Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros

Banco de datos
Tipo de estudio
Tipo del documento
Intervalo de año de publicación
1.
Breast Cancer Res Treat ; 149(1): 99-108, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25519042

RESUMEN

The urokinase receptor (uPAR) plays a critical role in breast cancer (BC) progression and metastases and is a validated target for novel therapies. The current study investigates the effects of MV-uPA, an oncolytic measles virus fully retargeted against uPAR in syngeneic and xenograft BC metastases models. In vitro replication and cytotoxicity of MVs retargeted against human (MV-h-uPA) or mouse (MV-m-uPA) uPAR were assessed in human and murine cancer and non-cancer mammary epithelial cells. The in vivo effects of species-specific uPAR retargeted MVs were assessed in syngeneic and xenograft models of experimental metastases, established by intravenous administration of luciferase expressing 4T1 or MDA-MD-231 cells. Metastases progression was assessed by in vivo bioluminescence imaging. Tumor targeting was evaluated by qRT-PCR of MV-N, rescue of viable viral particles, and immunostaining of MV particles in lungs from tumor bearing mice. In vitro, MV-h-uPA and MV-m-uPA selectively infected, replicated, and induced cytotoxicity in cancer compared to non-cancer cells in a species-specific manner. In vivo, MV-m-uPA delayed 4T1 lung metastases progression and prolonged survival. These effects were associated with identification of viable viral particles, viral RNA, and detection of MV-N by immunostaining from lung tissues in treated mice. In the human MDA-MB-231 metastases model, intravenous administration of MV-h-uPA markedly inhibited metastases progression and significantly improved survival, compared to controls. No significant treatment-related toxicity was observed in treated mice. The above preclinical findings strongly suggest that uPAR retargeted measles virotherapy is a novel and feasible systemic therapy strategy against metastatic breast cancer.


Asunto(s)
Neoplasias de la Mama/genética , Neoplasias Mamarias Experimentales/genética , Virus del Sarampión/genética , Viroterapia Oncolítica , Receptores del Activador de Plasminógeno Tipo Uroquinasa/biosíntesis , Administración Intravenosa , Animales , Neoplasias de la Mama/terapia , Neoplasias de la Mama/virología , Línea Celular Tumoral , Femenino , Humanos , Neoplasias Mamarias Experimentales/terapia , Neoplasias Mamarias Experimentales/virología , Ratones , Metástasis de la Neoplasia , Virus Oncolíticos/genética , Receptores del Activador de Plasminógeno Tipo Uroquinasa/antagonistas & inhibidores , Ensayos Antitumor por Modelo de Xenoinjerto
2.
J Hazard Mater ; 478: 135385, 2024 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-39121733

RESUMEN

The remediation of heavy metal-contaminated soils necessitated a holistic approach that encompassed water and fertilizer conservation alongside soil property restoration. This study introduced the synthesis of (poly)acrylamide-layered double hydroxide gel spheres (PAM-LDH beads), which were designed to simultaneously immobilize heavy metals, control the release of fertilizers, and enhance soil water retention. Laboratory soil experiments under diverse conditions highlighted the superior performance of PAM-LDH beads in the immobilization of hexavalent chromium (Cr(VI)). The layered double hydroxide (LDH) component was identified as the key player in Cr(VI) immobilization, with anion exchange being the predominant mechanism. Notably, the encapsulated urea within the beads was released independently of environmental influences, governed by a concentration gradient across the beads surface. This release process was characterized by an initial phase of absorptive swelling followed by a diffusive phase. The impact on plant growth was assessed, revealing that PAM-LDH beads significantly curtailed Cr(VI) accumulation and alleviated its phytotoxic effects. Changes in the carbon (C) and nitrogen (N) content of the plants suggested that the urea encapsulated within the beads served as a nutrient source, contributing to soil fertility. Moreover, the water-holding capacity and soil-water characteristic curves of PAM-LDH beads suggested that these superabsorbent beads could delay soil water evaporation. The observed shifts in microbial community structure provided evidence for the enhancement of soil carbon and nitrogen cycles, indicative of improved soil properties.

3.
Nutrients ; 16(11)2024 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-38892573

RESUMEN

With the global aging population, addressing prevalent age-related conditions such as osteoporosis and sarcopenia is crucial. Traditional nutritional strategies focusing on single nutrients like calcium, vitamin D, or protein have limitations, prompting a nuanced exploration of the relationship between aging, nutrition, and musculoskeletal health. This cross-sectional study examines the complex interplay between dietary intake of macronutrients, common micronutrients, and water, as well as their association with musculoskeletal health in adults aged 50 to 80 years, using U.S. National Health and Nutrition Examination Survey data (NHANES). Employing multiple linear regression, restricted cubic splines, weighted quantile sum (WQS), and quantile-based g-computation (QGC) regression models, our initial analysis using the WQS model revealed that a one-quartile increase in mixed macronutrient intake was associated with a significant 0.009 unit increase in bone mineral density (BMD) and a 0.670 unit increase in grip strength, while a similar increase in mixed micronutrient intake showed a 0.007 unit increase in BMD and a 0.442 unit increase in grip strength. Our findings highlight the importance of a balanced dietary approach in promoting musculoskeletal health in the elderly, offering holistic strategies for overall well-being.


Asunto(s)
Densidad Ósea , Micronutrientes , Nutrientes , Encuestas Nutricionales , Humanos , Anciano , Micronutrientes/administración & dosificación , Masculino , Femenino , Nutrientes/administración & dosificación , Persona de Mediana Edad , Estudios Transversales , Anciano de 80 o más Años , Densidad Ósea/efectos de los fármacos , Estado Nutricional , Envejecimiento/fisiología , Dieta/métodos , Fuerza de la Mano , Osteoporosis/prevención & control
4.
J Surg Res ; 162(2): 299-307, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19592020

RESUMEN

BACKGROUND: Although evidence has shown that very small electrical currents produce a beneficial therapeutic result for wounds, noninvasive electromagnetic field (EMF) therapy has consisted mostly of anecdotal clinical reports, with very few well-controlled laboratory mechanistic studies. In this study, we evaluate the effects and potential mechanisms of a noninvasive EMF device on skin wound repair. MATERIALS AND METHODS: The effects of noninvasive EMF on keratinocytes and fibroblasts were assessed via proliferation and incisional wound model migration assays. cDNA microarray and RT-PCR were utilized to assess genetic expression changes in keratinocytes after noninvasive EMF treatment. RESULTS: In vitro analyses with human skin keratinocyte cultures demonstrated that noninvasive EMFs have a strong effect on accelerating keratinocyte migration and a relatively weaker effect on promoting keratinocyte proliferation. The positive effects of noninvasive EMFs on cell migration and proliferation seem keratinocyte-specific without such effects seen on dermal fibroblasts. cDNA microarray and RT-PCR performed revealed increased expression of CRK7 and HOXC8 genes in treated keratinocytes. CONCLUSIONS: This study suggests that a noninvasive EMF accelerates wound re-epithelialization through a mechanism of promoting keratinocyte migration and proliferation, possibly due to upregulation of CRK7 and HOXC8 genes.


Asunto(s)
Campos Electromagnéticos , Queratinocitos/citología , Queratinocitos/fisiología , Actinas/genética , Animales , Quemaduras/fisiopatología , Quemaduras/prevención & control , Proteínas de Unión a Calmodulina/genética , División Celular , Movimiento Celular , Fibroblastos/citología , Fibroblastos/fisiología , Prepucio/citología , Prepucio/fisiología , Humanos , Recién Nacido , Masculino , Análisis de Secuencia por Matrices de Oligonucleótidos , ARN Mensajero/genética , ARN Mensajero/aislamiento & purificación , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Piel/lesiones , Porcinos , Cicatrización de Heridas
5.
Cancer Gene Ther ; 27(12): 910-922, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32231231

RESUMEN

The tumor stroma acts as a barrier that limits the efficacy of systemically administered oncolytic viruses (OV). We previously demonstrated that stromal-selective, retargeted oncolytic measles viruses (MVs) delay in vivo tumor progression. To further characterize the contribution of stromal targeting to MV's overall in vivo efficacy in an experimental cancer model, a dual targeted oncolytic measles virus (MV-CD46-muPA) able to simultaneously infect murine stromal (via murine uPAR) and human cancer (via CD46) cells was developed. MV-CD46-muPA infected, replicated, and induced cytotoxicity in both murine and human cancer cells. Viral infection was successfully transferred from stromal to tumor cells in vitro, leading to tumor cell oncolysis. Systemic administration of MV-CD46-muPA led to improved antitumor effects in colon (HT-29) cancer xenografts compared to vehicle or CD46 only targeted MVs. These effects were associated with improved tumor viral deposition, increased apoptosis, and decreases in murine stromal endothelial cells and fibroblasts. MV-CD46-muPA modulated cell cycle, survival, proliferation, and metabolic pathways, as determined by functional proteomic analysis of treated tumors. The above findings further validate the concept that dual stromal and tumor cell viral targeting enhances the therapeutic effects of systemically administered OVs and support further preclinical and clinical development of stromal directed virotherapies.


Asunto(s)
Técnicas In Vitro/métodos , Virus del Sarampión/genética , Virus Oncogénicos/genética , Virus Oncolíticos/genética , Animales , Línea Celular Tumoral , Modelos Animales de Enfermedad , Femenino , Humanos , Ratones
6.
J Biochem ; 141(5): 621-6, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17339230

RESUMEN

c-Abl non-receptor tyrosine kinase has been implicated in many cellular processes including cell differentiation, stress response and regulating gene transcription. The mechanism by which c-Abl is involved in the regulation of gene transcription remains to be elucidated. In this study, we investigated the functions of c-Abl in the activation of p21 promoter. Our results showed that overexpression of c-Abl tyrosine kinase activated p21 promoter and endogenous p21 transcription in U2OS cells. We found that p53 is involved in the activation of p21 promoter by c-Abl, and integrative structure of p53 is required for regulating p21 transcription. In addition, the chromatin immunoprecipitation study demonstrated that c-Abl and p53 can be recruited to the region containing p53 binding site of p21 promoter, and c-Abl increases the DNA binding activity of p53 to the p21 promoter. Furthermore, not only the activation of p21 promoter but also the recruitment to p21 promoter by c-Abl is dependent on the interaction between c-Abl and p53 protein.


Asunto(s)
Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Proteínas Proto-Oncogénicas c-abl/fisiología , Transcripción Genética/fisiología , Proteína p53 Supresora de Tumor/fisiología , Línea Celular Tumoral , Inmunoprecipitación de Cromatina , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Humanos , Regiones Promotoras Genéticas/fisiología
7.
Mol Cancer Res ; 15(10): 1410-1420, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28679779

RESUMEN

The tumor microenvironment (TME) is a relevant target for novel biological therapies. MV-m-uPA and MV-h-uPA are fully retargeted, species-specific, oncolytic measles viruses (MV) directed against murine or human urokinase receptor (PLAUR/uPAR), expressed in tumor and stromal cells. The effects of stromal-selective targeting by uPAR-retargeted MVs were investigated. In vitro infection, virus-induced GFP expression, and cytotoxicity by MV-h-uPA and MV-m-uPA were demonstrated in human and murine cancer cells and cancer-associated fibroblasts in a species-specific manner. In a murine fibroblast/human breast cancer 3D coculture model, selective fibroblast targeting by MV-m-uPA inhibited breast cancer cell growth. Systemic administration of murine-specific MV-m-uPA in mice bearing human MDA-MB-231 xenografts was associated with a significant delay in tumor progression and improved survival compared with controls. Experiments comparing tumor (MV-h-uPA) versus stromal (MV-m-uPA) versus combined virus targeting showed that tumor and stromal targeting was associated with improved tumor control over the other groups. Correlative studies confirmed in vivo viral targeting of tumor stroma by MV-m-uPA, increased apoptosis, and virus-induced differential regulation of murine stromal genes associated with inflammatory, angiogenesis, and survival pathways, as well as indirect regulation of human cancer pathways, indicating viral-induced modulation of tumor-stroma interactions. These data demonstrate the feasibility of stromal-selective targeting by an oncolytic MV, virus-induced modulation of tumor-stroma pathways, and subsequent tumor growth delay. These findings further validate the critical role of stromal uPAR in cancer progression and the potential of oncolytic viruses as antistromal agents.Implications: The current report demonstrates for the first time the biological, in vitro, and in vivo antitumor and molecular effects of stromal selective targeting by an oncolytic virus. Mol Cancer Res; 15(10); 1410-20. ©2017 AACR.


Asunto(s)
Neoplasias de la Mama/terapia , Virus del Sarampión/fisiología , Virus Oncolíticos/fisiología , Receptores del Activador de Plasminógeno Tipo Uroquinasa/genética , Células del Estroma/citología , Animales , Neoplasias de la Mama/genética , Fibroblastos Asociados al Cáncer/citología , Fibroblastos Asociados al Cáncer/virología , Línea Celular Tumoral , Proliferación Celular , Técnicas de Cocultivo , Femenino , Células HT29 , Humanos , Ratones , Viroterapia Oncolítica , Células del Estroma/virología , Microambiente Tumoral
8.
Mol Cancer Ther ; 15(2): 264-75, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26637370

RESUMEN

Interference with endothelial cell metabolism is a promising, yet unexploited strategy for angiogenesis inhibition. We reported that the glucose analogue 2-deoxy-D-glucose (2-DG) inhibits angiogenesis at significantly lower concentrations than those required for tumor cytotoxicity. Here, we found that hypersensitivity to 2-DG in endothelial cells is not associated with enhanced drug uptake compared with tumor cells, but with time-dependent, endothelial-selective inhibition of AKT and ERK phosphorylation. Downregulation of these critical survival pathways is shown to be due to 2-DG's interference with N-linked glycosylation, leading to alterations in VEGFR2 (and downstream signaling) as well as induction of endoplasmic reticulum (ER) stress, GSK3ß activation, and apoptosis. In vivo, periocular administration of 2-DG in LHBETATAG mice was associated with significant reduction of newly formed (CD105(+)) tumor capillaries, ER stress (GRP 78 expression), and endothelial apoptosis (TUNEL). These findings uniquely link N-linked glycosylation inhibition, ER stress, and ERK/AKT downregulation in endothelial cells, and provide a novel drug development strategy to overcome resistance mechanisms to currently available antiangiogenic agents.


Asunto(s)
Inhibidores de la Angiogénesis/administración & dosificación , Desoxiglucosa/administración & dosificación , Estrés del Retículo Endoplásmico/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Glucógeno Sintasa Quinasa 3/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Inhibidores de la Angiogénesis/farmacología , Animales , Apoptosis , Línea Celular Tumoral , Desoxiglucosa/farmacología , Regulación hacia Abajo , Células Endoteliales/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Glucógeno Sintasa Quinasa 3 beta , Glicosilación/efectos de los fármacos , Células HT29 , Células Endoteliales de la Vena Umbilical Humana , Humanos , Ratones , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
9.
Mol Cancer Res ; 10(10): 1271-81, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22912336

RESUMEN

Tumor proteases and inhibitors have been associated with paradoxical effects on tumor progression in preclinical and clinical settings. We previously reported that urokinase (uPA) overexpression delays tumor progression in mammary cancer. This study aimed to determine the role of plasminogen activator inhibitor-1 (PAI-1) on uPA's paradoxical in vivo effects. Using syngeneic murine models, we found that stable uPA overexpression promoted in vivo growth of colon tumors (MC-38) naturally expressing high PAI-1, whereas growth inhibition was observed in renal tumors (RENCA) expressing lower PAI-1 levels. In murine mammary carcinoma (4T1), uPA overexpression shifted the uPA/PAI-1 balance in favor of the protease, resulting in significantly reduced tumor growth and metastases in vivo. Conversely, increased tumor progression was observed in stable PAI-1 overexpressing 4T1 tumors as compared with uPA-overexpressing and control tumors. These effects were associated with downregulation of metastases promoting genes in uPA-overexpressing tumors, such as metalloproteinases, CXCL-1, c-Fos, integrin α-5, VEGF-A, PDGF-α, and IL-1ß. In PAI-1-overexpressing tumors, many of the above genes were upregulated. PAI-1 overexpressing tumors had increased total and new tumor microvessels, and increased tumor cell proliferation, whereas the opposite effects were found in uPA-overexpressing tumors. Finally, PAI-1 downregulation led to significant inhibition of 4T1 tumor growth and metastases in vivo. In conclusion, uPA's dual effects on tumor progression occur in the context of its interactions with endogenous PAI-1 expression. Our studies uncover novel mechanisms of in vivo tumor control by modulation of the balance between tumor proteases and inhibitors, which may be exploited therapeutically.


Asunto(s)
Neoplasias/metabolismo , Neoplasias/patología , Inhibidor 1 de Activador Plasminogénico/metabolismo , Activador de Plasminógeno de Tipo Uroquinasa/metabolismo , Animales , Línea Celular Tumoral , Proliferación Celular , Células Clonales , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Regulación hacia Abajo/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Inmunohistoquímica , Ratones , Ratones Endogámicos BALB C , Metástasis de la Neoplasia , Neoplasias/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , Fenotipo
10.
PLoS One ; 5(10): e13699, 2010 Oct 27.
Artículo en Inglés | MEDLINE | ID: mdl-21060881

RESUMEN

BACKGROUND: During tumor angiogenesis, endothelial cells (ECs) are engaged in a number of energy consuming biological processes, such as proliferation, migration, and capillary formation. Since glucose uptake and metabolism are increased to meet this energy need, the effects of the glycolytic inhibitor 2-deoxy-D-glucose (2-DG) on in vitro and in vivo angiogenesis were investigated. METHODOLOGY/PRINCIPAL FINDINGS: In cell culture, 2-DG inhibited EC growth, induced cytotoxicity, blocked migration, and inhibited actively forming but not established endothelial capillaries. Surprisingly, 2-DG was a better inhibitor of these EC properties than two more efficacious glycolytic inhibitors, 2-fluorodeoxy-D-glucose and oxamate. As an alternative to a glycolytic inhibitory mechanism, we considered 2-DG's ability to interfere with endothelial N-linked glycosylation. 2-DG's effects were reversed by mannose, an N-linked glycosylation precursor, and at relevant concentrations 2-DG also inhibited synthesis of the lipid linked oligosaccharide (LLO) N-glycosylation donor in a mannose-reversible manner. Inhibition of LLO synthesis activated the unfolded protein response (UPR), which resulted in induction of GADD153/CHOP and EC apoptosis (TUNEL assay). Thus, 2-DG's effects on ECs appeared primarily due to inhibition of LLOs synthesis, not glycolysis. 2-DG was then evaluated in two mouse models, inhibiting angiogenesis in both the matrigel plug assay and the LH(BETA)T(AG) transgenic retinoblastoma model. CONCLUSIONS/SIGNIFICANCE: In conclusion, 2-DG inhibits endothelial cell angiogenesis in vitro and in vivo, at concentrations below those affecting tumor cells directly, most likely by interfering with N-linked glycosylation rather than glycolysis. Our data underscore the importance of glucose metabolism on neovascularization, and demonstrate a novel approach for anti-angiogenic strategies.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Desoxiglucosa/farmacología , Animales , Apoptosis , Western Blotting , Inmunohistoquímica , Etiquetado Corte-Fin in Situ , Ratones , Ratones Transgénicos
11.
Cancer Res ; 69(4): 1459-68, 2009 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-19208845

RESUMEN

Oncolytic measles virus (MV) induces cell fusion and cytotoxicity in a CD46-dependent manner. Development of fully retargeted oncolytic MVs would improve tumor selectivity. The urokinase-type plasminogen activator receptor (uPAR) is a tumor and stromal target overexpressed in multiple malignancies. MV-H glycoproteins fully retargeted to either human or murine uPAR were engineered and their fusogenic activity was determined. Recombinant human (MV-h-uPA) and murine (MV-m-uPA) uPAR-retargeted MVs expressing enhanced green fluorescent protein (eGFP) were rescued and characterized. Viral expression of chimeric MV-H was shown by reverse transcription-PCR and Western blot. In vitro viral replication was comparable to MV-GFP control. The receptor and species specificity of MV-uPAs was shown in human and murine cells with different levels of uPAR expression. Removal of the NH(2)-terminal fragment ligand from MV-uPA by factor X(a) treatment ablated the MV-uPA functional activity. Cytotoxicity was shown in uPAR-expressing human and murine cells. MV-h-uPA efficiently infected human endothelial cells and capillary tubes in vitro. I.v. administration of MV-h-uPA delayed tumor growth and prolonged survival in the MDA-MB-231 breast cancer xenograft model. Viral tumor targeting was confirmed by immunohistochemistry. MV-m-uPA transduced murine mammary tumors (4T1) in vivo after intratumor administration. MV-m-uPA targeted murine tumor vasculature after systemic administration, as shown by dual (CD31 and MV-N) staining of tumor capillaries in the MDA-MB-231 model. In conclusion, MV-uPA is a novel oncolytic MV associated with potent and specific antitumor effects and tumor vascular targeting. This is the first retargeted oncolytic MV able to replicate in murine cells and target tumor vasculature in a uPAR-dependent manner.


Asunto(s)
Virus del Sarampión/genética , Viroterapia Oncolítica/métodos , Células 3T3 , Animales , Células CHO , Carcinoma Hepatocelular , Fusión Celular , Línea Celular Tumoral , Chlorocebus aethiops , Neoplasias del Colon , Cricetinae , Cricetulus , Eliminación de Gen , Haplorrinos , Humanos , Neoplasias Hepáticas , Virus del Sarampión/inmunología , Virus del Sarampión/fisiología , Ratones , Virus Oncolíticos/genética , Virus Oncolíticos/inmunología , Virus Oncolíticos/fisiología , ARN Neoplásico/genética , ARN Neoplásico/aislamiento & purificación , Receptores del Activador de Plasminógeno Tipo Uroquinasa/deficiencia , Receptores del Activador de Plasminógeno Tipo Uroquinasa/genética , Células Vero , Proteínas Virales/genética
12.
Arch Biochem Biophys ; 437(2): 199-204, 2005 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-15850559

RESUMEN

c-Abl tyrosine kinase, predominantly distributed in nucleus, has been implicated in many important cellular processes including the regulation of gene transcription. In this study, we showed that c-Abl promoted the transcription of c-fos gene, both exogenously and endogenously. The nuclear localization and tyrosine kinase activity of c-Abl were required for the activation of c-fos gene. c-Abl was associated with RNA polymerase II (RNAP II) in vivo and augmented the tyrosine phosphorylation of the largest subunit of RNAP II. In addition, c-Abl and RNAP II could be recruited to the region of c-fos promoter. The combined results suggest that c-Abl plays an important role in the transcriptional regulation of c-fos gene and the tyrosine phosphorylation of the largest subunit of RNAP II by c-Abl is involved in the regulating process.


Asunto(s)
Regulación de la Expresión Génica , Genes fos/genética , Proteínas Proto-Oncogénicas c-abl/metabolismo , ARN Polimerasa II/metabolismo , Transporte Activo de Núcleo Celular , Línea Celular , Humanos , Células K562 , Mutación/genética , Fosforilación , Fosfotirosina/metabolismo , Regiones Promotoras Genéticas/genética , Unión Proteica , Subunidades de Proteína/química , Subunidades de Proteína/metabolismo , Proteínas Proto-Oncogénicas c-abl/química , Proteínas Proto-Oncogénicas c-abl/genética , ARN Polimerasa II/química , Transcripción Genética/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA