenin ubiquitination. Shc3 bound ß-catenin and facilitated its nuclear translocation, prompting the ß-catenin/TCF pathway to elevate MDR1 transcription. ß-catenin blockage abolished the discrepancy in drug resistance between Shc3-depleted HCC cells and control cells, which further validating that ß-catenin is required for Shc3-mediated liver chemotherapy. We also determined the effect of Shc3 on the sensitivity of HCC to chemotherapy in vivo. Collectively, this study provides a potential strategy to target these pathways concurrently with systemic chemotherapy that can improve the clinical treatment of HCC.


Asunto(s)
Antineoplásicos/farmacología , Carcinoma Hepatocelular/tratamiento farmacológico , Doxorrubicina/farmacología , Resistencia a Antineoplásicos , Neoplasias Hepáticas/tratamiento farmacológico , Células Madre Neoplásicas/efectos de los fármacos , Proteína Transformadora 3 que Contiene Dominios de Homología 2 de Src/metabolismo , beta Catenina/metabolismo , Subfamilia B de Transportador de Casetes de Unión a ATP/genética , Subfamilia B de Transportador de Casetes de Unión a ATP/metabolismo , Animales , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Femenino , Regulación Neoplásica de la Expresión Génica , Células Hep G2 , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Masculino , Ratones Endogámicos BALB C , Ratones Desnudos , Persona de Mediana Edad , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Proteolisis , Transducción de Señal , Proteína Transformadora 3 que Contiene Dominios de Homología 2 de Src/genética , Factores de Transcripción TCF/genética , Factores de Transcripción TCF/metabolismo , Ubiquitinación , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Cells ; 9(5)2020 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-32443613

RESUMEN

Adaptation of glioblastoma to caloric restriction induces compensatory changes in tumor metabolism that are incompletely known. Here we show that in human glioblastoma cells maintained in exhausted medium, SHC adaptor protein 3 (SHC3) increases due to down-regulation of SHC3 protein degradation. This effect is reversed by glucose addition and is not present in normal astrocytes. Increased SHC3 levels are associated to increased glucose uptake mediated by changes in membrane trafficking of glucose transporters of the solute carrier 2A superfamily (GLUT/SLC2A). We found that the effects on vesicle trafficking are mediated by SHC3 interactions with adaptor protein complex 1 and 2 (AP), BMP-2-inducible protein kinase and a fraction of poly ADP-ribose polymerase 1 (PARP1) associated to vesicles containing GLUT/SLC2As. In glioblastoma cells, PARP1 inhibitor veliparib mimics glucose starvation in enhancing glucose uptake. Furthermore, cytosol extracted from glioblastoma cells inhibits PARP1 enzymatic activity in vitro while immunodepletion of SHC3 from the cytosol significantly relieves this inhibition. The identification of a new pathway controlling glucose uptake in high grade gliomas represents an opportunity for repositioning existing drugs and designing new ones.


Asunto(s)
Adaptación Fisiológica , Neoplasias Encefálicas/patología , Glioblastoma/patología , Glucosa/deficiencia , Transducción de Señal , Adaptación Fisiológica/efectos de los fármacos , Bencimidazoles/farmacología , Neoplasias Encefálicas/ultraestructura , Línea Celular Tumoral , Endocitosis/efectos de los fármacos , Glioblastoma/ultraestructura , Transportador de Glucosa de Tipo 1/metabolismo , Glicosilación/efectos de los fármacos , Humanos , Ácido Láctico/biosíntesis , Poli(ADP-Ribosa) Polimerasa-1/metabolismo , Poli Adenosina Difosfato Ribosa/metabolismo , Unión Proteica/efectos de los fármacos , Dominios Proteicos , Estabilidad Proteica/efectos de los fármacos , Transporte de Proteínas/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Proteína Transformadora 3 que Contiene Dominios de Homología 2 de Src/química , Proteína Transformadora 3 que Contiene Dominios de Homología 2 de Src/metabolismo , Vesículas Transportadoras/efectos de los fármacos , Vesículas Transportadoras/metabolismo
5.
Biomolecules ; 10(2)2020 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-32024191

RESUMEN

In the last decade, Nerve Growth Factor (NGF)-based clinical approaches have lacked specific and efficient Tyrosine Kinase A (TrkA) agonists for brain delivery. Nowadays, the characterization of novel small peptidomimetic is taking centre stage in preclinical studies, in order to overcome the main size-related limitation in brain delivery of NGF holoprotein for Central Nervous System (CNS) pathologies. Here we investigated the NGF mimetic properties of the human NGF 1-14 sequence (hNGF1-14) and its derivatives, by resorting to primary cholinergic and dorsal root ganglia (DRG) neurons. Briefly, we observed that: 1) hNGF1-14 peptides engage the NGF pathway through TrkA phosphorylation at tyrosine 490 (Y490), and activation of ShcC/PI3K and Plc-γ/MAPK signalling, promoting AKT-dependent survival and CREB-driven neuronal activity, as seen by levels of the immediate early gene c-Fos, of the cholinergic marker Choline Acetyltransferase (ChAT), and of Brain Derived Neurotrophic Factor (BDNF); 2) their NGF mimetic activity is lost upon selective TrkA inhibition by means of GW441756; 3) hNGF1-14 peptides are able to sustain DRG survival and differentiation in absence of NGF. Furthermore, the acetylated derivative Ac-hNGF1-14 demonstrated an optimal NGF mimetic activity in both neuronal paradigms and an electrophysiological profile similar to NGF in cholinergic neurons. Cumulatively, the findings here reported pinpoint the hNGF1-14 peptide, and in particular its acetylated derivative, as novel, specific and low molecular weight TrkA specific agonists in both CNS and PNS primary neurons.


Asunto(s)
Neuronas Colinérgicas/metabolismo , Ganglios Espinales/metabolismo , Factor de Crecimiento Nervioso/química , Receptor trkA/agonistas , Receptor trkA/metabolismo , Proteína Transformadora 3 que Contiene Dominios de Homología 2 de Src/metabolismo , Animales , Bioensayo , Diferenciación Celular , Supervivencia Celular , Células Cultivadas , Humanos , Péptidos/química , Fosforilación , Ratas , Transducción de Señal , Tirosina/química
6.
BMC Neurosci ; 20(1): 57, 2019 12 10.
Artículo en Inglés | MEDLINE | ID: mdl-31823725

RESUMEN

BACKGROUND: Mammalian Shc (Src homology and collagen) proteins comprise a family of four phosphotyrosine adaptor molecules which exhibit varied spatiotemporal expression and signaling functions. ShcD is the most recently discovered homologue and it is highly expressed in the developing central nervous system (CNS) and adult brain. Presently however, its localization within specific cell types of mature neural structures has yet to be characterized. RESULTS: In the current study, we examine the expression profile of ShcD in the adult rat CNS using immunohistochemistry, and compare with those of the neuronally enriched ShcB and ShcC proteins. ShcD shows relatively widespread distribution in the adult brain and spinal cord, with prominent levels of staining throughout the olfactory bulb, as well as in sub-structures of the cerebellum and hippocampus, including the subgranular zone. Co-localization studies confirm the expression of ShcD in mature neurons and progenitor cells. ShcD immunoreactivity is primarily localized to axons and somata, consistent with the function of ShcD as a cytoplasmic adaptor. Regional differences in expression are observed among neural Shc proteins, with ShcC predominating in the hippocampus, cerebellum, and some fiber tracts. Interestingly, ShcD is uniquely expressed in the olfactory nerve layer and in glomeruli of the main olfactory bulb. CONCLUSIONS: Together our findings suggest that ShcD may provide a distinct signaling contribution within the olfactory system, and that overlapping expression of ShcD with other Shc proteins may allow compensatory functions in the brain.


Asunto(s)
Sistema Nervioso Central/metabolismo , Proteínas Adaptadoras de la Señalización Shc/metabolismo , Animales , Sistema Nervioso Central/citología , Inmunohistoquímica , Masculino , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Neuronas/citología , Neuronas/metabolismo , Ratas Sprague-Dawley , Proteína Transformadora 2 que Contiene Dominios de Homología 2 de Src/metabolismo , Proteína Transformadora 3 que Contiene Dominios de Homología 2 de Src/metabolismo
7.
Front Immunol ; 10: 1041, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31134091

RESUMEN

Multiple sclerosis is an autoimmune disease caused by autoreactive immune cell infiltration into the central nervous system leading to inflammation, demyelination, and neuronal loss. While myelin-reactive Th1 and Th17 are centrally implicated in multiple sclerosis pathogenesis, the local CNS microenvironment, which is shaped by both infiltrated immune cells and central nervous system resident cells, has emerged a key player in disease onset and progression. We have recently demonstrated that ShcC/Rai is as a novel astrocytic adaptor whose loss in mice protects from experimental autoimmune encephalomyelitis. Here, we have explored the mechanisms that underlie the ability of Rai-/- astrocytes to antagonize T cell-dependent neuroinflammation. We show that Rai deficiency enhances the ability of astrocytes to upregulate the expression and activity of the ectonucleotidase CD39, which catalyzes the conversion of extracellular ATP to the immunosuppressive metabolite adenosine, through both contact-dependent and-independent mechanisms. As a result, Rai-deficient astrocytes acquire an enhanced ability to suppress T-cell proliferation, which involves suppression of T cell receptor signaling and upregulation of the inhibitory receptor CTLA-4. Additionally, Rai-deficient astrocytes preferentially polarize to the neuroprotective A2 phenotype. These results identify a new mechanism, to which Rai contributes to a major extent, by which astrocytes modulate the pathogenic potential of autoreactive T cells.


Asunto(s)
Antígenos CD/metabolismo , Apirasa/metabolismo , Astrocitos/inmunología , Linfocitos T CD4-Positivos/inmunología , Encefalomielitis Autoinmune Experimental/inmunología , Proteína Transformadora 3 que Contiene Dominios de Homología 2 de Src/genética , Animales , Linfocitos T CD8-positivos/inmunología , Antígeno CTLA-4/metabolismo , Proliferación Celular/genética , Células Cultivadas , Sistema Nervioso Central/inmunología , Sistema Nervioso Central/patología , Encefalomielitis Autoinmune Experimental/patología , Femenino , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína Transformadora 3 que Contiene Dominios de Homología 2 de Src/metabolismo
8.
Cell Physiol Biochem ; 49(3): 971-984, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30184529

RESUMEN

BACKGROUND/AIMS: Parkinson's disease (PD) is a prevalent disease that leads to motor and cognitive disabilities, and oxidative stress (OS) injury was found to be related to the etiology of PD. Increasing evidence has shown that SHC3 is aberrantly expressed in neurons. The current study examines the involvement of SHC3 silencing in OS injury in the nigral dopamine neurons in rats with PD via the PI3K-AKT-FoxO signaling pathway. METHODS: To study the mechanisms and functions of SHC3 silencing in PD at the tissue level, 170 rats were selected, and a lentivirus-based packaging system was designed to silence SHC3 expression in rats. Furthermore, PC12 cells were selected for in vitro experimentation. To evaluate the effect of SHC3 silencing in nigral dopamine neuronal growth, an MTT assay, propidium iodide (PI) single staining and Annexin V-PI double staining were performed to detect cell viability, cell cycle progression and cell apoptosis, respectively. RESULTS: SHC3 shRNA led to decreased SOD and MDA levels and enhanced GSH activity, indicating that SHC3 silencing leads to motor retardation. SHC3 silencing repressed the extent of Akt and FoxO phosphorylation, thereby inhibiting the PI3K-AKT-FoxO signaling pathway. Furthermore, in cell experiments, SHC3 silencing suppressed PC12 cell proliferation and cell cycle progression, whereas it enhanced cell apoptosis. CONCLUSION: The current study provides evidence suggesting that SHC3 silencing may aggravate OS injury in nigral dopamine neurons via downregulation of the PI3K-AKT-FoxO signaling pathway in PD rats.


Asunto(s)
Neuronas Dopaminérgicas/metabolismo , Estrés Oxidativo , Transducción de Señal , Proteína Transformadora 3 que Contiene Dominios de Homología 2 de Src/metabolismo , Animales , Conducta Animal , Modelos Animales de Enfermedad , Factores de Transcripción Forkhead/metabolismo , Lentivirus/genética , Masculino , Malondialdehído/metabolismo , Células PC12 , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/patología , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Ratas , Ratas Sprague-Dawley , Proteína Transformadora 3 que Contiene Dominios de Homología 2 de Src/antagonistas & inhibidores , Proteína Transformadora 3 que Contiene Dominios de Homología 2 de Src/genética , Superóxido Dismutasa/metabolismo
9.
J Cell Biol ; 215(5): 667-685, 2016 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-27872138

RESUMEN

Macropinocytosis, by which cells ingest large amounts of fluid, and autophagy, the lysosome-based catabolic process, involve vesicular biogenesis (early stage) and turnover (end stage). Much is known about early-stage events; however, our understanding of how the end stages of these processes are governed is incomplete. Here we demonstrate that the microRNA-103/107(miR-103/107) family, which is preferentially expressed in the stem cell-enriched limbal epithelium, coordinately regulates aspects of both these activities. Loss of miR-103/107 causes dysregulation of macropinocytosis with the formation of large vacuoles, primarily through up-regulation of Src, Ras, and Ankfy1. Vacuole accumulation is not a malfunction of early-stage autophagy; rather, miR-103/107 ensure proper end-stage autophagy by regulating diacylglycerol/protein kinase C and cyclin-dependent kinase 5 signaling, which enables dynamin to function in vacuole clearance. Our findings unveil a key biological function for miR-103/107 in coordinately suppressing macropinocytosis and preserving end-stage autophagy, thereby contributing to maintenance of a stem cell-enriched epithelium.


Asunto(s)
Autofagia , MicroARNs/metabolismo , Pinocitosis , Animales , Biomarcadores/metabolismo , Proliferación Celular , Diglicéridos/metabolismo , Dinaminas/metabolismo , Células Epiteliales/citología , Células Epiteliales/metabolismo , Humanos , Queratinocitos/citología , Queratinocitos/metabolismo , Queratinocitos/ultraestructura , Lisosomas/metabolismo , Proteínas de la Membrana , Ratones Transgénicos , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas de Unión a Fosfato , Proteína Transformadora 3 que Contiene Dominios de Homología 2 de Src/metabolismo , Vacuolas/metabolismo , Vacuolas/ultraestructura , Proteínas de Unión al GTP rab/metabolismo
10.
Sci Rep ; 6: 27511, 2016 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-27273072

RESUMEN

BDNF-TrkB signaling is implicated in experimental seizures and epilepsy. However, the downstream signaling involved in the epileptiform activity caused by TrkB receptor activation is still unknown. The aim of the present study was to determine whether TrkB-mediated N-Shc signal transduction was involved in kainic acid (KA)-induced epileptiform activity. We investigated KA-induced behavioral seizures, epileptiform activities and neuronal cell loss in hippocampus between N-Shc deficient and control mice. There was a significant reduction in seizure severity and the frequency of epileptiform discharges in N-Shc deficient mice, as compared with wild-type and C57BL/6 mice. KA-induced neuronal cell loss in the CA3 of hippocampus was also inhibited in N-Shc deficient mice. This study demonstrates that the activation of N-Shc signaling pathway contributes to an acute KA-induced epileptiform activity and neuronal cell loss in the hippocampus. We propose that the N-Shc-mediated signaling pathway could provide a potential target for the novel therapeutic approaches of epilepsy.


Asunto(s)
Ácido Kaínico/farmacología , Neuronas/metabolismo , Fosfotirosina/metabolismo , Convulsiones/metabolismo , Transducción de Señal , Proteína Transformadora 3 que Contiene Dominios de Homología 2 de Src/metabolismo , Animales , Ratones , Convulsiones/inducido químicamente
11.
Aging Cell ; 15(4): 661-72, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27076121

RESUMEN

NGF has been implicated in forebrain neuroprotection from amyloidogenesis and Alzheimer's disease (AD). However, the underlying molecular mechanisms are still poorly understood. Here, we investigated the role of NGF signalling in the metabolism of amyloid precursor protein (APP) in forebrain neurons using primary cultures of septal neurons and acute septo-hippocampal brain slices. In this study, we show that NGF controls the basal level of APP phosphorylation at Thr668 (T668) by downregulating the activity of the Ser/Thr kinase JNK(p54) through the Tyr kinase signalling adaptor SH2-containing sequence C (ShcC). We also found that the specific NGF receptor, Tyr kinase A (TrkA), which is known to bind to APP, fails to interact with the fraction of APP molecules phosphorylated at T668 (APP(pT668) ). Accordingly, the amount of TrkA bound to APP is significantly reduced in the hippocampus of ShcC KO mice and of patients with AD in which elevated APP(pT668) levels are detected. NGF promotes TrkA binding to APP and APP trafficking to the Golgi, where APP-BACE interaction is hindered, finally resulting in reduced generation of sAPPß, CTFß and amyloid-beta (1-42). These results demonstrate that NGF signalling directly controls basal APP phosphorylation, subcellular localization and BACE cleavage, and pave the way for novel approaches specifically targeting ShcC signalling and/or the APP-TrkA interaction in AD therapy.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Precursor de Proteína beta-Amiloide/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Factor de Crecimiento Nervioso/farmacología , Fosfotreonina/metabolismo , Adulto , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Animales , Ácido Aspártico Endopeptidasas/metabolismo , Activación Enzimática/efectos de los fármacos , Eliminación de Gen , Aparato de Golgi/efectos de los fármacos , Aparato de Golgi/metabolismo , Hipocampo/patología , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Masculino , Ratones Endogámicos C57BL , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Células PC12 , Fosforilación/efectos de los fármacos , Unión Proteica/efectos de los fármacos , Ratas , Receptor trkA/metabolismo , Proteína Transformadora 3 que Contiene Dominios de Homología 2 de Src/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA