Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
J Virol ; 86(11): 6286-302, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22457526

RESUMEN

We have recently reported that a group of human adenoviruses (HAdVs) uses desmoglein 2 (DSG2) as a receptor for infection. Among these are the widely distributed serotypes HAdV-B3 and HAdV-B7, as well as a newly emerged strain derived from HAdV-B14. These serotypes do not infect rodent cells and could not up until now be studied in small-animal models. We therefore generated transgenic mice containing the human DSG2 locus. These mice expressed human DSG2 (hDSG2) at a level and in a pattern similar to those found for humans and nonhuman primates. As an initial application of hDSG2-transgenic mice, we used a green fluorescent protein (GFP)-expressing HAdV-B3 vector (Ad3-GFP) and studied GFP transgene expression by quantitative reverse transcription-PCR (qRT-PCR) and immunohistochemistry subsequent to intranasal and intravenous virus application. After intranasal application, we found efficient transduction of bronchial and alveolar epithelial cells in hDSG2-transgenic mice. Intravenous Ad3-GFP injection into hDSG2-transgenic mice resulted in hDSG2-dependent transduction of epithelial cells in the intestinal and colon mucosa. Our findings give an explanation for clinical symptoms associated with infection by DSG2-interacting HAdVs and provide a rationale for using Ad3-derived vectors in gene therapy.


Asunto(s)
Infecciones por Adenovirus Humanos/patología , Adenovirus Humanos/patogenicidad , Desmogleína 2/genética , Modelos Animales de Enfermedad , Receptores Virales/genética , Tropismo Viral , Infecciones por Adenovirus Humanos/virología , Animales , Genes Reporteros , Proteínas Fluorescentes Verdes/genética , Humanos , Inmunohistoquímica , Ratones , Ratones Transgénicos , Coloración y Etiquetado/métodos , Transducción Genética
2.
Mol Ther ; 19(3): 479-89, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21081901

RESUMEN

Extracellular matrix (ECM) in solid tumors affects the effectiveness of therapeutics through blocking of intratumoral diffusion and/or physical masking of target receptors on malignant cells. In immunohistochemical studies of tumor sections from breast cancer patients and xenografts, we observed colocalization of ECM proteins and Her2/neu, a tumor-associated antigen that is the target for the widely used monoclonal antibody trastuzumab (Herceptin). We tested whether intratumoral expression of the peptide hormone relaxin (Rlx) would result in ECM degradation and the improvement of trastuzumab therapy. As viral gene delivery into epithelial tumors with extensive tumor ECM is inefficient, we used a hematopoietic stem cell (HSC)-based approach to deliver the Rlx gene to the tumor. In mouse models with syngeneic breast cancer tumors, HSC-mediated intratumoral Rlx expression resulted in a decrease of ECM proteins and enabled control of tumor growth. Moreover, in a model with Her2/neu-positive BT474-M1 tumors and more treatment-refractory tumors derived from HCC1954 cells, we observed a significant delay of tumor growth when trastuzumab therapy was combined with Rlx expression. Our results have implications for antibody therapy of cancer as well as for other anticancer treatment approaches that are based on T-cells or encapsulated chemotherapy drugs.


Asunto(s)
Anticuerpos Monoclonales , Neoplasias de la Mama/terapia , Matriz Extracelular , Receptor ErbB-2 , Animales , Anticuerpos Monoclonales/metabolismo , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales Humanizados , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Matriz Extracelular/metabolismo , Femenino , Orden Génico , Terapia Genética , Vectores Genéticos/genética , Vectores Genéticos/metabolismo , Trasplante de Células Madre Hematopoyéticas , Humanos , Espacio Intracelular/metabolismo , Lentivirus/genética , Lentivirus/metabolismo , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , Unión Proteica , Transporte de Proteínas , Receptor ErbB-2/genética , Receptor ErbB-2/metabolismo , Relaxina/genética , Relaxina/metabolismo , Trastuzumab , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Mol Cancer Res ; 18(2): 278-286, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31704732

RESUMEN

Mechanisms of intrinsic resistance of serous ovarian cancers to standard treatment with carboplatin and paclitaxel are poorly understood. Seventeen primary serous ovarian cancers classified as responders or nonresponders to standard treatment were screened with DigiWest protein array analysis for 279 analytes. Histone methyl transferase EZH2, an interaction partner of ataxia telangiectasia mutated (ATM), was found as one of the most significantly represented proteins in responsive tumors. Survival analysis of 616 patients confirmed a better outcome in patients with high EZH2 expression, but a worse outcome in patients with low EZH2 and high-ATM-expressing tumors compared with patients with low EZH2 and low-ATM-expressing tumors. A proximity ligation assay further confirmed an association between ATM and EZH2 in tumors of patients with an increased disease-free survival. Knockdown of EZH2 resulted in treatment-resistant cells, but suppression of both EZH2 and ATM, or ATM alone, had no effect. DigiWest protein analysis of EZH2-knockdown cells revealed a decrease in proteins involved in mitotic processes and checkpoint regulation, suggesting that deregulated ATM may induce treatment resistance. IMPLICATIONS: Ovarian cancer is a malignancy with high mortality rates, with to date, no successful molecular characterization strategies. Our study uncovers in a comprehensive approach the involvement of checkpoint regulation via ATM and EZH2, potentially providing a new therapeutic perspective for further investigations.


Asunto(s)
Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Carboplatino/farmacología , Cistadenocarcinoma Seroso/tratamiento farmacológico , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Neoplasias Ováricas/tratamiento farmacológico , Paclitaxel/farmacología , Anciano , Proteínas de la Ataxia Telangiectasia Mutada/genética , Cistadenocarcinoma Seroso/genética , Cistadenocarcinoma Seroso/metabolismo , Daño del ADN , ADN de Neoplasias/genética , Resistencia a Antineoplásicos , Proteína Potenciadora del Homólogo Zeste 2/deficiencia , Proteína Potenciadora del Homólogo Zeste 2/genética , Femenino , Humanos , Neoplasias Ováricas/genética , Neoplasias Ováricas/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transducción de Señal
4.
Cancer Biol Ther ; 17(10): 1079-1088, 2016 10 02.
Artículo en Inglés | MEDLINE | ID: mdl-27574825

RESUMEN

Chemotherapy often fails to eradicate cancer stem cells (CSCs) that drive cancer recurrence. In fact, the treated tumors often contain a higher frequency of chemo-resistant CSCs. It is thought that CSC formation is supported by exposure of cancer cells to sub-cytotoxic chemotherapy doses as a result of poor drug penetration in epithelial tumors. We have shown that low-dos cisplatin triggers the transdifferentiation of ovarian cancer cells into CSCs through processes that are also involved in the generation and maintenance of induced pluripotent stem (iPS) cells. Considering similarities between CSCs and iPS cells, we screened a library of 60 synthetic small-molecule compounds, designed to influence EMT/MET signaling in iPS cells on primary ovarian cancer cells. Using a Nanog reporter system we identified a series of compounds capable of blocking the cisplatin triggered formation of CSCs. We then focused on compound GHDM-1515, a drug that acts on pathways that regulate histone demethylases. We demonstrated that co-treatment of primary ovarian cancer cells with GHDM-1515 significantly increased cisplatin induced apoptosis, specifically apoptosis of CSCs. GHDM-1515 inhibited EMT and the cisplatin-induced formation of CSCs. This suggests that GHDM-1515 can sensitize ovarian cancer cells to low-dose cisplatin and potentially enhance the efficacy of cisplatin chemotherapy.


Asunto(s)
Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/patología , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/patología , Pirimidinas/farmacología , Animales , Antineoplásicos/farmacología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Cisplatino/farmacología , Transición Epitelial-Mesenquimal/efectos de los fármacos , Femenino , Humanos , Ratones , Ratones Endogámicos NOD , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Tissue Barriers ; 1(1): e23647, 2013 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-24665377

RESUMEN

Most solid tumors are of epithelial origin and, although malignant cells are de-differentiated, they maintain intercellular junctions, a key feature of epithelial cells, both in the primary tumor as well as in metastatic lesions. These intercellular junctions represent a protective mechanism against attacks by the host's immune system and pose as physical barriers that prevent intratumoral penetration and dissemination of cancer therapeutics. A key protein of epithelial junctions is desmoglein 2 (DSG2). DSG2 is consistently upregulated in all cancers analyzed. Recently, we demonstrated that a group of human adenoviruses (Ad serotypes 3, 7, 11 and 14) use DSG2 as a primary attachment receptor for the infection of cells. We subsequently created a small recombinant protein derived from Ad serotype 3, which binds to DSG2 and triggers transient opening of epithelial intercellular junctions. We named the protein "JO-1" ("junction opener -1"). JO-1 is a small protein that can easily be produced in E. coli. JO-1 binding to and clustering of DSG2 triggers an epithelial-to-mesenchymal-transition that results in transient opening of epithelial junctions. We have shown in over 25 xenograft tumor models that the intravenous injection of JO-1 increased the efficacy of monoclonal and chemotherapy, subsequently reducing the required treatment dose and concomitantly reducing the toxic side effect of these treatments. The application of JO-1 has not been associated with toxicities in safety studies performed in human DSG2-transgenic mice and monkeys.

6.
Front Syst Neurosci ; 6: 54, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22811660

RESUMEN

The role of the histamine H(3) receptor (H(3)R) in anxiety is controversial, due to limitations in drug selectivity and limited validity of behavioral tests used in previous studies. In the present report, we describe two experiments. In the first one, Wistar rats were treated with an H(3)R agonist (methimepip), and exposed to an open-field. In the second one, Balb/c mice were treated with H(3)R agonist (methimepip) or antagonist (JNJ-5207852), and exposed to an open space 3D maze which is a modified version of the radial-arm maze. C57BL/6J saline treated mice were included for comparisons. When exposed to an empty open field, Wistar rats spent more time in the outer area and made very low number of brief crossings in the central area. However, when an object occupied the central area, rats crossed frequently into and spent a long time in the central area. Administration of a range of different doses of methimepip (selective H(3)R agonist) reduced the entries into the central area with a novel object, indicating enhanced avoidance response. In the 3D maze, both Balb/c and C57BL/6J saline-treated mice crossed frequently onto the bridges that radiate from the central platform but only C57BL/6J mice crossed onto the arms which extend the bridges. This suggests that Balb/c mice are more anxious than C57BL/6J mice. Neither methimepip nor JNJ-5207852 (selective H(3)R antagonist/inverse agonist) induced entry into the arms of the maze, indicative of lack of anxiolytic effects.

7.
Clin Cancer Res ; 18(12): 3340-51, 2012 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-22535153

RESUMEN

PURPOSE: Epithelial junctions between tumor cells inhibit the penetration of anticancer drugs into tumors. We previously reported on recombinant adenovirus serotype 3-derived protein (JO-1), which triggers transient opening of intercellular junctions in epithelial tumors through binding to desmoglein 2 (DSG2), and enhances the antitumor effects of several therapeutic monoclonal antibodies. The goal of this study was to evaluate whether JO-1 cotherapy can also improve the efficacy of chemotherapeutic drugs. EXPERIMENTAL DESIGN: The effect of intravenous application of JO-1 in combination with several chemotherapy drugs, including paclitaxel/Taxol, nanoparticle albumin-bound paclitaxel/Abraxane, liposomal doxorubicin/Doxil, and irinotecan/Camptosar, was tested in xenograft models for breast, colon, ovarian, gastric and lung cancer. Because JO-1 does not bind to mouse cells, for safety studies with JO-1, we also used human DSG2 (hDSG2) transgenic mice with tumors that overexpressed hDSG2. RESULTS: JO-1 increased the efficacy of chemotherapeutic drugs, and in several models overcame drug resistance. JO-1 treatment also allowed for the reduction of drug doses required to achieve antitumor effects. Importantly, JO-1 coadmininstration protected normal tissues, including bone marrow and intestinal epithelium, against toxic effects that are normally associated with chemotherapeutic agents. Using the hDSG2-transgenic mouse model, we showed that JO-1 predominantly accumulates in tumors. Except for a mild, transient diarrhea, intravenous injection of JO-1 (2 mg/kg) had no critical side effects on other tissues or hematologic parameters in hDSG2-transgenic mice. CONCLUSIONS: Our preliminary data suggest that JO-1 cotherapy has the potential to improve the therapeutic outcome of cancer chemotherapy.


Asunto(s)
Antineoplásicos/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Uniones Intercelulares/efectos de los fármacos , Proteínas Virales/administración & dosificación , Adenoviridae , Animales , Línea Celular Tumoral , Desmogleína 2/metabolismo , Quimioterapia Combinada , Humanos , Ratones , Ratones SCID , Ratones Transgénicos , Proteínas Recombinantes/administración & dosificación , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Cancer Res ; 71(22): 7080-90, 2011 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-21990319

RESUMEN

The efficacy of monoclonal antibodies (mAb) used to treat solid tumors is limited by intercellular junctions which tightly link epithelial tumor cells to each another. In this study, we define a small, recombinant adenovirus serotype 3-derived protein, termed junction opener 1 (JO-1), which binds to the epithelial junction protein desmoglein 2 (DSG2). In mouse xenograft models employing Her2/neu- and EGFR-positive human cancer cell lines, JO-1 mediated cleavage of DSG2 dimers and activated intracellular signaling pathways which reduced E-cadherin expression in tight junctions. Notably, JO-1-triggered changes allowed for increased intratumoral penetration of the anti-Her2/neu mAb trastuzumab (Herceptin) and improved access to its target receptor, Her2/neu, which is partly trapped in tight junctions. This effect translated directly into increased therapeutic efficacy of trastuzumab in mouse xenograft models using breast, gastric, and ovarian cancer cells that were Her2/neu-positive. Furthermore, combining JO-1 with the EGFR-targeting mAb cetuximab (Erbitux) greatly improved therapeutic outcomes in a metastatic model of EGFR-positive lung cancer. A combination of JO-1 with an approach that triggered transient degradation of tumor stroma proteins elicited eradication of tumors. Taken together, our findings offer preclinical proof of concept to employ JO-1 in combination with mAb therapy.


Asunto(s)
Adenovirus Humanos/fisiología , Anticuerpos Monoclonales Humanizados/uso terapéutico , Anticuerpos Monoclonales/uso terapéutico , Desmogleína 2/fisiología , Neoplasias Experimentales/tratamiento farmacológico , Proteínas Virales/farmacología , Animales , Línea Celular Tumoral , Cetuximab , Femenino , Humanos , Ratones , Neoplasias Experimentales/metabolismo , Proteínas Recombinantes/uso terapéutico , Trastuzumab
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA