Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 637
Filtrar
1.
Cancer Med ; 10(15): 5218-5234, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34264013

RESUMEN

BACKGROUND: The carcinogenic effect of NUP37 has been reported recently in a variety of tumors, but its research in the field of glioma has not been paid attention. The main purpose of this study is to reveal the relationship between NUP37 and prognosis or clinical characteristics of glioma patients. METHODS: First, as a retrospective study, this study included thousands of tissue samples based on a variety of public databases and clinicopathological tissues. Second, a series of bioinformatics analysis methods were used to analyze the NUP37 and glioma samples from multiple databases such as the CGGA, TCGA, GEO, HPA, and GEPIA. Third, to analyze the relationship between the expression level of NUP37 in tumor tissues and cells and a variety of clinical prognostic molecular characteristics, whether it can be an independent risk factor leading to poor prognosis in glioma and whether it has clinical diagnostic value; GSEA was used to analyze the cancer-related signaling pathways that may be activated by high expression of NUP37. Fifth, CMap was used to analyze small molecule drugs that may inhibit NUP37 expression. Finally, the meta-analysis of thousands of tissue samples from seven datasets and cell proliferation and migration experiments confirmed that NUP37 has a malignant effect on glioma. RESULTS: NUP37 is highly expressed in glioma patient tissues and glioma cells, significantly correlates with reduced overall survival, and may serve as an independent prognostic factor with some diagnostic value. Silencing NUP37 suppresses malignant biological behaviors of glioma cells. 4 small molecule drugs that had potential targeting inhibitory effects on NUP37 overexpression. CONCLUSIONS: This study demonstrates for the first time a malignant role of NUP37 in glioma and provides a vision to unravel the complex pathological mechanisms of glioma and a potentially valuable biomarker for implementing individualized diagnosis and treatment of glioma.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Proliferación Celular/fisiología , Glioma/metabolismo , Glioma/patología , Proteínas de Neoplasias/metabolismo , Proteínas de Complejo Poro Nuclear/metabolismo , Neoplasias Encefálicas/mortalidad , Línea Celular Tumoral , Movimiento Celular , Biología Computacional/métodos , Bases de Datos Factuales/estadística & datos numéricos , Bases de Datos Genéticas , Glioma/mortalidad , Humanos , Proteínas de Neoplasias/efectos de los fármacos , Proteínas de Neoplasias/genética , Proteínas de Complejo Poro Nuclear/efectos de los fármacos , Proteínas de Complejo Poro Nuclear/genética , Pronóstico , Análisis por Matrices de Proteínas , Estudios Retrospectivos , Transducción de Señal
2.
Clin Pharmacol Drug Dev ; 10(11): 1335-1344, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34109764

RESUMEN

This phase 1, 2-part, 2-period, open-label, drug-drug interaction study evaluated the potential for pharmacokinetic interactions between upadacitinib and rosuvastatin, an organic anion transporting polypeptide (OATP) 1B1 and breast cancer resistance protein substrate, or atorvastatin, a cytochrome P450 3A, OATP1B1, and OATP1B3 substrate, in 36 healthy volunteers. During period 1, a single dose of rosuvastatin (5 mg; part 1) or atorvastatin (10 mg; part 2) was administered on day 1, followed by a washout period of 5 days. During period 2, once-daily doses of upadacitinib extended-release (30 mg) were administered on days 1 to 10, and a single dose of rosuvastatin (5 mg; part 1) or atorvastatin (10 mg; part 2) was administered 1 hour after the upadacitinib dose on day 7. Serial blood samples were collected for assays of drug concentrations. In Part 1, rosuvastatin maximum observed plasma concentration (Cmax ) and area under the plasma concentration-time curve from time 0 to infinity (AUCinf ) were 23% and 33% lower, respectively, when administered with upadacitinib relative to when administered alone. In part 2, atorvastatin Cmax and AUCinf was 11% and 23% lower, respectively, when administered with upadacitinib relative to when administered alone. The Cmax and AUCinf of the active metabolite ortho-hydroxyatorvastatin remained unchanged. Administration of a single 5-mg dose of rosuvastatin or a single 10-mg dose of atorvastatin had no relevant effect on upadacitinib Cmax or area under the plasma concentration-time curve. These results demonstrated that upadacitinib has no clinically relevant effect on the pharmacokinetics of rosuvastatin and atorvastatin or on substrates transported by OATP1B or breast cancer resistance protein.


Asunto(s)
Anticolesterolemiantes/farmacocinética , Atorvastatina/farmacocinética , Interacciones Farmacológicas , Compuestos Heterocíclicos con 3 Anillos/farmacología , Inhibidores de las Cinasas Janus/farmacología , Rosuvastatina Cálcica/farmacocinética , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/efectos de los fármacos , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/metabolismo , Adulto , Femenino , Voluntarios Sanos , Humanos , Transportador 1 de Anión Orgánico Específico del Hígado/efectos de los fármacos , Transportador 1 de Anión Orgánico Específico del Hígado/metabolismo , Masculino , Persona de Mediana Edad , Proteínas de Neoplasias/efectos de los fármacos , Proteínas de Neoplasias/metabolismo , Adulto Joven
3.
BMC Cancer ; 21(1): 493, 2021 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-33941107

RESUMEN

BACKGROUND: Glioblastoma (GBM) is the deadliest and the most common primary brain tumor in adults. The invasiveness and proliferation of GBM cells can be decreased through the inhibition of Wnt/ß-catenin pathway. In this regard, celecoxib is a promising agent, but other COXIBs and 2,5-dimethylcelecoxib (2,5-DMC) await elucidation. Thus, the aim of this study was to analyze the impact of celecoxib, 2,5-DMC, etori-, rofe-, and valdecoxib on GBM cell viability and the activity of Wnt/ß-catenin pathway. In addition, the combination of the compounds with temozolomide (TMZ) was also evaluated. Cell cycle distribution and apoptosis, MGMT methylation level, COX-2 and PGE2 EP4 protein levels were also determined in order to better understand the molecular mechanisms exerted by these compounds and to find out which of them can serve best in GBM therapy. METHODS: Celecoxib, 2,5-DMC, etori-, rofe- and valdecoxib were evaluated using three commercially available and two patient-derived GBM cell lines. Cell viability was analyzed using MTT assay, whereas alterations in MGMT methylation level were determined using MS-HRM method. The impact of COXIBs, in the presence and absence of TMZ, on Wnt pathway was measured on the basis of the expression of ß-catenin target genes. Cell cycle distribution and apoptosis analysis were performed using flow cytometry. COX-2 and PGE2 EP4 receptor expression were evaluated using Western blot analysis. RESULTS: Wnt/ß-catenin pathway was attenuated by COXIBs and 2,5-DMC irrespective of the COX-2 expression profile of the treated cells, their MGMT methylation status, or radio/chemoresistance. Celecoxib and 2,5-DMC were the most cytotoxic. Cell cycle distribution was altered, and apoptosis was induced after the treatment with celecoxib, 2,5-DMC, etori- and valdecoxib in T98G cell line. COXIBs and 2,5-DMC did not influence MGMT methylation status, but inhibited COX-2/PGE2/EP4 pathway. CONCLUSIONS: Not only celecoxib, but also 2,5-DMC, etori-, rofe- and valdecoxib should be further investigated as potential good anti-GBM therapeutics.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Inhibidores de la Ciclooxigenasa 2/farmacología , Glioblastoma/metabolismo , Proteínas de Neoplasias/efectos de los fármacos , Pirazoles/farmacología , Sulfonamidas/farmacología , Vía de Señalización Wnt/efectos de los fármacos , Anciano , Antineoplásicos Alquilantes/farmacología , Apoptosis/efectos de los fármacos , Neoplasias Encefálicas/tratamiento farmacológico , Celecoxib/farmacología , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Ciclooxigenasa 2/metabolismo , Metilasas de Modificación del ADN/efectos de los fármacos , Metilasas de Modificación del ADN/metabolismo , Enzimas Reparadoras del ADN/efectos de los fármacos , Enzimas Reparadoras del ADN/metabolismo , Dinoprostona/metabolismo , Relación Dosis-Respuesta a Droga , Etoricoxib/farmacología , Femenino , Glioblastoma/tratamiento farmacológico , Humanos , Isoxazoles/farmacología , Lactonas/farmacología , Masculino , Metilación , Persona de Mediana Edad , Proteínas de Neoplasias/metabolismo , Subtipo EP4 de Receptores de Prostaglandina E/efectos de los fármacos , Subtipo EP4 de Receptores de Prostaglandina E/metabolismo , Sulfonas/farmacología , Temozolomida/farmacología , Proteínas Supresoras de Tumor/efectos de los fármacos , Proteínas Supresoras de Tumor/metabolismo , beta Catenina/efectos de los fármacos , beta Catenina/metabolismo
4.
Nat Rev Drug Discov ; 20(5): 384-405, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33692549

RESUMEN

Gasdermins were recently identified as the mediators of pyroptosis - inflammatory cell death triggered by cytosolic sensing of invasive infection and danger signals. Upon activation, gasdermins form cell membrane pores, which release pro-inflammatory cytokines and alarmins and damage the integrity of the cell membrane. Roles for gasdermins in autoimmune and inflammatory diseases, infectious diseases, deafness and cancer are emerging, revealing potential novel therapeutic avenues. Here, we review current knowledge of the family of gasdermins, focusing on their mechanisms of action and roles in normal physiology and disease. Efforts to develop drugs to modulate gasdermin activity to reduce inflammation or activate more potent immune responses are highlighted.


Asunto(s)
Antiinflamatorios/farmacología , Mediadores de Inflamación/metabolismo , Proteínas de Neoplasias/efectos de los fármacos , Proteínas de Neoplasias/fisiología , Piroptosis/efectos de los fármacos , Piroptosis/fisiología , Animales , Citocinas/metabolismo , Humanos , Inmunidad/efectos de los fármacos
5.
J Chem Neuroanat ; 114: 101942, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33675952

RESUMEN

Methamphetamine (Meth) is a neuro-stimulator substrate which might lead to neural cell death and the activation of several interconnected cellular pathways as well. However, the precise molecular mechanisms underlying Meth-induced neural cell death remained unclear yet. The current study aimed to assess the specific relationship between long-term Meth exposure and several endoplasmic reticulum stress, autophagy, and apoptosis associated markers including C/EBP homologous protein (CHOP), Tribbles homolog 3(Trib3), Nuclear protein 1(NUPR1), and Beclin-1 expression in postmortem human striatum. Therefore, the effects of long-term Meth exposure on autophagy and apoptosis in the striatum of postmortem users were evaluated and molecular, immunehistochemical, and histological examinations were performed on 10 control and 10 Meth-addicted brains. The level of CHOP, Trib3, NUPR1, and Beclin-1, Microtubule-associated proteins 1A/1B light chain 3B(LC3), Caspase 3, and Autophagy protein 5 (ATG5) were measured by using qPCR and immunohistochemistry. Stereological neural cell counting, Hematoxylin and Eosin, Nissl and Tunel staining were also performed. Based on our findings, the expression level of CHOP, Trib3, NUPR1, and Beclin-1 in the striatum of Meth group were significantly higher than the control group. Besides, the neuronal cell death was substantially increased in the striatum based on data obtained from the Tunel assay and the stereological analysis. Long-term presence of Meth in the brain can induce ER stress and overexpression of NUPR1 which is associated with the upregulation of CHOP, a pro-apoptotic transcription factor. Moreover, an increase in Trib3 expression is implicated in CHOP-dependent autophagic cell death during Meth-induced ER stress accompanied by an increase in neuronal cell death in the striatum of the postmortem human brains. Beclin 1 expression was also upregulated which may due to the activation of autophagic mechanisms upon prolonged Meth exposure.


Asunto(s)
Trastornos Relacionados con Anfetaminas/complicaciones , Autofagosomas/efectos de los fármacos , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/efectos de los fármacos , Cuerpo Estriado/efectos de los fármacos , Proteínas de Neoplasias/efectos de los fármacos , Factor de Transcripción CHOP/efectos de los fármacos , Adulto , Apoptosis/efectos de los fármacos , Autopsia , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/biosíntesis , Estimulantes del Sistema Nervioso Central/efectos adversos , Humanos , Masculino , Metanfetamina/efectos adversos , Proteínas de Neoplasias/biosíntesis , Factor de Transcripción CHOP/biosíntesis
6.
J Mol Med (Berl) ; 99(3): 349-358, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33486566

RESUMEN

Breast cancer is currently the most common cancer and the leading cause of cancer death among women worldwide. Advanced breast cancer is prone to metastasis, and there is currently no drug to cure metastatic breast cancer. The purinergic ligand-gated ion channel 7 receptor is an ATP-gated nonselective cation channel receptor and is involved in signal transduction, growth regulation, cytokine secretion, and tumor cell development. Recent studies have shown that upregulation of the P2X7 receptor in breast cancer can mediate AKT signaling pathways, Ca2 þ-activated SK3 potassium channels, and EMT and regulate the secretion of small extracellular vesicles to promote breast cancer invasion and migration, which are affected by factors such as hypoxia and ATP. In addition, studies have shown that microRNAs can bind to the 3' untranslated region of the P2X7 receptor, which affects the occurrence and development of breast cancer by upregulating and downregulating P2X7 receptor expression. Studies have shown that new P2X7 receptor inhibitors, such as emodin and Uncaria tomentosa, can inhibit P2X7 receptor-mediated breast cancer invasion and are expected to be used clinically. This article reviews the research progress on the relationship between the P2X7 receptor and breast cancer to provide new ideas and a basis for clinical diagnosis and treatment.


Asunto(s)
Antineoplásicos/uso terapéutico , Neoplasias de la Mama/metabolismo , Terapia Molecular Dirigida/métodos , Proteínas de Neoplasias/fisiología , Antagonistas del Receptor Purinérgico P2X/uso terapéutico , Receptores Purinérgicos P2X7/fisiología , Adenosina Trifosfato/metabolismo , Antineoplásicos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Uña de Gato , Cationes/metabolismo , Progresión de la Enfermedad , Emodina/uso terapéutico , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Transporte Iónico , Invasividad Neoplásica , Metástasis de la Neoplasia , Proteínas de Neoplasias/efectos de los fármacos , Extractos Vegetales/uso terapéutico , Antagonistas del Receptor Purinérgico P2X/farmacología , Receptores Purinérgicos P2X7/química , Receptores Purinérgicos P2X7/efectos de los fármacos , Transducción de Señal/fisiología , Relación Estructura-Actividad , Regulación hacia Arriba
7.
Invest New Drugs ; 39(2): 458-468, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33475937

RESUMEN

Chemotherapy is usually the subsequent treatment for non-small cell lung cancer patients with acquired radioresistance after long-term fractionated radiotherapy. However, few studies have focused on the selection of chemotherapeutic drugs to treat lung adenocarcinoma patients with radioresistance. Our study compared the sensitivity changes of lung adenocarcinoma cells to conventional chemotherapeutic drugs under radioresistant circumstances by using three lung adenocarcinoma cell models, which were irradiated with fractionated X-rays at a total dose of 60 Gy. The results showed that the toxicities of paclitaxel, docetaxel and SN-38 were increased in radioresistant cells. The IC50 values of docetaxel and SN-38 decreased 0 ~ 3 times and 3 ~ 36 times in radioresistant cells, respectively. Notably, the A549 radioresistant cells were approximately 36 times more sensitive to SN-38 than the parental cells. Further results revealed that the downregulation of the efflux transporter BCRP by long-term fractionated irradiation was an important factor contributing to the increased cytotoxicity of SN-38. In addition, the reported miRNAs and transcriptional factors that regulate BCRP did not participate in the downregulation. In conclusion, these results presented important data on the sensitivity changes of lung adenocarcinoma cells to chemotherapeutic drugs after acquiring radioresistance and suggested that irinotecan (the prodrug of SN-38) might be a promising drug candidate for lung adenocarcinoma patients with acquired radioresistance.


Asunto(s)
Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/efectos de los fármacos , Antineoplásicos/farmacología , Carcinoma de Pulmón de Células no Pequeñas/terapia , Neoplasias Pulmonares/terapia , Proteínas de Neoplasias/efectos de los fármacos , Células A549 , Carcinoma de Pulmón de Células no Pequeñas/patología , Regulación hacia Abajo , Humanos , Concentración 50 Inhibidora , Irinotecán/farmacología , Neoplasias Pulmonares/patología
8.
Mol Oncol ; 15(2): 487-502, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33188726

RESUMEN

Erlotinib is highly effective in lung cancer patients with epidermal growth factor receptor (EGFR) mutations. However, despite initial favorable responses, most patients rapidly develop resistance to erlotinib soon after the initial treatment. This study aims to identify new genes and pathways associated with erlotinib resistance mechanisms in order to develop novel therapeutic strategies. Here, we induced knockout (KO) mutations in erlotinib-resistant human lung cancer cells (NCI-H820) using a genome-scale CRISPR-Cas9 sgRNA library to screen for genes involved in erlotinib susceptibility. The spectrum of sgRNAs incorporated among erlotinib-treated cells was substantially different to that of the untreated cells. Gene set analyses showed a significant depletion of 'cell cycle process' and 'protein ubiquitination pathway' genes among erlotinib-treated cells. Chemical inhibitors targeting genes in these two pathways, such as nutlin-3 and carfilzomib, increased cancer cell death when combined with erlotinib in both in vitro cell line and in vivo patient-derived xenograft experiments. Therefore, we propose that targeting cell cycle processes or protein ubiquitination pathways are promising treatment strategies for overcoming resistance to EGFR inhibitors in lung cancer.


Asunto(s)
Ciclo Celular , Resistencia a Antineoplásicos , Clorhidrato de Erlotinib/farmacología , Neoplasias Pulmonares , Proteínas de Neoplasias , Ubiquitinación , Animales , Sistemas CRISPR-Cas , Línea Celular Tumoral , Resistencia a Antineoplásicos/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Femenino , Estudio de Asociación del Genoma Completo , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Ratones , Ratones Desnudos , Ratones SCID , Proteínas de Neoplasias/efectos de los fármacos , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo
9.
Aging (Albany NY) ; 12(24): 25564-25580, 2020 12 02.
Artículo en Inglés | MEDLINE | ID: mdl-33264103

RESUMEN

The Wnt/ß-catenin pathway plays vital roles in diverse biological processes, including cell differentiation, proliferation, migration, and insulin sensitivity. A recent study reported that the DNA-binding transcriptional factor SIX3 is essential during embryonic development in vertebrates and capable of downregulating target genes of the Wnt/ß-catenin pathway in lung cancer, indicating negative regulation of Wnt/ß-catenin activation. However, regulation of the SIX3-Wnt/ß-catenin pathway axis remains unknown. We measured the expression of TRIM27 and SIX3 as well as investigated whether there was a correlation between them in lung cancer tissue samples. Herein, we found that the E3 ubiquitin ligase, TRIM27, ubiquitinates, and degrades SIX3. TRIM27 induces non-small cell lung cancer (NSCLC) cell proliferation and metastasis, and the expression of ß-catenin, S100P, TGFB3, and MMP-9 were significantly inhibited by SIX3. Furthermore, XAV939 is a selective ß-catenin-mediated transcription inhibitor that inhibited TRIM27- and SIX3-mediated NSCLC cell proliferation, migration, and invasion. Clinically, lung tissue samples of cancer patients showed increased TRIM27 expression and decreased SIX3 expression. Taken together, these data demonstrate that TRIM27 acts as an oncogene regulating cell proliferation and metastasis in NSCLC through SIX3-ß-catenin signaling.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/genética , Proliferación Celular/genética , Proteínas de Unión al ADN/genética , Proteínas del Ojo/genética , Proteínas de Homeodominio/genética , Neoplasias Pulmonares/genética , Proteínas del Tejido Nervioso/genética , Proteínas Nucleares/genética , Células A549 , Proteínas de Unión al Calcio/efectos de los fármacos , Proteínas de Unión al Calcio/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/patología , Movimiento Celular/efectos de los fármacos , Movimiento Celular/genética , Proliferación Celular/efectos de los fármacos , Proteínas de Unión al ADN/metabolismo , Proteínas del Ojo/metabolismo , Femenino , Compuestos Heterocíclicos con 3 Anillos/farmacología , Proteínas de Homeodominio/metabolismo , Humanos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Masculino , Metaloproteinasa 9 de la Matriz/efectos de los fármacos , Metaloproteinasa 9 de la Matriz/metabolismo , Persona de Mediana Edad , Metástasis de la Neoplasia , Proteínas de Neoplasias/efectos de los fármacos , Proteínas de Neoplasias/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Proteínas Nucleares/metabolismo , Oncogenes , Transducción de Señal , Factor de Crecimiento Transformador beta3/efectos de los fármacos , Factor de Crecimiento Transformador beta3/metabolismo , Ubiquitinación/efectos de los fármacos , Ubiquitinación/genética , beta Catenina/efectos de los fármacos , beta Catenina/metabolismo , Proteína Homeobox SIX3
10.
Mol Cell Neurosci ; 109: 103566, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33049367

RESUMEN

Human SH-SY5Y neuroblastoma cells stably expressing exogenous CB1 (CB1XS) or CB2 (CB2XS) receptors were developed to investigate endocannabinoid signaling in the extension of neuronal projections. Expression of cannabinoid receptors did not alter proliferation rate, viability, or apoptosis relative to parental SH-SY5Y. Transcripts for endogenous cannabinoid system enzymes (diacylglycerol lipase, monoacylglycerol lipase, α/ß-hydrolase domain containing proteins 6 and 12, N-acyl phosphatidylethanolamine-phospholipase D, and fatty acid amide hydrolase) were not altered by CB1 or CB2 expression. Endocannabinoid ligands 2-arachidonoylglycerol (2-AG) and anandamide were quantitated in SH-SY5Y cells, and diacylglycerol lipase inhibitor tetrahydrolipstatin decreased 2-AG abundance by 90% but did not alter anandamide abundance. M3 muscarinic agonist oxotremorine M, and inhibitors of monoacylglycerol lipase and α/ß hydrolase domain containing proteins 6 &12 increased 2-AG abundance. CB1 receptor expression increased lengths of short (<30 µm) and long (>30 µm) projections, and this effect was significantly reduced by tetrahydrolipstatin, indicative of stimulation by endogenously produced 2-AG. Pertussis toxin, Gßγ inhibitor gallein, and ß-arrestin inhibitor barbadin did not significantly alter long projection length in CB1XS, but significantly reduced short projections, with gallein having the greatest inhibition. The rho kinase inhibitor Y27632 increased CB1 receptor-mediated long projection extension, indicative of actin cytoskeleton involvement. CB1 receptor expression increased GAP43 and ST8SIA2 mRNA and decreased ITGA1 mRNA, whereas CB2 receptor expression increased NCAM and SYT mRNA. We propose that basal endogenous production of 2-AG provides autocrine stimulation of CB1 receptor signaling through Gi/o, Gßγ, and ß-arrestin mechanisms to promote neuritogenesis, and rho kinase influences process extension.


Asunto(s)
Endocannabinoides/fisiología , Neuritas/ultraestructura , Receptor Cannabinoide CB1/fisiología , Receptor Cannabinoide CB2/fisiología , Citoesqueleto de Actina/ultraestructura , Amidas/farmacología , Apoptosis/efectos de los fármacos , Ácidos Araquidónicos/biosíntesis , Línea Celular Tumoral , Endocannabinoides/biosíntesis , Regulación de la Expresión Génica/efectos de los fármacos , Glicéridos/biosíntesis , Humanos , Lipoproteína Lipasa/antagonistas & inhibidores , Lipoproteína Lipasa/metabolismo , Proteínas de Neoplasias/efectos de los fármacos , Proteínas de Neoplasias/fisiología , Proteínas del Tejido Nervioso/biosíntesis , Proteínas del Tejido Nervioso/genética , Neuroblastoma , Orlistat/farmacología , Oxotremorina/farmacología , Toxina del Pertussis/farmacología , Alcamidas Poliinsaturadas , Piridinas/farmacología , Pirimidinas/farmacología , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Receptor Cannabinoide CB1/efectos de los fármacos , Receptor Cannabinoide CB2/efectos de los fármacos , Proteínas Recombinantes/biosíntesis , Transducción de Señal , Xantenos/farmacología
11.
Clin Pharmacol Drug Dev ; 9(8): 995-1002, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32592633

RESUMEN

Aprocitentan is an investigational, orally active, dual, endothelin receptor antagonist that targets a novel pathway in the treatment of difficult-to-control (resistant) hypertension. The drug-drug interaction potential of aprocitentan on the breast cancer resistance protein (BCRP) transporter substrate rosuvastatin was investigated in this single-center, open-label, single-sequence study. Twenty healthy male subjects received a single dose of 10-mg rosuvastatin on days 1 and 13 followed by pharmacokinetic and tolerability assessments for up to 120 hours. From day 5 to day 17, subjects received 25 mg of aprocitentan once daily. Seventeen of 20 enrolled subjects completed the treatment. At steady state, aprocitentan did not affect the pharmacokinetics of rosuvastatin in a clinically relevant way. The maximum plasma concentration was increased by 40% with a 90% confidence interval of 1.19 to 1.65. However, the ratio of the geometric means for both area under the plasma concentration-time curve from time 0 to time t and area under the plasma concentration-time curve from time 0 to infinity was close to 1 with the 90% confidence interval within a reference interval of 0.80 to 1.25. Adverse events leading to study discontinuation were reported in 2 subjects. Overall, the combination of rosuvastatin and aprocitentan was well tolerated. Based on these data, aprocitentan does not affect BCRP and can be administered concomitantly with drugs dependent on BCRP transport.


Asunto(s)
Anticolesterolemiantes/farmacocinética , Antagonistas de los Receptores de Endotelina/efectos adversos , Pirimidinas/efectos adversos , Rosuvastatina Cálcica/farmacocinética , Sulfonamidas/efectos adversos , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/efectos de los fármacos , Administración Oral , Adulto , Anticolesterolemiantes/administración & dosificación , Anticolesterolemiantes/sangre , Área Bajo la Curva , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Interacciones Farmacológicas/fisiología , Quimioterapia Combinada/efectos adversos , Quimioterapia Combinada/estadística & datos numéricos , Tolerancia a Medicamentos , Antagonistas de los Receptores de Endotelina/administración & dosificación , Antagonistas de los Receptores de Endotelina/uso terapéutico , Voluntarios Sanos/estadística & datos numéricos , Humanos , Hipertensión/tratamiento farmacológico , Masculino , Persona de Mediana Edad , Proteínas de Neoplasias/efectos de los fármacos , Pirimidinas/administración & dosificación , Pirimidinas/uso terapéutico , Rosuvastatina Cálcica/administración & dosificación , Rosuvastatina Cálcica/sangre , Sulfonamidas/administración & dosificación , Sulfonamidas/uso terapéutico
12.
Int J Mol Sci ; 21(9)2020 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-32397371

RESUMEN

Multiple kinase inhibitors are available for patients with advanced hepatocellular carcinoma (HCC). It is largely unknown whether regorafenib or lenvatinib modulates innate immunity including Toll-like receptor (TLR)-signaling pathways in HCC. We performed real-time RT-PCR to investigate 84 TLR-associated gene expression levels and compared these gene expression levels in each hepatoma cells treated with or without regorafenib or lenvatinib. In response to regorafenib, nine and 10 genes were upregulated in Huh7 and HepG2 cells, respectively, and only C-X-C motif chemokine ligand 10 was upregulated in both cell lines. A total of 14 and 12 genes were downregulated in Huh7 and HepG2 cells, respectively, and two genes (Fos proto-oncogene, AP-1 transcription factor subunit, and ubiquitin conjugating enzyme E2 N) were downregulated in both cell lines. In response to lenvatinib, four and 16 genes were upregulated in Huh7 and HepG2 cells, respectively, and two genes (interleukin 1 alpha and TLR4) were upregulated in both cells. Six and one genes were downregulated in Huh7 and HepG2, respectively, and no genes were downregulated in both cell lines. In summary, regorafenib and lenvatinib affect TLR signaling pathways in human hepatoma cell lines. Modulation of TLR signaling pathway may improve the treatment of HCC patients with refractory disease.


Asunto(s)
Antineoplásicos/farmacología , Carcinoma Hepatocelular/patología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Neoplasias Hepáticas/patología , Proteínas de Neoplasias/efectos de los fármacos , Compuestos de Fenilurea/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Piridinas/farmacología , Quinolinas/farmacología , Receptores Toll-Like/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Redes Reguladoras de Genes , Células Hep G2 , Humanos , Inmunidad Innata/efectos de los fármacos , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética , Proto-Oncogenes Mas , Reacción en Cadena en Tiempo Real de la Polimerasa , Transducción de Señal , Sorafenib/farmacología , Transcriptoma/efectos de los fármacos
13.
Int J Nanomedicine ; 15: 2947-2955, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32425526

RESUMEN

INTRODUCTION: ZnO quantum dots (QDs) have drawn much attention recently as they are Cd-free, low-cost, and have excellent optical properties. With the expanded production and application of ZnO nanoparticles, concerns about their potential toxicity have also been raised. MATERIALS AND METHODS: We used RNA sequencing (RNA-seq) to analyze the global gene expression of liver and lung tissues after ZnO QDs treatment. Differentially expressed genes (DEGs) were screened, with a fold change >1.5 and padj <0.05. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes enrichment analyses were performed, and padj <0.05 was considered significantly enriched. The RNA-seq results were validated by quantitative real-time polymerase chain reaction (qRT-PCR). RESULTS: A total of 47 and 218 genes were significantly differentially expressed in the liver and lung. Eight GO terms were enriched in the liver and lung, and retinol metabolism and the peroxisome proliferator-activated receptor (PPAR) signaling pathway were shared in different tissues. DISCUSSION: According to DEGs and pathway enrichment analyses, inflammation might be induced in liver and lung tissues after intravenous injection of ZnO QDs. These findings will be helpful for future research and application of ZnO QDs.


Asunto(s)
Hígado/efectos de los fármacos , Proteínas de Neoplasias/efectos de los fármacos , Proteínas Nucleares/efectos de los fármacos , Puntos Cuánticos/toxicidad , Ubiquitina-Proteína Ligasas/efectos de los fármacos , Óxido de Zinc/toxicidad , Animales , Perfilación de la Expresión Génica/métodos , Ontología de Genes , Hígado/fisiología , Masculino , Ratones , Proteínas de Neoplasias/fisiología , Proteínas Nucleares/fisiología , Reacción en Cadena en Tiempo Real de la Polimerasa , Análisis de Secuencia de ARN , Pruebas de Toxicidad , Ubiquitina-Proteína Ligasas/fisiología
14.
Cancer Discov ; 10(7): 916-921, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32444466

RESUMEN

The mapping of SARS-CoV-2 human protein-protein interactions by Gordon and colleagues revealed druggable targets that are hijacked by the virus. Here, we highlight several oncogenic pathways identified at the host-virus interface of SARS-CoV-2 to enable cancer biologists to apply their knowledge for rapid drug repurposing to treat COVID-19, and help inform the response to potential long-term complications of the disease.


Asunto(s)
Betacoronavirus , Infecciones por Coronavirus/tratamiento farmacológico , Reposicionamiento de Medicamentos , Proteínas de Neoplasias/antagonistas & inhibidores , Neoplasias/tratamiento farmacológico , Neumonía Viral/tratamiento farmacológico , COVID-19 , Ciclo Celular , Infecciones por Coronavirus/genética , Infecciones por Coronavirus/metabolismo , Infecciones por Coronavirus/fisiopatología , Daño del ADN , Epigenómica , Humanos , Proteínas de Neoplasias/efectos de los fármacos , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/fisiopatología , Pandemias , Neumonía Viral/genética , Neumonía Viral/metabolismo , Neumonía Viral/fisiopatología , Biosíntesis de Proteínas , SARS-CoV-2
15.
Toxicology ; 439: 152459, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32278787

RESUMEN

Endocrine disrupting pesticides (EDPs) are exogenous compounds that disrupt endocrine activity. Human exposure to EDPs can occur through occupational contact, and through the consumption of food, milk and water with trace amounts of these pollutants. Several EDPs are epidemiologically linked to breast cancer or are considered as possible carcinogens. However, current evidence is not fully conclusive and their mechanisms of action remain unknown. Thus, the potential interactions between 262 EDPs and 189 proteins associated with breast cancer were evaluated by using a virtual high-throughput screening approach, with AutoDock Vina 1.1.1. The molecular coordinates were previously downloaded from Protein Data Bank and EDCs DataBank, and used for preparation and optimization in Sybyl X-2.0. The best affinity score (-11.0 kcal/mol) was obtained for flucythrinate with the nuclear receptor for vitamin D (VDR). This synthetic pyrethroid, along with other EDPs, such as fluvalinate, bifenthrin, cyhalothrin and cypermethrin, are proposed as multi-target ligands of several proteins related to breast cancer. In addition, the validation of our protocol showed a good accuracy in terms of binding pose prediction and affinity estimation. This study provides a guide to prioritize EDPs for which further in vitro and in vivo analysis could be done to evaluate the risk and possible mechanisms of action of these contaminants and their potential association with breast cancer.


Asunto(s)
Neoplasias de la Mama/metabolismo , Disruptores Endocrinos/química , Disruptores Endocrinos/toxicidad , Proteínas de Neoplasias/efectos de los fármacos , Plaguicidas/química , Plaguicidas/toxicidad , Femenino , Ensayos Analíticos de Alto Rendimiento , Humanos , Modelos Moleculares , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Unión Proteica , Reproducibilidad de los Resultados , Relación Estructura-Actividad
16.
Mol Pharm ; 17(6): 1884-1898, 2020 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-32271581

RESUMEN

We developed a practical synthetic method for fluorine-18 (18F)-labeled pitavastatin ([18F]PTV) as a positron emission tomography (PET) tracer to assess hepatobiliary transporter activity and conducted a PET scan as a preclinical study for proof-of-concept in rats. This method is a one-pot synthesis involving aromatic 18F-fluorination of an arylboronic acid ester followed by deprotection under acidic conditions, which can be reproduced in general clinical sites equipped with a standard radiolabeling system due to the simplified procedure. PET imaging confirmed that intravenously administered [18F]PTV was rapidly accumulated in the liver and gradually transferred into the intestinal lumen through the bile duct. Radiometabolite analysis showed that [18F]PTV was metabolically stable, and 80% of the injected dose was detected as the unchanged form in both blood and bile. We applied integration plot analysis to assess tissue uptake clearance (CLuptake, liver and CLuptake, kidney) and canalicular efflux clearance (CLint, bile), and examined the effects of inhibitors on membrane transport. Treatment with rifampicin, an organic anion transporting polypeptide inhibitor, significantly reduced CLuptake, liver and CLuptake, kidney to 44% and 64% of control, respectively. In contrast, Ko143, a breast cancer resistance protein inhibitor, did not affect CLuptake, liver but significantly reduced CLint, bile to 39% of control without change in [18F]PTV blood concentration. In addition, we found decreased CLuptake, liver and increased CLint, bile in Eisai hyperbilirubinemic rats in response to altered expression levels of transporters. We expect that [18F]PTV can be translated into clinical application, as our synthetic method does not need special apparatus in the radiolabeling system and PET scan with [18F]PTV can quantitatively evaluate transporter activity in vivo.


Asunto(s)
Radioisótopos de Flúor/química , Quinolinas/química , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/efectos de los fármacos , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/metabolismo , Animales , Western Blotting , Cromatografía en Capa Delgada , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Estructura Molecular , Proteínas de Neoplasias/efectos de los fármacos , Proteínas de Neoplasias/metabolismo , Transportadores de Anión Orgánico/efectos de los fármacos , Transportadores de Anión Orgánico/metabolismo , Tomografía de Emisión de Positrones , Ratas , Ratas Sprague-Dawley , Rifampin/química
17.
Theranostics ; 10(8): 3488-3502, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32206103

RESUMEN

Rationale: Whole-genome sequencing has identified many amplified genes in esophageal squamous-cell carcinoma (ESCC). This study investigated the role and clinical relevance of these genes in ESCC. Methods: We collected ESCC and non-tumor esophageal tissues from 225 individuals who underwent surgery. Clinical data were collected and survival time was measured from the date of diagnosis to the date of last follow-up or death. Patient survival was compared with immunohistochemical staining score using Kaplan-Meier methods and hazard ratios were calculated by Cox models. Cells with gene overexpression and knockout were analyzed in proliferation, migration and invasion assays. Cells were also analyzed for levels of intracellular lactate, NADPH, ATP and mRNA and protein expression patterns. Protein levels in cell line and tissue samples were measured by immunoblotting or immunohistochemistry. ESCC cell were grown as xenograft tumors in nude mice. Primary ESCC in genetically engineered mice and patient-derived xenograft mouse models were established for test of therapeutic effects. Results: We show that TP53-induced glycolysis and apoptosis regulator (TIGAR) is a major player in ESCC progression and chemoresistance. TIGAR reprograms glucose metabolism from glycolysis to the glutamine pathway through AMP-activated kinase, and its overexpression is correlated with poor disease outcomes. Tigar knockout mice have reduced ESCC tumor burden and growth rates. Treatment of TIGAR-overexpressing ESCC cell xenografts and patient-derived tumor xenografts in mice with combination of glutaminase inhibitor and chemotherapeutic agents achieves significant more efficacy than chemotherapy alone. Conclusion: These findings shed light on an important role of TIGAR in ESCC and might provide evidence for targeted treatment of TIGAR-overexpressing ESCC.


Asunto(s)
Antineoplásicos/uso terapéutico , Proteínas Reguladoras de la Apoptosis/genética , Neoplasias Esofágicas/tratamiento farmacológico , Carcinoma de Células Escamosas de Esófago/tratamiento farmacológico , Proteínas de Neoplasias/genética , Monoéster Fosfórico Hidrolasas/genética , Animales , Proteínas Reguladoras de la Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular , Progresión de la Enfermedad , Sistemas de Liberación de Medicamentos , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/mortalidad , Carcinoma de Células Escamosas de Esófago/genética , Carcinoma de Células Escamosas de Esófago/mortalidad , Femenino , Glutaminasa/antagonistas & inhibidores , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Proteínas de Neoplasias/efectos de los fármacos , Oncogenes , Monoéster Fosfórico Hidrolasas/efectos de los fármacos , Tasa de Supervivencia , Ensayos Antitumor por Modelo de Xenoinjerto
18.
Am J Respir Cell Mol Biol ; 62(2): 178-190, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31419911

RESUMEN

ATP-binding cassette (ABC) transporters are evolutionarily conserved membrane proteins that pump a variety of endogenous substrates across cell membranes. Certain subfamilies are known to interact with pharmaceutical compounds, potentially influencing drug delivery and treatment efficacy. However, the role of drug resistance-associated ABC transporters has not been examined in idiopathic pulmonary fibrosis (IPF) or its animal model: the bleomycin (BLM)-induced murine model. Here, we investigate the expression of two ABC transporters, P-gp (permeability glycoprotein) and BCRP (breast cancer resistance protein), in human IPF lung tissue and two different BLM-induced mouse models of pulmonary fibrosis. We obtained human IPF specimens from patients during lung transplantation and administered BLM to male C57BL/6J mice either by oropharyngeal aspiration (1 U/kg) or subcutaneous osmotic infusion (100 U/kg over 7 d). We report that P-gp and BCRP expression in lungs of patients with IPF was comparable to controls. However, murine lungs expressed increased levels of P-gp and BCRP after oropharyngeal and subcutaneous BLM administration. We localized this upregulation to multiple pulmonary cell types, including alveolar fibroblasts, endothelial cells, and type 2 epithelial cells. Functionally, this effect reduced murine lung exposure to nintedanib, a U.S. Food and Drug Administration-approved IPF therapy known to be a P-gp substrate. The study reveals a discrepancy between IPF pathophysiology and the common animal model of lung fibrosis. BLM-induced drug efflux in the murine lungs may present an uncontrolled confounding variable in the preclinical study of IPF drug candidates, and these findings will facilitate disease model validation and enhance new drug discoveries that will ultimately improve patient outcomes.


Asunto(s)
Bleomicina/farmacología , Células Endoteliales/efectos de los fármacos , Fibroblastos/efectos de los fármacos , Fibrosis Pulmonar Idiopática/tratamiento farmacológico , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/efectos de los fármacos , Animales , Modelos Animales de Enfermedad , Células Endoteliales/metabolismo , Femenino , Fibroblastos/metabolismo , Humanos , Fibrosis Pulmonar Idiopática/metabolismo , Masculino , Ratones Endogámicos C57BL , Proteínas de Neoplasias/efectos de los fármacos , Proteínas de Neoplasias/metabolismo
19.
Anatol J Cardiol ; 22(5): 240-249, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31674929

RESUMEN

OBJECTIVE: Endocan, chemerin, and galectin-3 are discrete biomarkers associated with cardiovascular diseases and acting through different pathophysiological pathways. The aim of this study is to investigate and compare the effects of high doses of atorvastatin and rosuvastatin on serum endocan, chemerin, and galectin-3 levels in patients with acute myocardial infarction (AMI). METHODS: Sixty-three patients with AMI were randomized to receive atorvastatin (80 mg/day) or rosuvastatin (40 mg/day) after percutaneous revascularization. Serum levels of endocan, chemerin, and galectin-3 were evaluated at baseline and after 4-week therapy. RESULTS: Endocan levels were not decreased statistically significantly with atorvastatin 80 mg, but rosuvastatin 40 mg markedly decreased the levels of endocan according to baseline [from 110.27 (86.03-143.69) pg/mL to 99.22 (78.30-122.87) pg/mL with atorvastatin 80 mg and from 110.73 (77.28-165.22) pg/mL to 93.40 (70.48-115.13) pg/mL with rosuvastatin 40 mg, p=0.242 for atorvastatin 80 mg and p=0.014 for rosuvastatin 40 mg]. Chemerin levels significantly decreased in both groups according to baseline [from 264.90 (196.00-525.95) ng/mL to 135.00 (105.95-225.65) ng/mL with atorvastatin 80 mg and from 309.95 (168.87-701.27) ng/mL to 121.25 (86.60-212.65) ng/mL with rosuvastatin 40 mg, p<0.001, respectively, for both groups]. Galectin-3 levels did not change markedly with atorvastatin 80 mg, but they decreased with rosuvastatin 40 mg [from 17.00 (13.10-22.25) ng/mL to 19.30 (15.25-23.45) ng/mL with atorvastatin 80 mg, p=0.721, and from 18.25 (12.82-23.82) ng/mL to 16.60 (10.60-20.15) ng/mL with rosuvastatin 40 mg, p=0.074]. There were no significant between-group differences in terms of absolute and percentage changes of endocan, chemerin, and galectin-3 at 4 weeks. CONCLUSION: We reported that both statins similarly decreased the endocan levels, whereas rosuvastatin seems to have more prominent effects on the reduction of the chemerin and galectin-3 levels in patients with AMI.


Asunto(s)
Atorvastatina/farmacología , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Infarto del Miocardio/sangre , Rosuvastatina Cálcica/farmacología , Angioplastia , Biomarcadores/sangre , Proteínas Sanguíneas , Quimiocinas/sangre , Quimiocinas/efectos de los fármacos , Femenino , Galectina 3/sangre , Galectina 3/efectos de los fármacos , Galectinas , Humanos , Masculino , Persona de Mediana Edad , Infarto del Miocardio/terapia , Proteínas de Neoplasias/sangre , Proteínas de Neoplasias/efectos de los fármacos , Proteoglicanos/sangre , Proteoglicanos/efectos de los fármacos , Resultado del Tratamiento
20.
J Proteomics ; 209: 103491, 2019 10 30.
Artículo en Inglés | MEDLINE | ID: mdl-31472280

RESUMEN

Neuroblastoma, a cancer of the sympathetic nervous system, is the second most common pediatric cancer. A unique feature of neuroblastoma is remission in some patients due to spontaneous differentiation of metastatic tumors. 13-cis retinoic acid (13-cis RA) is currently used in the clinic to treat neuroblastoma due to its differentiation inducing effects. In this study, we used shotgun proteomics to identify proteins affected by 13-cis RA treatment in neuroblastoma SK-N-SH cells. Our results showed that 13-cis RA reduced proteins involved in extracellular matrix synthesis and organization and increased proteins involved in cell adhesion and neurofilament formation. These changes indicate that 13-cis RA induces tumor cell differentiation by decreasing extracellular matrix rigidity and increasing neurite overgrowth. Differentially-affected proteins identified in this study may be novel biomarkers of drug efficacy in the treatment of neuroblastoma. SIGNIFICANCE: As neuroblastoma can spontaneously differentiate, determining which proteins are involved in differentiation can guide development of novel treatments. 13-cis retinoic acid is currently used in the clinic as a differentiation inducer. Here we have established a proteome map of SK-N-SH cells treated with 13-cis retinoic acid. Bioinformatic analysis revealed the involvement of development, differentiation, extracellular matrix assembly, collagen biosynthesis, and neurofilament bundle association. This proteome map provides information as to which proteins are important for differentiation and identifies networks that can be targeted by drugs to treat neuroblastoma [1].


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Isotretinoína/farmacología , Proteínas de Neoplasias/análisis , Neuroblastoma/tratamiento farmacológico , Proteómica/métodos , Línea Celular Tumoral , Matriz Extracelular/efectos de los fármacos , Humanos , Isotretinoína/uso terapéutico , Proteínas de Neoplasias/efectos de los fármacos , Neuritas/efectos de los fármacos , Neuroblastoma/patología , Proteoma/análisis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...