Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Curr Protoc ; 2(3): e394, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-35316583

RESUMEN

Forward genetics begins with a biological phenotype and attempts to identify genetic changes that influence that phenotype. These changes can be induced in a selected group of genes, for instance, by using libraries of cDNAs, shRNAs, CRISPR guide RNAs, or genetic suppressor elements (GSEs), or randomly throughout the genome using chemical or insertional mutagens, with each approach creating distinct genetic changes. The Validation-Based Insertional Mutagenesis (VBIM) strategy utilizes modified lentiviruses as insertional mutagens, placing strong promoters throughout the genome. Generating libraries with millions of cells carrying one or a few VBIM promoter insertions is straightforward, allowing selection of cells in which overexpression of VBIM-driven RNAs or proteins promote the phenotype of interest. VBIM-driven RNAs may encode full-length proteins, truncated proteins (which may have wild-type, constitutive, or dominant-negative activity), or antisense RNAs that can disrupt gene expression. The diversity in VBIM-driven changes allows for the identification of both gain-of-function and loss-of-function mutations in a single screen. Additionally, VBIM can target any genomic locus, regardless of whether it is expressed in the cells under study or known to have a biological function, allowing for true whole-genome screens without the complication and cost of constructing, maintaining, and delivering a comprehensive library. Here, we review the VBIM strategy and discuss examples in which VBIM has been successfully used in diverse screens to identify novel genes or novel functions for known genes. In addition, we discuss considerations for transitioning the VBIM strategy to in vivo screens. We hope that other laboratories will be encouraged to use the VBIM strategy to identify genes that influence their phenotypes of interest. © 2022 Wiley Periodicals LLC.


Asunto(s)
Pruebas Genéticas , Lentivirus , Lentivirus/genética , Mutagénesis Insercional , Mutación , Fenotipo
2.
Proc Natl Acad Sci U S A ; 118(47)2021 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-34795058

RESUMEN

High expression of programmed death-ligand 1 (PD-L1) in cancer cells drives immune-independent, cell-intrinsic functions, leading to resistance to DNA-damaging therapies. We find that high expression of the ubiquitin E3 ligase FBXO22 sensitizes nonsmall cell lung cancer (NSCLC) cells to ionizing radiation (IR) and cisplatin, and that activation of FBXO22 by phosphorylation is necessary for this function. Importantly, FBXO22 activates PD-L1 ubiquitination and degradation, which in turn increases the sensitivity of NSCLC cells to DNA damage. Cyclin-dependent kinase 5 (CDK5), aberrantly active in cancer cells, plays a crucial role in increasing the expression of PD-L1 in medulloblastoma [R. D. Dorand et al, Science 353, 399-403 (2016)]. We show in NSCLC cells that inhibiting CDK5 or reducing its expression increases the level of FBXO22, decreases that of PD-L1, and increases the sensitivity of the cells to DNA damage. We conclude that FBXO22 is a substrate of CDK5, and that inhibiting CDK5 reduces PD-L1 indirectly by increasing FBXO22. Pairing inhibitors of CDK5 with immune checkpoint inhibitors may increase the efficacy of immune checkpoint blockade alone or in combination with DNA-damaging therapies.


Asunto(s)
Antígeno B7-H1/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Proteínas F-Box/metabolismo , Neoplasias Pulmonares/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Células A549 , Antígeno B7-H1/genética , Carcinoma de Pulmón de Células no Pequeñas/genética , Quinasa 5 Dependiente de la Ciclina , Daño del ADN , Proteínas F-Box/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Pulmonares/genética , Meduloblastoma , Fosforilación , Receptores Citoplasmáticos y Nucleares/genética , Ubiquitina-Proteína Ligasas/genética , Ubiquitinación , Ubiquitinas/metabolismo
3.
Br J Cancer ; 124(5): 893-895, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33257843

RESUMEN

Small cell lung cancer (SCLC) is characterised by high relapse rates. Tumour-initiating cells (TICs) are responsible for drug resistance and recurrence of cancer. Rovalpituzumab tesirine (Rova-T), a potent humanised antibody-drug conjugate, selectively targets delta-like protein 3, which is highly expressed in SCLC TICs. The experimental drug CBL0137 (CBL) inhibits the histone chaperone FACT (facilitates chromatin transcription), which is required for the expression of transcription factors that are essential for TIC maintenance. Rova-T and CBL each target SCLC TICs as single agents. However, acquired or intrinsic resistance to single agents is a major problem in cancer. Therefore, we investigated the potential effect of combining Rova-T and CBL in SCLC to eradicate TICs more effectively. Our preclinical studies report a novel and highly translatable therapeutic strategy of dual targeting TICs using Rova-T in combination with CBL to potentially increase survival of SCLC patients.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Neoplasias Pulmonares/tratamiento farmacológico , Carcinoma Pulmonar de Células Pequeñas/tratamiento farmacológico , Animales , Anticuerpos Monoclonales Humanizados/administración & dosificación , Apoptosis , Benzodiazepinonas/administración & dosificación , Carbazoles/administración & dosificación , Proliferación Celular , Resistencia a Antineoplásicos , Humanos , Inmunoconjugados/administración & dosificación , Neoplasias Pulmonares/patología , Ratones , Ratones Endogámicos NOD , Ratones SCID , Pronóstico , Carcinoma Pulmonar de Células Pequeñas/patología , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Cancer Res ; 78(9): 2396-2406, 2018 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-29440145

RESUMEN

Traditional treatments of small-cell lung cancer (SCLC) with cisplatin, a standard-of-care therapy, spare the tumor-initiating cells (TIC) that mediate drug resistance. Here we report a novel therapeutic strategy that preferentially targets TICs in SCLC, in which cisplatin is combined with CBL0137, an inhibitor of the histone chaperone facilitates chromatin transcription (FACT), which is highly expressed in TICs. Combination of cisplatin and CBL0137 killed patient-derived and murine SCLC cell lines synergistically. In response to CBL0137 alone, TICs were more sensitive than non-TICs, in part, because CBL0137 increased expression of the tumor suppressor NOTCH1 by abrogating the binding of negative regulator SP3 to the NOTCH1 promoter, and in part because treatment decreased the high expression of stem cell transcription factors. The combination of cisplatin and CBL0137 greatly reduced the growth of a patient-derived xenograft in mice and also the growth of a syngeneic mouse SCLC tumor. Thus, CBL0137 can be a highly effective drug against SCLC, especially in combination with cisplatin.Significance: These findings reveal a novel therapeutic regimen for SCLC, combining cisplatin with an inhibitor that preferentially targets tumor-initiating cells. Cancer Res; 78(9); 2396-406. ©2018 AACR.


Asunto(s)
Carbazoles/farmacología , Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas del Grupo de Alta Movilidad/antagonistas & inhibidores , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Células Madre Neoplásicas/metabolismo , Receptor Notch1/genética , Carcinoma Pulmonar de Células Pequeñas/genética , Carcinoma Pulmonar de Células Pequeñas/patología , Factores de Elongación Transcripcional/antagonistas & inhibidores , Animales , Línea Celular Tumoral , Autorrenovación de las Células/efectos de los fármacos , Autorrenovación de las Células/genética , Supervivencia Celular/efectos de los fármacos , Cisplatino/farmacología , Modelos Animales de Enfermedad , Sinergismo Farmacológico , Humanos , Células Madre Neoplásicas/patología , Receptor Notch1/metabolismo , Transducción de Señal/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Proc Natl Acad Sci U S A ; 112(31): 9680-5, 2015 Aug 04.
Artículo en Inglés | MEDLINE | ID: mdl-26195767

RESUMEN

Several components of the canonical pathway of response to lipopolysaccharide (LPS) are required for the EGF-dependent activation of NFκB. Conversely, the ability of Toll-like Receptor 4 (TLR4) to activate NFκB in response to LPS is impaired by down regulating EGF receptor (EGFR) expression or by using the EGFR inhibitor erlotinib. The LYN proto-oncogene (LYN) is required for signaling in both directions. LYN binds to the EGFR upon LPS stimulation, and erlotinib impairs this association. In mice, erlotinib blocks the LPS-induced expression of tumor necrosis factor α (TNFα) and interleukin-6 (IL-6) and ameliorates LPS-induced endotoxity, revealing that EGFR is essential for LPS-induced signaling in vivo.


Asunto(s)
Receptores ErbB/metabolismo , Lipopolisacáridos/toxicidad , Sustancias Protectoras/farmacología , Quinazolinas/farmacología , Transducción de Señal/efectos de los fármacos , Receptor Toll-Like 4/metabolismo , Animales , Línea Celular , Citocinas/metabolismo , Factor de Crecimiento Epidérmico/farmacología , Clorhidrato de Erlotinib , Silenciador del Gen/efectos de los fármacos , Humanos , Quinasas Quinasa Quinasa PAM/metabolismo , Ratones , Factor 88 de Diferenciación Mieloide/metabolismo , FN-kappa B/metabolismo , Fosforilación/efectos de los fármacos , Proto-Oncogenes Mas , Factor de Necrosis Tumoral alfa/farmacología , Familia-src Quinasas/metabolismo
6.
Mol Cancer Ther ; 13(9): 2203-14, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25028470

RESUMEN

Erlotinib is a tyrosine kinase inhibitor approved for the treatment of patients with advanced non-small cell lung cancer (NSCLC). In these patients, erlotinib prolongs survival but its benefit remains modest because many tumors express wild-type (wt) EGFR or develop a second-site EGFR mutation. To test drug combinations that could improve the efficacy of erlotinib, we combined erlotinib with quinacrine, which inhibits the FACT (facilitates chromatin transcription) complex that is required for NF-κB transcriptional activity. In A549 (wtEGFR), H1975 (EGFR-L858R/T790M), and H1993 (MET amplification) NSCLC cells, this drug combination was highly synergistic, as quantified by Chou-Talalay combination indices, and slowed xenograft tumor growth. At a sub-IC50 but more clinically attainable concentration of erlotinib, quinacrine, alone or in combination with erlotinib, significantly inhibited colony formation and induced cell-cycle arrest and apoptosis. Quinacrine decreased the level of active FACT subunit SSRP1 and suppressed NF-κB-dependent luciferase activity. Knockdown of SSRP1 decreased cell growth and sensitized cells to erlotinib. Moreover, transcriptomic profiling showed that quinacrine or combination treatment significantly affected cell-cycle-related genes that contain binding sites for transcription factors that regulate SSRP1 target genes. As potential biomarkers of drug combination efficacy, we identified genes that were more strongly suppressed by the combination than by single treatment, and whose increased expression predicted poorer survival in patients with lung adenocarcinoma. This preclinical study shows that quinacrine overcomes erlotinib resistance by inhibiting FACT and cell-cycle progression, and supports a clinical trial testing erlotinib alone versus this combination in advanced NSCLC.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Proteínas de Unión al ADN/metabolismo , Resistencia a Antineoplásicos , Proteínas del Grupo de Alta Movilidad/metabolismo , Neoplasias Pulmonares/tratamiento farmacológico , Subunidad p50 de NF-kappa B/metabolismo , Quinacrina/química , Quinazolinas/farmacología , Factores de Elongación Transcripcional/metabolismo , Animales , Antineoplásicos/química , Apoptosis , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , ADN/química , Clorhidrato de Erlotinib , Regulación Neoplásica de la Expresión Génica , Humanos , Luciferasas/metabolismo , Neoplasias Pulmonares/metabolismo , Ratones , Ratones Desnudos , ARN Interferente Pequeño/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Proc Natl Acad Sci U S A ; 111(32): 11721-6, 2014 Aug 12.
Artículo en Inglés | MEDLINE | ID: mdl-25071181

RESUMEN

Activation of nuclear factor κB (NFκB) is a central event in the responses of normal cells to inflammatory signals, and the abnormal constitutive activation of NFκB is important for the survival of most cancer cells. In nonmalignant human cells, EGF stimulates robust activation of NFκB. The kinase activity of the EGF receptor (EGFR) is required, because the potent and specific inhibitor erlotinib blocks the response. Down-regulating EGFR expression or inhibiting EGFR with erlotinib impairs constitutive NFκB activation in several different types of cancer cells and, conversely, increased activation of NFκB leads to erlotinib resistance in these cells. We conclude that EGF is an important mediator of NFκB activation in cancer cells. To explore the mechanism, we selected an erlotinib-resistant cell line in which the guanine nucleotide exchange factor Son of Sevenless 1 (SOS1), well known to be important for EGF-dependent signaling to MAP kinases, is overexpressed. Increased expression of SOS1 increases NFκB activation in several different types of cancer cells, and ablation of SOS1 inhibits EGF-induced NFκB activation in these cells, indicating that SOS1 is a functional component of the pathway connecting EGFR to NFκB activation. Importantly, the guanine nucleotide exchange activity of SOS1 is not required for NFκB activation.


Asunto(s)
Receptores ErbB/metabolismo , FN-kappa B/metabolismo , Neoplasias/metabolismo , Proteína SOS1/metabolismo , Mama/citología , Mama/metabolismo , Línea Celular Tumoral , Células Cultivadas , Resistencia a Antineoplásicos , Factor de Crecimiento Epidérmico/metabolismo , Células Epiteliales/metabolismo , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/genética , Clorhidrato de Erlotinib , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Inhibidores de Proteínas Quinasas/farmacología , Quinazolinas/farmacología , Proteína SOS1/genética , Transducción de Señal/efectos de los fármacos , Regulación hacia Arriba
8.
Breast Cancer Res Treat ; 131(3): 849-58, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21479552

RESUMEN

Breast cancer is a genetically heterogenous disease with subtypes differing in prognosis and chemosensitivity. The basal-like breast cancer (BLBC) molecular subtype is associated with poorer outcomes, but is more responsive to taxane-based chemotherapy. Kinesins are intracellular transport proteins that interact with microtubules, which are also the mechanistic target for taxanes. We investigated the relationship between taxane resistance in BLBC and kinesins using both expression and functional studies. Kinesin (KIF) expression was evaluated in three settings in relation to taxane resistance: (i) the NCI-60 cancer cell lines, (ii) pre-treatment samples from four BLBC patient cohorts receiving neoadjuvant chemotherapy regimens with and without taxanes, and (iii) post-treatment samples from residual breast cancer following neoadjuvant taxane-containing chemotherapy. We used a novel functional approach to gene modification, validation-based insertional mutagenesis, to select kinesin-overexpressing clones of BLBC cells for evaluation of related mechanisms of taxane resistance. In the NCI-60 cell line dataset, overexpression of the kinesin KIFC3 is significantly correlated with resistance to both docetaxel (P < 0.001) and paclitaxel (P < 0.001), but not to platinum-based chemotherapy, including carboplatin (P = 0.49) and cisplatin (P = 0.10). Overexpression of KIFC3 and KIF5A in pre-chemotherapy samples similarly predicted resistance to paclitaxel in the MDACC cohorts (P = 0.01); no KIF predicted resistance to fluorouracil-epirubicin-cyclophosphamide or cisplatin in BLBC patient cohorts treated without taxanes. KIF12 is the most overexpressed KIF gene in post-chemotherapy taxane-resistant residual breast cancers (2.8-fold-change). Functional studies established that overexpression of KIFC3, KIF5A, and KIF12 were specific in mediating resistance to docetaxel and not vincristine or doxorubicin. Mutation of the ATP-binding domain of a kinesin abolished its ability to mediate docetaxel resistance. Overall, kinesin overexpression correlates with specific taxane resistance in BLBC cell lines and tissues. Our results suggest a novel approach for drug development to overcome taxane resistance in breast cancer through concurrent or sequential use of kinesin inhibitors.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias de la Mama/genética , Hidrocarburos Aromáticos con Puentes/farmacología , Resistencia a Antineoplásicos/genética , Cinesinas/genética , Taxoides/farmacología , Adenosina Trifosfato/metabolismo , Adulto , Anciano , Antraciclinas/farmacología , Antineoplásicos/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Hidrocarburos Aromáticos con Puentes/uso terapéutico , Línea Celular Tumoral , Análisis por Conglomerados , Docetaxel , Femenino , Expresión Génica , Perfilación de la Expresión Génica , Células HEK293 , Humanos , Cinesinas/metabolismo , Persona de Mediana Edad , Estadificación de Neoplasias , Paclitaxel/farmacología , Unión Proteica , Taxoides/uso terapéutico , Vincristina/farmacología
9.
Cancer Res ; 69(20): 8035-42, 2009 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-19789344

RESUMEN

Resistance to chemotherapy remains a major barrier to the successful treatment of cancer. To understand mechanisms underlying docetaxel resistance in breast cancer, we used an insertional mutagenesis strategy to identify proteins whose overexpression confers resistance. A strong promoter was inserted approximately randomly into the genomes of tumor-derived breast cancer cells, using a novel lentiviral vector. We isolated a docetaxel-resistant clone in which the level of the kinesin KIFC3 was elevated. When KIFC3 or the additional kinesins KIFC1, KIF1A, or KIF5A were overexpressed in the breast cancer cell lines MDA-MB231 and MDA-MB 468, the cells became more resistant to docetaxel. The binding of kinesins to microtubules opposes the stabilizing effect of docetaxel that prevents cytokinesis and leads to apoptosis. Our finding that kinesins can mediate docetaxel resistance might lead to novel therapeutic approaches in which kinesin inhibitors are paired with taxanes.


Asunto(s)
Antineoplásicos/uso terapéutico , Neoplasias de la Mama/metabolismo , Resistencia a Antineoplásicos , Cinesinas/metabolismo , Taxoides/uso terapéutico , Apoptosis/efectos de los fármacos , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Docetaxel , Perfilación de la Expresión Génica , Humanos , Immunoblotting , Cinesinas/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Tubulina (Proteína)/metabolismo , Células Tumorales Cultivadas
11.
Lung India ; 25(4): 152-4, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21264082

RESUMEN

A 47 year old female who had past history of incomplete treatment for pulmonary tuberculosis presented with increased breathlessness, generalized swelling and loss of ap-petite for last one month. X-ray chest PA view showed bilateral fibrocalcific opacities with blunting of costophrenic angle on both sides. She underwent bronchoscopy to collect bronchial wash to rule out relapse of tuberculosis. On bronchoscopy a loose broncholith with sharp and speculated margins were detected in right middle lobe bronchus. This broncholith was successfully removed through flexible bronchoscope without any complications.

12.
Indian J Crit Care Med ; 12(4): 194-7, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19742266

RESUMEN

Tracheal stenosis following prolonged intubation is a relatively rare but a serious problem. However, some degree of airway injury is common following intubation, no matter whether it is prolonged or of short duration. Here, we are reporting a fifty six year old male patient who developed multiple web like tracheal stenosis following intubation with high volume low pressure cuff endotracheal tube. Subsequently, the stenosis was successfully dilated by balloon bronchoplasty.

13.
Proc Natl Acad Sci U S A ; 102(21): 7589-94, 2005 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-15897451

RESUMEN

Cross-talk between the major angiogenic growth factor, VEGF, and integrin cell adhesion receptors has emerged recently as a critical factor in the regulation of angiogenesis and tumor development. However, the molecular mechanisms and consequences of this intercommunication remain unclear. Here, we define a mechanism whereby integrin alpha v beta3, through activation, clustering, and signaling by means of p66 Shc (Src homology 2 domain containing), regulates the production of VEGF in tumor cells expressing this integrin. Tumors with "activatable" but not "inactive" beta3 integrin secrete high levels of VEGF, which in turn promotes extensive neovascularization and augments tumor growth in vivo. This stimulation of VEGF expression depends upon the ability of alpha v beta3 integrin to cluster and promote phosphorylation of p66 Shc. These observations identify a link between beta3 integrins and VEGF in tumor growth and angiogenesis and, therefore, may influence anti-integrin as well as anti-VEGF therapeutic strategies.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Integrina alfaVbeta3/metabolismo , Neoplasias/metabolismo , Neovascularización Patológica/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Línea Celular Tumoral , Movimiento Celular/fisiología , Ensayo de Inmunoadsorción Enzimática , Humanos , Immunoblotting , Inmunoprecipitación , Integrina beta3/genética , Imagen por Resonancia Magnética , Masculino , Ratones , Ratones Endogámicos , Mutación/genética , Fosforilación , Plásmidos/genética , Proteínas Adaptadoras de la Señalización Shc , Proteína Transformadora 1 que Contiene Dominios de Homología 2 de Src
14.
Cell Commun Adhes ; 11(1): 1-11, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15500293

RESUMEN

Mechanisms of metastasis, the major complication of prostate cancer, are poorly understood. In this study, we define molecular mechanisms that may contribute to the highly invasive potential of prostate cancer cells. Vascular endothelial growth factor (VEGF), its receptors (VEGFRs), and alpha5beta1 integrin were expressed by prostate cancer cells in vitro and by prostate tumors in vivo, and their expression was elevated at sites of bone metastasis compared to original prostate tumor. VEGF, through interaction with its receptors, regulated adhesive and migratory properties of the cancer cells. Specifically, the highly metastatic prostate cancer cell subline LNCaP-C4-2 showed a decreased adhesive but an enhanced migratory response to fibronectin, a ligand for alpha5beta1 integrin, compared to its nonmetastatic counterpart. A similar pattern was also observed when bone sialoprotein was used as a ligand in migration assays. Increased migration of metastatic prostate cancer cells to fibronectin and bone sialoprotein was regulated by VEGF via VEGFR-2. Tumor suppressor PTEN was involved in control of VEGF/VEGFR-2 stimulated prostate cancer cell adhesion as well as proliferation.


Asunto(s)
Metástasis de la Neoplasia/fisiopatología , Neoplasias de la Próstata/fisiopatología , Factor A de Crecimiento Endotelial Vascular/fisiología , Neoplasias Óseas/secundario , Adhesión Celular/efectos de los fármacos , Adhesión Celular/fisiología , División Celular/efectos de los fármacos , División Celular/fisiología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Movimiento Celular/fisiología , Fibronectinas/fisiología , Regulación Neoplásica de la Expresión Génica , Humanos , Inmunohistoquímica/métodos , Integrina alfa5beta1/análisis , Integrina alfa5beta1/genética , Integrina alfa5beta1/fisiología , Masculino , Microscopía Fluorescente/métodos , Fosfohidrolasa PTEN , Monoéster Fosfórico Hidrolasas/farmacología , Monoéster Fosfórico Hidrolasas/fisiología , Neoplasias de la Próstata/genética , Receptores de Factores de Crecimiento Endotelial Vascular/análisis , Receptores de Factores de Crecimiento Endotelial Vascular/genética , Receptores de Factores de Crecimiento Endotelial Vascular/fisiología , Proteínas Supresoras de Tumor/farmacología , Proteínas Supresoras de Tumor/fisiología , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/farmacología , Receptor 2 de Factores de Crecimiento Endotelial Vascular/análisis , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/fisiología
15.
Blood ; 104(6): 1703-10, 2004 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-15105289

RESUMEN

We investigated the role of Akt-1, one of the major downstream effectors of phosphoinositide 3-kinase (PI3K), in platelet function using mice in which the gene for Akt-1 had been inactivated. Using ex vivo techniques, we showed that Akt-1-deficient mice exhibited impaired platelet aggregation and spreading in response to various agonists. These differences were most apparent in platelets activated with low concentrations of thrombin. Although Akt-1 is not the predominant Akt isoform in mouse platelets, its absence diminished the amount of total phospho-Akt and inhibited increases in intracellular Ca(2+) concentration in response to thrombin. Moreover, thrombin-induced platelet alpha-granule release as well as release of adenosine triphosphate from dense granules was also defective in Akt-1-null platelets. Although the absence of Akt-1 did not influence expression of the major platelet receptors for thrombin and collagen, fibrinogen binding in response to these agonists was significantly reduced. As a consequence of impaired alpha(IIb)beta(3) activation and platelet aggregation, Akt-1 null mice showed significantly longer bleeding times than wild-type mice.


Asunto(s)
Plaquetas/efectos de los fármacos , Colágeno/farmacología , Proteínas Serina-Treonina Quinasas/deficiencia , Proteínas Proto-Oncogénicas/deficiencia , Trombina/farmacología , Adenosina Difosfato/farmacología , Adenosina Trifosfato/metabolismo , Animales , Tiempo de Sangría , Plaquetas/citología , Plaquetas/metabolismo , Calcio/metabolismo , Fibrinógeno/metabolismo , Integrina beta3/metabolismo , Ratones , Ratones Noqueados , Selectina-P/metabolismo , Forbol 12,13-Dibutirato/farmacología , Fosforilación , Adhesividad Plaquetaria/efectos de los fármacos , Agregación Plaquetaria/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/genética , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas c-akt
16.
J Biol Chem ; 278(40): 39044-50, 2003 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-12885781

RESUMEN

The molecular mechanism leading to the cancer metastasis to bone is poorly understood but yet determines prognosis and therapy. Here, we define a new molecular pathway that may account for the extraordinarily high osteotropism of prostate cancer. By using SPARC (secreted protein, acidic and rich in cysteine)-deficient mice and recombinant SPARC, we demonstrated that SPARC selectively supports the migration of highly metastatic relative to less metastatic prostate cancer cell lines to bone. Increased migration to SPARC can be traced to the activation of integrins alphaVbeta3 and alphaVbeta5 on tumor cells. Such activation is induced by an autocrine vascular endothelial growth factor (VEGF)/VEGF receptor (VEGFR)-2 loop on the tumor cells, which also supports the growth and proliferation of prostate cancer cells. A consequence of SPARC recognition by alphaVbeta5 is enhanced VEGF production. Thus, prostate cancer cells expressing VEGF/VEGFR-2 will activate alphaVbeta3 and alphaVbeta5 on their surface and use these integrins to migrate toward SPARC in bone. Within the bone environment, SPARC engagement of these integrins will stimulate growth of the tumor and further production of VEGF to support neoangiogenesis, thereby favoring the development of the metastatic tumor. Supporting this model, activated integrins were found to colocalize with VEGFR-2 in tissue samples of metastatic prostate tumors from patients.


Asunto(s)
Neoplasias Óseas/patología , Neoplasias Óseas/secundario , Metástasis de la Neoplasia , Osteonectina/genética , Agar/farmacología , Animales , Huesos , División Celular , Movimiento Celular , Citometría de Flujo , Humanos , Inmunohistoquímica , Integrina alfaVbeta3/metabolismo , Integrinas/metabolismo , Masculino , Ratones , Modelos Biológicos , Trasplante de Neoplasias , Osteonectina/metabolismo , Neoplasias de la Próstata/patología , Receptores de Vitronectina/metabolismo , Proteínas Recombinantes/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Tumorales Cultivadas , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
17.
Teratog Carcinog Mutagen ; Suppl 1: 71-8, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-12616598

RESUMEN

Many natural agents including fruits and vegetables are known to provide protection from different degenerative diseases including cancer, by preventing damage to the cellular components. The effect of two important dietary agents, alpha tocopherol, and the flavonoid quercetin, along with two commonly consumed vegetables, bitter gourd and tomato, were investigated on spontaneous and dimethylbenz(a)anthracene (DMBA)-induced DNA damage in murine lymphocytes in vitro. DNA damage was determined by single cell gel electrophoresis (comet assay). The rationale for such an approach for this study is that DNA damage can lead to genetic disorders that occur at different stages of carcinogenesis and protection from such damages may in the long run help to prevent development of cancer. Both alpha tocopherol and quercetin as single agents were found to be potent inhibitors of DNA damage (spontaneous and carcinogen induced) in a dose-dependent manner. Fresh juices of bitter gourd and tomato could also protect from DMBA-induced DNA damage but not as effectively as the single agents. The anticarcinogenic role of nutrients as well as non-nutrient dietary components need to be explored more extensively. The Comet assay is a simple, fast, and reliable method to determine the protective effect against DNA damage, one of the prerequisites for carcinogenesis.


Asunto(s)
9,10-Dimetil-1,2-benzantraceno/antagonistas & inhibidores , Antineoplásicos/farmacología , Carcinógenos/antagonistas & inhibidores , Carcinógenos/farmacología , Ensayo Cometa/métodos , Daño del ADN , Dieta , Linfocitos/efectos de los fármacos , Linfocitos/metabolismo , Quercetina/farmacología , alfa-Tocoferol/farmacología , 9,10-Dimetil-1,2-benzantraceno/farmacología , Animales , Antineoplásicos/metabolismo , Separación Celular , Daño del ADN/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Solanum lycopersicum/química , Linfocitos/química , Ratones , Momordica charantia/química , Quercetina/metabolismo , alfa-Tocoferol/metabolismo
18.
Asian Pac J Cancer Prev ; 2(1): 43-47, 2001.
Artículo en Inglés | MEDLINE | ID: mdl-12718653

RESUMEN

The anticarcinogenic effect of tomato juice, a natural source of antioxidants and other chemopreventive / antimutagenic agents, was studied in a skin carcinogenesis model in mice. The possible mode of action was also investigated. Oral administration of tomato juice afforded protection from development of skin tumour and increased life expectancy which may be attributed to the combined action of a number of chemical compounds with cancer chemopreventive properties present in tomato. The protective role may be associated with a decreased level of lipid peroxides noted in the tomato treated group and modulation of host detoxification enzymes. Exposure to the carcinogen resulted in a depression of the liver enzymes- glutathione-S-transferase (GST), glutathione peroxidase (GPx) and superoxide dismutase (SOD). Oral administration of tomato juice resulted in significant activation of all these enzymes (p<0.001). These results suggest a preventive role of tomato juice during carcinogenesis which is mediated possibly by their modulatory effects on biotransformation enzymes and the detoxification system of the host.

19.
Asian Pac J Cancer Prev ; 1(3): 203-206, 2000.
Artículo en Inglés | MEDLINE | ID: mdl-12718665

RESUMEN

The protective role of two commonly consumed natural dietary items- bitter gourd and tomato against endogenous as well as 7,12- dimethylbenz(a)anthracene (DMBA) induced lipid peroxidation in the livers of mice was investigated. The rationale for such an approach is that lipid peroxidation has been suggested to play a key role in human cancer development. There was a sharp rise in lipid peroxidation (measured as thiobarbituric acid reactive substances formation) during skin carcinogenesis induced by DMBA in mice. Aqueous extracts of bitter gourd and tomato juice were found to be very potent inhibitors of lipid peroxidation both in normal and DMBA treated mice. Our observations support the hypothesis that natural combinations of phytochemicals present in the fruit juices exert cancer-protective effects via a decrease in lipid peroxidation.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...