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










Base de datos
Intervalo de año de publicación
1.
J Gastroenterol ; 59(3): 229-249, 2024 03.
Artículo en Inglés | MEDLINE | ID: mdl-38310161

RESUMEN

BACKGROUND: Liver fibrosis can progress to cirrhosis and hepatic carcinoma without treatment. CircDCBLD2 was found to be downregulated in liver fibrosis. However, the precise underlying mechanism requires further investigation. METHODS: qRT-PCR, Western blot, and immunohistochemistry assays were used to detect the related molecule levels. HE, Masson's trichrome, and Sirius Red staining were used to assess the pathological changes in mice's liver tissues. Flow cytometric analysis and commercial kit were used to assess the levels of lipid reactive oxygen species (ROS), malonaldehyde (MDA), glutathione (GSH), and iron. Cell viability was assessed by MTT. Immunoprecipitation was used to study the ubiquitination of PARK7. Mitophagy was determined by immunostaining and confocal imaging. RIP and Co-IP assays were used to assess the interactions of circDCBLD2/HuR, HuR/STUB1, and STUB1/PARK7. Fluorescence in situ hybridization and immunofluorescence staining were used to assess the co-localization of circDCBLD2 and HuR. RESULTS: CircDCBLD2 was downregulated, whereas PARK7 was upregulated in liver fibrosis. Ferroptosis activators increased circDCBLD2 while decreasing PARK7 in hepatic stellate cells (HSCs) and mice with liver fibrosis. CircDCBLD2 overexpression reduced cell viability and GSH, PARK7, and GPX4 expression in erastin-treated HSCs while increasing MDA and iron levels, whereas circDCBLD2 knockdown had the opposite effect. CircDCBLD2 overexpression increased STUB1-mediated PARK7 ubiquitination by promoting HuR-STUB1 binding and thus increasing STUB1 mRNA stability. PARK7 overexpression or HuR knockdown reversed the effects of circDCBLD2 overexpression on HSC activation and ferroptosis. CircDCBLD2 reduced liver fibrosis in mice by inhibiting PARK7. CONCLUSION: CircDCBLD2 overexpression increased PARK7 ubiquitination degradation by upregulating STUB1 through its interaction with HuR, inhibiting HSC activation and promoting HSC ferroptosis, ultimately enhancing liver fibrosis.


Asunto(s)
Ferroptosis , Neoplasias Hepáticas , Animales , Ratones , Células Estrelladas Hepáticas/metabolismo , Hibridación Fluorescente in Situ , Hierro/metabolismo , Hierro/farmacología , Cirrosis Hepática/patología , Neoplasias Hepáticas/patología , Proteína Desglicasa DJ-1/genética , Proteína Desglicasa DJ-1/metabolismo , Proteína Desglicasa DJ-1/farmacología , Ubiquitinación
2.
ACS Chem Neurosci ; 14(12): 2294-2301, 2023 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-37289979

RESUMEN

Parkinson's disease (PD) is an incurable neurodegenerative disorder caused by the selective loss of dopaminergic neurons in the substantia nigra pars compacta. Current therapies are only symptomatic and are not able to stop or delay its progression. In order to search for new and more effective therapies, our group carried out a high-throughput screening assay, identifying several candidate compounds that are able to improve locomotor ability in DJ-1ß mutant flies (a Drosophila model of familial PD) and reduce oxidative stress (OS)-induced lethality in DJ-1-deficient SH-SY5Y human cells. One of them was vincamine (VIN), a natural alkaloid obtained from the leaves of Vinca minor. Our results showed that VIN is able to suppress PD-related phenotypes in both Drosophila and human cell PD models. Specifically, VIN reduced OS levels in PD model flies. Besides, VIN diminished OS-induced lethality by decreasing apoptosis, increased mitochondrial viability, and reduced OS levels in DJ-1-deficient human cells. In addition, our results show that VIN might be exerting its beneficial role, at least partially, by the inhibition of voltage-gated sodium channels. Therefore, we propose that these channels might be a promising target in the search for new compounds to treat PD and that VIN represents a potential therapeutic treatment for the disease.


Asunto(s)
Proteínas de Drosophila , Neuroblastoma , Enfermedad de Parkinson , Vincamina , Animales , Humanos , Suplementos Dietéticos , Drosophila/genética , Proteínas de Drosophila/genética , Proteínas del Tejido Nervioso/genética , Estrés Oxidativo , Enfermedad de Parkinson/tratamiento farmacológico , Enfermedad de Parkinson/genética , Proteína Desglicasa DJ-1/genética , Proteína Desglicasa DJ-1/farmacología , Proteína Desglicasa DJ-1/uso terapéutico , Vincamina/farmacología , Vincamina/uso terapéutico
3.
Adv Sci (Weinh) ; 10(15): e2206007, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36967569

RESUMEN

Doxorubicin (DOX)-induced cardiotoxicity (DoIC) is a major side effect for cancer patients. Recently, ferroptosis, triggered by iron overload, is demonstrated to play a role in DoIC. How iron homeostasis is dysregulated in DoIC remains to be elucidated. Here, the authors demonstrate that DOX challenge exhibits reduced contractile function and induction of ferroptosis-related phenotype in cardiomyocytes, evidenced by iron overload, lipid peroxide accumulation, and mitochondrial dysfunction. Compared to Ferric ammonium citrate (FAC) induced secondary iron overload, DOX-challenged cardiomyocytes show a dysfunction of iron homeostasis, with decreased cytoplasmic and mitochondrial iron-sulfur (FeS) cluster-mediated aconitase activity and abnormal expression of iron homeostasis-related proteins. Mechanistically, mass spectrometry analysis identified DOX-treatment induces p53-dependent degradation of Parkinsonism associated deglycase (Park7) which results in iron homeostasis dysregulation. Park7 counteracts iron overload by regulating iron regulatory protein family transcription while blocking mitochondrial iron uptake. Knockout of p53 or overexpression of Park7 in cardiomyocytes remarkably restores the activity of FeS cluster and iron homeostasis, inhibits ferroptosis, and rescues cardiac function in DOX treated animals. These results demonstrate that the iron homeostasis plays a key role in DoIC ferroptosis. Targeting of the newly identified p53-Park7 signaling axis may provide a new approach to prevent DoIC.


Asunto(s)
Sobrecarga de Hierro , Miocitos Cardíacos , Animales , Proteína p53 Supresora de Tumor/metabolismo , Proteína Desglicasa DJ-1/metabolismo , Proteína Desglicasa DJ-1/farmacología , Doxorrubicina/efectos adversos , Hierro/metabolismo , Homeostasis
4.
Toxicol Lett ; 379: 56-66, 2023 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-36965608

RESUMEN

AIMS: DJ-1, a multifunctional protein encoded by the Park7 gene, is tightly related to mitochondrial dysfunction, oxidative stress, protein aggregation, and autophagy regulation. The current study was designed to investigate whether DJ-1 is expressed in auditory cells and, if so, to explore the possible correlation between DJ-1 and cisplatin-induced ototoxicity in this type of cells. METHODS: The location and dynamic expression of DJ-1 in mouse cochlea hair cells (HCs) and House Ear Institute-Organ of Corti 1 (HEI-OC1 cells) were detected by immunofluorescence, real-time PCR, and western blot. The apoptosis of auditory cells was assessed by TUNEL staining and flow cytometry. The levels of ROS were evaluated by MitoSox red staining. The expression of protein cleaved caspase-9, cleaved caspase-3, and LC3B was examined by immunofluorescence and western blot. The expressions of certain key factors relevant to apoptosis (Bcl-2 and Bax) and autophagy (Beclin1, p-JNK, and p-c-Jun) were determined by western blot. The dynamic alterations of those factors in response to DJ-1 knockdown in HEI-OC1 cells (DJ-1-KD) were measured by western blot and MitoSox red staining. RESULTS: The expression of DJ-1 was clearly shown in both HCs and HEI-OC1 cells and cisplatin led to the reduction of DJ-1 expression in a concentration and time-dependent manner. Meanwhile, cisplatin-induced apoptotic process was implemented by promoting reactive oxygen species (ROS) production and activating the mitochondrial pathway. Furthermore, DJ-1 explicitly participated in cisplatin-trigged cell damage by regulating autophagy. CONCLUSIONS: Findings from this work clearly reveal, for the first time, that DJ-1 is expressed in the cochlea. Of particular importance, DJ-1 exerts its protective action against cisplatin-elicited injury on auditory cells via regulating apoptosis and autophagy, which provides a new strategy for the prevention of cisplatin-induced ototoxicity.


Asunto(s)
Antineoplásicos , Ototoxicidad , Ratones , Animales , Cisplatino/toxicidad , Antineoplásicos/toxicidad , Especies Reactivas de Oxígeno/metabolismo , Ototoxicidad/prevención & control , Apoptosis , Autofagia , Supervivencia Celular , Proteína Desglicasa DJ-1/genética , Proteína Desglicasa DJ-1/farmacología
5.
Tumori ; 109(1): 47-53, 2023 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-34918581

RESUMEN

INTRODUCTION: PARK7/DJ-1 is an oncogene that is associated with tumorigenesis in many cancers. Recent studies have demonstrated the importance of DJ-1 in the origin and development of uveal melanoma (UM). We present an analysis of the role of the DJ-1 protein in UM cells, especially in its effect on proliferation and migration. METHODS: UM cells from a primary tumor, Mel 270, and its liver metastasis, OMM2.5, were transfected with lentiviral-delivered shRNA against PARK7/DJ-1. Evaluation of cell migration and proliferation was performed using the xCELLigence real-time cell analyzer (RTCA). The effect of DJ-1 inhibition on the PTEN-Akt signaling pathway was also studied by immunoblotting. RESULTS: The silencing of PARK7/DJ-1 oncoprotein expression produced a significant decrease of phosphorylated Akt (S473) in Mel270 and in metastatic OMM2.5 UM cells with no alteration on tumor suppressor PTEN expression. The diminution of PARK7/DJ-1 expression significantly inhibited real-time proliferation and invasion of Mel270 and OMM2.5 and the invasion potential of the metastatic cells. CONCLUSION: DJ-1 appears to play a key role on the PTEN/Akt pathway in UM. DJ-1 inhibition appears to have a negative effect on proliferation and invasion of UM cells. This suggests DJ-1 as a potential therapeutic target in UM.


Asunto(s)
Proteínas Proto-Oncogénicas c-akt , Neoplasias de la Úvea , Humanos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteína Desglicasa DJ-1/genética , Proteína Desglicasa DJ-1/metabolismo , Proteína Desglicasa DJ-1/farmacología , Proliferación Celular , Transducción de Señal , Neoplasias de la Úvea/genética , Neoplasias de la Úvea/metabolismo , Neoplasias de la Úvea/patología , Línea Celular Tumoral
6.
Pharmacol Res ; 187: 106607, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36509316

RESUMEN

BACKGROUND: As a pathological myocardial remodeling process in a variety of cardiovascular diseases, cardiac hypertrophy still has no effective treatment. Human mesenchymal stem cells (hMSCs) derived extracellular vesicles (EVs) has been recognized as a promising treatment strategy for cardiac disease. METHODS: In this study, the inhibitory effects on cardiac hypertrophy are compared between normoxia-conditioned hMSC-derived EVs (Nor-EVs) and hypoxia-conditioned hMSC-derived EVs (Hypo-EVs) in neonatal rat cardiomyocytes (NRCMs) after angiotensin II (Ang II) stimulation and in a mouse model of transverse aortic constriction (TAC). RESULTS: We demonstrate that Hypo-EVs exert an increased inhibitory effect on cardiac hypertrophy compared with Nor-EVs. Parkinson disease protein 7 (PARK7/DJ-1) is identify as a differential protein between Nor-EVs and Hypo-EVs by quantitative proteomics analysis. Results show that DJ-1, which is rich in Hypo-EVs, alleviates mitochondrial dysfunction and excessive mitochondrial reactive oxygen species (mtROS) production as an antioxidant. Mechanistic studies demonstrate for the first time that DJ-1 may suppress cardiac hypertrophy by inhibiting the activity of proteasome subunit beta type 10 (PSMB10) through a direct physical interaction. This interaction can inhibit angiotensin II type 1 receptor (AT1R)-mediated signaling pathways resulting in cardiac hypertrophy through alleviating ubiquitination degradation of AT1R-associated protein (ATRAP). CONCLUSIONS: When taken together, our study suggests that Hypo-EVs have significant potential as a novel therapeutic agent for the treatment of cardiac hypertrophy.


Asunto(s)
Vesículas Extracelulares , Células Madre Mesenquimatosas , Ratones , Ratas , Humanos , Animales , Cardiomegalia/metabolismo , Proteolisis , Transducción de Señal , Vesículas Extracelulares/metabolismo , Miocitos Cardíacos/metabolismo , Células Madre Mesenquimatosas/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteína Desglicasa DJ-1/metabolismo , Proteína Desglicasa DJ-1/farmacología
7.
Angew Chem Int Ed Engl ; 62(9): e202215801, 2023 02 20.
Artículo en Inglés | MEDLINE | ID: mdl-36550087

RESUMEN

Single-cell protein therapeutics is expected to promote our in-depth understanding of how a specific protein with a therapeutic dosage treats the cell without population averaging. However, it has not yet been tackled by current single-cell nanotools. We address this challenge by the use of a double-barrel nanopipette, in which one lumen was used for electroosmotic cytosolic protein delivery and the other was customized for ionic evaluation of the consequence. Upon injection of protein DJ-1 through the delivery lumen, upregulation of the antioxidant protein could protect neural PC-12 cells against oxidative stress from phorbol myristate acetate exposure, as deduced by targeting of the cytosolic hydrogen peroxide by the detecting lumen. The nanotool developed in this study for single-cell protein therapeutics provides a perspective for future single-cell therapeutics involving different therapeutic modalities, such as peptides, enzymes and nucleic acids.


Asunto(s)
Tratamiento Basado en Trasplante de Células y Tejidos , Proteína Desglicasa DJ-1 , Iones , Péptidos , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Sistema de Administración de Fármacos con Nanopartículas , Proteína Desglicasa DJ-1/farmacología , Proteína Desglicasa DJ-1/uso terapéutico , Estrés Oxidativo , Acetato de Tetradecanoilforbol
8.
Life Sci ; 276: 119089, 2021 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-33476627

RESUMEN

BACKGROUND: Chronic obstructive pulmonary disease (COPD) is an inflammatory disease characterized by airway obstruction and abnormal inflammatory responses. Multidrug resistance-related protein 1 (MRP1) can reduce lung inflammation and damage by excreting various toxic exogenous substances and certain pro-inflammatory molecules. AIMS: We studied whether DJ-1 modulates nuclear factor erythroid 2-related factor 2 (Nrf2) by activating the Wnt3a/ß-catenin signalling pathway to further regulate MRP1 expression and pulmonary antioxidant defences in alveolar epithelial (A549) cells treated with smoke extract (CSE) and lipopolysaccharide (LPS). MAIN METHODS: Marker expression was studied by western blot analysis, quantitative real-time PCR and immunofluorescence staining of A549 cells. KEY FINDINGS: A549 cells exposed to CSE and LPS showed downregulation of DJ-1, Wnt3a, MRP1 and haem oxygenase-1 (HO-1) and upregulation of inflammatory factors. Additionally, Nrf2 protein levels were significantly decreased, while there was no change in Nrf2 mRNA levels. Overexpression of DJ-1 and Wnt3a activated Nrf2 signalling, increased MRP1 and HO-1 levels and decreased IL-6 protein expression, while knockdown of DJ-1 and Wnt3a had the opposite effects. Furthermore, DJ-1 overexpression and DJ-1 knockdown increased and decreased, respectively, the levels of Wnt3a and ß-catenin. Interestingly, Nrf2 and Wnt3a deficiency reduced the protective effects of Wnt3a and DJ-1, respectively, in A549 cells. However, the levels of DJ-1 and Wnt3a were not altered by Wnt3a and Nrf2 deletion, respectively. SIGNIFICANCE: In A549 cells treated with CSE and LPS, DJ-1 regulates Nrf2-mediated MRP1 expression and antioxidant defences by activating the Wnt3a/ß-catenin signalling pathway. These findings may provide potential therapeutic targets for COPD intervention.


Asunto(s)
Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Inflamación/prevención & control , Lipopolisacáridos/efectos adversos , Estrés Oxidativo/efectos de los fármacos , Proteína Desglicasa DJ-1/farmacología , Humo/efectos adversos , Productos de Tabaco/efectos adversos , Células A549 , Humanos , Inflamación/inducido químicamente , Inflamación/metabolismo , Inflamación/patología , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/genética , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/metabolismo , Factor 2 Relacionado con NF-E2/genética , Factor 2 Relacionado con NF-E2/metabolismo , Proteína Desglicasa DJ-1/administración & dosificación , Proteína Wnt3A/genética , Proteína Wnt3A/metabolismo , beta Catenina/genética , beta Catenina/metabolismo
9.
Adv Exp Med Biol ; 1037: 187-202, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29147910

RESUMEN

Parkinson's disease (PD) is a progressive neurodegenerative disorder that is primarily characterized by the degeneration of dopaminergic neurons in the nigrostriatal pathway. Loss-of-function mutations in the gene encoding PARK7/DJ-1 were identified in familial PD. Wild-type DJ-1 acts as an oxidative stress sensor in neural cells. Previously, we identified binding compounds of DJ-1, including UCP0045037/compound A, UCP0054278/compound B, and compound-23 (comp-23), by in silico virtual screening. These compounds prevented oxidative stress-induced dopaminergic neuronal death and restored locomotion defects in animal models of PD. In addition, these binding partners reduced infarct size in cerebral ischemia in rats. The neuroprotective effects of these compounds are lost in DJ-1-knockdown cells and DJ-1-knockout animal. These results suggest that these compounds interact with endogenous DJ-1 and then produce antioxidant and neuroprotective responses in both animal models for PD and cerebral ischemia in rats. This raises the possibility that interaction partners of DJ-1, such as UCP0045037, UCP0054278, and comp-23, may represent a novel dopaminergic neuroprotective drug for the treatment of PD.


Asunto(s)
Benzamidas/farmacología , Benzodioxoles/farmacología , Enfermedades Neurodegenerativas/prevención & control , Estrés Oxidativo/efectos de los fármacos , Proteína Desglicasa DJ-1/farmacología , Animales , Benzamidas/metabolismo , Benzodioxoles/metabolismo , Isquemia Encefálica/metabolismo , Isquemia Encefálica/prevención & control , Línea Celular Tumoral , Modelos Animales de Enfermedad , Neuronas Dopaminérgicas/efectos de los fármacos , Neuronas Dopaminérgicas/metabolismo , Humanos , Enfermedades Neurodegenerativas/metabolismo , Fármacos Neuroprotectores/metabolismo , Fármacos Neuroprotectores/farmacología , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/prevención & control , Unión Proteica , Proteína Desglicasa DJ-1/metabolismo , Ratas , Especies Reactivas de Oxígeno/metabolismo
10.
Synapse ; 71(5)2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28165637

RESUMEN

Using an in vitro model of ischemic stroke we treated differentiated SH-SY5Y cells to oxygen-glucose deprivation (OGD) followed by a reperfusion period where normal growth conditions were restored. Cells undergoing OGD exhibited significant cell death as measure by propidium iodide staining. However, cells treated with exogenous extracellular DJ-1 during reperfusion exhibited significant rescue from OGD-induced cell death.


Asunto(s)
Glucosa/deficiencia , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Oxígeno/metabolismo , Proteína Desglicasa DJ-1/farmacología , Hipoxia de la Célula , Línea Celular Tumoral , Humanos , Neuronas/metabolismo , Estrés Oxidativo
11.
Zhejiang Da Xue Xue Bao Yi Xue Ban ; 45(4): 349-355, 2016 05 25.
Artículo en Chino | MEDLINE | ID: mdl-27868407

RESUMEN

Objective: To investigate the effect of silencing DJ-1 on xenografted human laryngeal squamous cell carcinoma (LSCC) Hep-2 cells in nude mice. Methods: Xenograft model of human LSCC was established by subcutaneous transplantation of Hep-2 cells in 24 nude mice. The LSCC-bearing nude mice were randomly divided into 3 groups (n=8 in each):DJ-1 siRNA low dose group and DJ-1 siRNA high dose group were injected in tumors with 20 µg of DJ-1 siRNA or 40 µg of DJ-1 siRNA in 50 µL, respectively; control group was injected with 5% glucose solution in 50 µL, twice a week for 3 weeks. The weight and size of tumors were measured before injection. The animals were sacrificed 48 h after the final treatment, and the tumors were harvested and weighed. The apoptosis and proliferation of tumor cells were determined; the expressions of Caspase-3 and Ki-67 in tumor specimens were detected with immunohistochemistry. The expression of DJ-1, PTEN, survivin mRNA and protein in tumor tissues were detected by RT-PCR and Western blotting, respectively. Results: Tumor weight in low dose group[(0.66±0.15)g] and high dose group[(0.48±0.11)g] were significantly lower than that in control group[(0.83±0.16)g, all P<0.05]. The inhibition rates of low dose group and high dose group were (20.48±0.18)% and (42.16±0.13)%, respectively. Immunohistochemistry showed that the expression of Caspase-3 was increased and Ki-67 was reduced in tumor specimens, compared with the control group (all P<0.05). RT-PCR and Western blot results showed that in low dose group and high dose group the mRNA and protein expression of DJ-1 and survivin significantly decreased (all P<0.05), while PTEN mRNA and protein content increased (all P<0.05). Conclusion: High dose DJ-1 siRNA can inhibit the tumor growth in human LSCC xenograft nude mouse model, which indicates that down-regulating DJ-1 and survivin, and up-regulating PTEN expression may lead to blockage of PI3K-PKB/Akt signaling pathway and promoting tumor cell apoptosis.


Asunto(s)
Carcinoma de Células Escamosas/química , Carcinoma de Células Escamosas/fisiopatología , Línea Celular Tumoral/química , Línea Celular Tumoral/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Neoplasias de Cabeza y Cuello/química , Neoplasias de Cabeza y Cuello/fisiopatología , Neoplasias Laríngeas/química , Neoplasias Laríngeas/fisiopatología , Proteína Desglicasa DJ-1/farmacología , Interferencia de ARN/fisiología , ARN Mensajero/farmacología , ARN Interferente Pequeño/fisiología , Animales , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Apoptosis/genética , Carcinoma de Células Escamosas/genética , Caspasa 3/análisis , Caspasa 3/efectos de los fármacos , Línea Celular Tumoral/fisiología , Línea Celular Tumoral/trasplante , Proliferación Celular/efectos de los fármacos , Regulación hacia Abajo , Regulación de la Expresión Génica/genética , Regulación de la Expresión Génica/fisiología , Neoplasias de Cabeza y Cuello/genética , Xenoinjertos/efectos de los fármacos , Xenoinjertos/fisiología , Humanos , Proteínas Inhibidoras de la Apoptosis/análisis , Proteínas Inhibidoras de la Apoptosis/efectos de los fármacos , Antígeno Ki-67/análisis , Antígeno Ki-67/efectos de los fármacos , Neoplasias Laríngeas/genética , Ratones Desnudos , Fosfohidrolasa PTEN/análisis , Fosfohidrolasa PTEN/efectos de los fármacos , Fosfatidilinositol 3-Quinasas/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Transducción de Señal/fisiología , Carcinoma de Células Escamosas de Cabeza y Cuello
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...