Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
Antiviral Res ; 77(2): 120-7, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18164447

RESUMEN

Antioxidant molecules can be used both to replenish the depletion of reduced glutathione (GSH) occurring during HIV infection, and to inhibit HIV replication. The purpose of this work was to assess the efficacy of two pro-GSH molecules able to cross the cell membrane more easily than GSH. We used an experimental animal model consisting of C57BL/6 mice infected with the LP-BM5 viral complex; the treatments were based on the intramuscular administration of I-152, a pro-drug of N-acetylcysteine and S-acetyl-beta-mercaptoethylamine, and S-acetylglutathione, an acetylated GSH derivative. The results show that I-152, at a concentration of 10.7 times lower than GSH, caused a reduction in lymph node and spleen weights of about 55% when compared to infected animals and an inhibition of about 66% in spleen and lymph node virus content. S-acetylglutathione, at half the concentration of GSH, caused a reduction in lymph node weight of about 17% and in spleen and lymph node virus content of about 70% and 30%, respectively. These results show that the administration of pro-GSH molecules may favorably substitute for the use of GSH as such.


Asunto(s)
Acetilcisteína/análogos & derivados , Fármacos Anti-VIH/uso terapéutico , Cisteamina/análogos & derivados , Glutatión/análogos & derivados , Síndrome de Inmunodeficiencia Adquirida del Murino/tratamiento farmacológico , Profármacos/uso terapéutico , Acetilcisteína/farmacología , Acetilcisteína/uso terapéutico , Animales , Fármacos Anti-VIH/farmacología , Proliferación Celular/efectos de los fármacos , Cisteamina/farmacología , Cisteamina/uso terapéutico , ADN Viral/efectos de los fármacos , ADN Viral/genética , Modelos Animales de Enfermedad , Femenino , Glutatión/farmacología , Glutatión/uso terapéutico , Hipergammaglobulinemia/tratamiento farmacológico , Inmunoglobulina G/sangre , Virus de la Leucemia Murina/efectos de los fármacos , Virus de la Leucemia Murina/genética , Virus de la Leucemia Murina/aislamiento & purificación , Ganglios Linfáticos/efectos de los fármacos , Ganglios Linfáticos/fisiopatología , Linfocitos/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Tamaño de los Órganos/efectos de los fármacos , Reacción en Cadena de la Polimerasa , Profármacos/farmacología , Bazo/efectos de los fármacos , Bazo/fisiopatología
2.
Curr Med Chem ; 13(15): 1749-55, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16787218

RESUMEN

Reduced glutathione (GSH) is present in millimolar concentrations in mammalian cells. It is involved in many cellular functions such as detoxification, amino acid transport, production of coenzymes, and the recycling of vitamins E and C. GSH acts as a redox buffer to preserve the reduced intracellular environment. Decreased glutathione levels have been found in numerous diseases such as cancer, viral infections, and immune dysfunctions. Many antioxidant molecules, such as GSH and N-acetylcysteine (NAC), have been demonstrated to inhibit in vitro and in vivo viral replication through different mechanisms of action. Accumulating evidence suggests that intracellular GSH levels in antigen-presenting cells such as macrophages, influence the Th1/Th2 cytokine response pattern, and more precisely, GSH depletion inhibits Th1-associated cytokine production and/or favours Th2 associated responses. It is known that GSH is not transported to most cells and tissues in a free form. Therefore, a number of different approaches have been developed in the last years to circumvent this problem. This review discusses the capacity of some new molecules with potent pro-GSH effects either to exert significant antiviral activity or to augment GSH intracellular content in macrophages to generate and maintain the appropriate Th1/Th2 balance. The observations reported herein show that pro-GSH molecules represent new therapeutic agents to treat antiviral infections and Th2-mediated diseases such as allergic disorders and AIDS.


Asunto(s)
Adyuvantes Inmunológicos/farmacología , Antivirales/farmacología , Glutatión/farmacología , Animales , Glutatión/fisiología , Humanos , Ratones , Células TH1/inmunología , Células Th2/inmunología , Virosis/fisiopatología
3.
Cell Mol Life Sci ; 61(16): 2100-12, 2004 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15316659

RESUMEN

To identify a model for the study of intestinal pathogenesis of severe acute respiratory syndrome (SARS) we tested the sensitivity of six human intestinal epithelial cell lines to infection with SARS coronavirus (SARS-CoV). In permissive cell lines, effects of SARS-CoV on cellular gene expression were analysed using high-density oligonucleotide arrays. Caco-2 and CL-14 cell lines were found to be highly permissive to SARS-CoV, due to the presence of angiotensin-converting enzyme 2 as a functional receptor. In both cell lines, SARS-CoV infection deregulated expression of cellular genes which may be important for the intestinal pathogenesis of SARS.


Asunto(s)
Células Epiteliales/virología , Intestinos/virología , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/patogenicidad , Células CACO-2 , Supervivencia Celular , Células Epiteliales/metabolismo , Células Epiteliales/ultraestructura , Expresión Génica , Humanos , Mucosa Intestinal/metabolismo , Intestinos/ultraestructura , Microscopía Electrónica , Modelos Biológicos , Análisis de Secuencia por Matrices de Oligonucleótidos , Receptores Virales/metabolismo , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/crecimiento & desarrollo , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/metabolismo
4.
Cell Mol Life Sci ; 61(11): 1307-16, 2004 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15170509

RESUMEN

Recent pathological findings reveal a higher frequency of human cytomegalovirus (HCMV) in tumor cells from different tumors compared with surrounding tissues. Experimental investigations suggest possible supportive effects of HCMV for tumor development and progression. One HCMV effect on tumor cells is the inhibition of apoptosis, leading to the promotion of tumor cell survival. Decreased sensitivity to treatment-induced tumor cell death is a major reason for failure of anticancer chemotherapy. HCMV infection interferes with both the intrinsic and extrinsic cellular apoptosis pathways. HCMV promotes cell survival signaling influencing the tumor suppressor p53 and its relative p73, and stimulates the antiapoptotic Ras/Raf/MEK/Erk- and PI-3K-signaling pathways. Antiapoptotic effects mediated by HCMV are inhibited by antiviral treatment in cell culture. Therefore, a better understanding of the influence of HCMV infection on tumor cell apoptosis might translate into improved anti-cancer therapy.


Asunto(s)
Apoptosis , Infecciones por Citomegalovirus/patología , Neoplasias/virología , Humanos , Neoplasias/etiología , Neoplasias/patología , Transducción de Señal
5.
Antiviral Res ; 55(1): 179-88, 2002 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12076762

RESUMEN

Antiviral activity of the metal chelator ethylenediaminedisuccinic acid (EDDS) was examined in vitro against human cytomegalovirus (HCMV) wild type strains and strains that are resistant against ganciclovir (GCV) and cidofovir (HPMPC). EDDS inhibited the replication of wild-type as well as GCV- and HPMPC-resistant strains with a 50% effective concentration of 7.4-12 microg/ml. At concentrations of 100 microg/ml EDDS, unlike GCV or HPMPC, suppressed HCMV-induced up-regulation of intercellular adhesion molecule-1 (ICAM-1) and reduced T-cell adhesion to HCMV-infected cells in a monolayer adhesion model. In vitro EDDS inhibited murine cytomegalovirus (MCMV) replication (EC50 8.6 microg/ml) and caused in mice some protection against MCMV induced mortality at a non-toxic dose. Since immunopathological factors may play a significant role in HCMV disease it will be of interest to further study whether EDDS is effective in terms of modulation of inflammatory responses to HCMV infections.


Asunto(s)
Adyuvantes Inmunológicos/farmacología , Antivirales/farmacología , Quelantes/farmacología , Citomegalovirus/efectos de los fármacos , Citosina/análogos & derivados , Etilenodiaminas/farmacología , Organofosfonatos , Succinatos/farmacología , Animales , Adhesión Celular/efectos de los fármacos , Línea Celular , Cidofovir , Citomegalovirus/inmunología , Citomegalovirus/fisiología , Citosina/farmacología , Farmacorresistencia Microbiana , Infecciones por Herpesviridae/tratamiento farmacológico , Infecciones por Herpesviridae/mortalidad , Humanos , Molécula 1 de Adhesión Intercelular/metabolismo , Células Jurkat , Ratones , Ratones SCID , Muromegalovirus/efectos de los fármacos , Compuestos Organofosforados/farmacología , Replicación Viral/efectos de los fármacos
6.
J Virol Methods ; 101(1-2): 149-57, 2002 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-11849693

RESUMEN

The COBAS Amplicor CMV Monitor test (Roche Diagnostics), an automated polymerase chain reaction (PCR) assay for the quantification of cytomegalovirus (CMV) DNA in plasma samples, was evaluated in a routine diagnostic laboratory. Using cell culture-derived CMV and CMV-negative human plasma, the linear detection range of the assay as well as its intra-and inter-assay variabilities were assessed. The study design allowed distinguishing variations in results related to amplification and detection from those caused by differences in the efficiency of DNA extraction. The assay was able to identify the majority of samples correctly as positive with CMV DNA concentrations above the limit of detection. However, the reported values were often twofold or more different from the (theoretical) input, which could be explained partly by inefficient DNA extraction. The following values were computed for the coefficients of determination R(2): inter-assay variability excluding DNA extraction, R(2)=0.982; including DNA extraction, R(2)=0.977; intra-assay variability excluding DNA extraction, R(2)=0.992; including DNA extraction, R(2)=0.992. On balance, the test has acceptable within-run and between-run reproducibility. It therefore allows the comparison of results obtained at different time-points as well as in different laboratories, e.g. in multi-centre studies.


Asunto(s)
Infecciones por Citomegalovirus/diagnóstico , Citomegalovirus/aislamiento & purificación , ADN Viral/sangre , Reacción en Cadena de la Polimerasa/métodos , Automatización , Células Cultivadas , Citomegalovirus/genética , Pruebas Diagnósticas de Rutina , Humanos , Reproducibilidad de los Resultados , Carga Viral
7.
Artículo en Inglés | MEDLINE | ID: mdl-11563036

RESUMEN

ISIS 2922, but not ganciclovir (GCV), inhibits HCMV immediate early protein (IE) expression in different infected cell lines and prevents down-modulation of extracellular matrix proteins thrombospondin-1 and -2 induced by IE proteins. While action of ISIS 2922 is mainly due to specific inhibition of IE 2 mRNA, there is also evidence for unspecific effects in terms of inhibition of virus adhesion and penetration.


Asunto(s)
Antígenos Virales/biosíntesis , Antivirales/farmacología , Proteínas Inmediatas-Precoces/biosíntesis , Oligonucleótidos Antisentido/farmacología , Tionucleótidos/farmacología , Trombospondina 1/biosíntesis , Trombospondinas/biosíntesis , Humanos , Proteínas Inmediatas-Precoces/antagonistas & inhibidores , Trombospondina 1/antagonistas & inhibidores , Trombospondinas/antagonistas & inhibidores
8.
J Immunol ; 167(4): 1900-8, 2001 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-11489969

RESUMEN

The human CMV (HCMV) is a persistent virus that may cause severe inflammatory responses especially in immunocompromised hosts. In different cell types, HCMV infection leads to the activation of the pleiotropic transcription factor, NF-kappaB, which triggers virus replication but also propagates cell-mediated inflammatory mechanisms that largely depend on PG synthesis. We investigated the interactions of HCMV and the NF-kappaB-dependent PG synthesis pathway in cultures of retinal pigment epithelial (RPE) cells that are known to be infected in HCMV retinitis patients. Unlike in other cell types, HCMV increased neither NF-kappaB activity nor p65 and p105/50 mRNA levels in RPE cells. Both TNF-alpha and phorbol ester 12,0-tetradecanoylphorbol 13-acetate (TPA) enhanced NF-kappaB activity but only TPA increased HCMV replication. Cyclooxygenase-2 expression and PGE2 release was increased by TPA and TNF-alpha but not by HCMV infection. Stimulatory activity of TPA on HCMV replication was suppressed by protein kinase C inhibitors and inhibitors of p42/44 and p38 mitogen-activated protein kinases but not by NF-kappaB inhibitors. In conclusion, HCMV circumvents the NF-kappaB route in favor of the protein kinase C-dependent mitogen-activated protein kinase pathway in RPE cells. This virus/host cell interaction might be a mechanism that promotes HCMV persistence in immune-privileged organs such as the eye.


Asunto(s)
Citomegalovirus/fisiología , FN-kappa B/fisiología , Epitelio Pigmentado Ocular/metabolismo , Epitelio Pigmentado Ocular/virología , Células Cultivadas , Ciclooxigenasa 2 , Citomegalovirus/efectos de los fármacos , Dinoprostona/metabolismo , Humanos , Isoenzimas/biosíntesis , Isoenzimas/genética , Proteínas de la Membrana , Proteínas Quinasas Activadas por Mitógenos/fisiología , FN-kappa B/genética , FN-kappa B/metabolismo , Subunidad p50 de NF-kappa B , Epitelio Pigmentado Ocular/efectos de los fármacos , Epitelio Pigmentado Ocular/enzimología , Prostaglandina-Endoperóxido Sintasas/biosíntesis , Prostaglandina-Endoperóxido Sintasas/genética , Proteína Quinasa C/fisiología , ARN Mensajero/metabolismo , Acetato de Tetradecanoilforbol/farmacología , Factor de Transcripción ReIA , Transcripción Genética , Factor de Necrosis Tumoral alfa/farmacología , Regulación hacia Arriba , Replicación Viral/efectos de los fármacos
9.
Anticancer Res ; 21(2A): 1035-42, 2001.
Artículo en Inglés | MEDLINE | ID: mdl-11396137

RESUMEN

BACKGROUND: Anaplastic thyroid carcinoma is an aggressive solid tumor that fails to adequately respond to any known chemotherapeutic regimen. The development of effective chemotherapy agents would provide the best chance for long-term survival of patients. MATERIALS AND METHODS: The cytotoxic effects of bovine seminal ribonuclease (BS-RNase) against thyroid carcinoma cell lines with different degrees of differentiation in comparison to non-malignant cells, including human foreskin fibroblasts (HFF) and retinal pigment epithelial cells (RPE), were tested using the MTT dye reduction assay. Induction of apoptosis was demonstrated by annexin V assay and expression of proteins related to apoptosis was investigated by flow cytometry. The antitumoral in vivo effects of BS-RNase were assessed on established xenografts of anaplastic thyroid carcinoma cell line 8505C in nude mice using subcutaneous injections of BS-RNase (12.5 mg/kg once a day, on 20 consecutive days). RESULTS: All the tumor cell lines exhibited marked sensitivity against BS-RNase in comparison to HFF and RPE cells. The greatest growth inhibition was seen in the 8505C line, while IC50 values for papillary (B-CPAP) and poorly-differentiated thyroid carcinoma cells were about 6-fold higher. The cytotoxic action of BS-RNase was associated with induction of apoptosis. Expressions of Fas and Fas-ligand were not influenced by BS-RNase completely, while the down-regulation of Bcl-2 in treated cells was observed. In vivo treatment induced significant tumor regression after the course of 20 consecutive days. No apparent toxic effects of BS-RNase toward non-malignant cells were observed during the in vivo treatment. After cessation of therapy (day 20) tumor volume continued to decrease and the tumor was no longer detectable after 30 days of treatment induction in all animals. CONCLUSION: BS-RNase may have beneficial effects for treatment of aggressive anaplastic thyroid cancer.


Asunto(s)
Antineoplásicos/uso terapéutico , Endorribonucleasas/uso terapéutico , Neoplasias de la Tiroides/tratamiento farmacológico , Animales , Antineoplásicos/farmacología , Apoptosis , Bovinos , Endorribonucleasas/farmacología , Proteína Ligando Fas , Femenino , Humanos , Glicoproteínas de Membrana/biosíntesis , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Neoplasias Experimentales , Proteínas Proto-Oncogénicas c-bcl-2/biosíntesis , Neoplasias de la Tiroides/patología , Células Tumorales Cultivadas , Receptor fas/biosíntesis
10.
Anticancer Drugs ; 12(5): 467-73, 2001 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-11395575

RESUMEN

Treatment failure in most neuroblastoma (NB) patients is related to primary and/or acquired resistance to conventional chemotherapeutic agents. Aphidicolin (APH), a tetracyclic diterpene, exhibits specific cytotoxic action against NB cells. The purpose of this study was to compare antitumoral efficacy of APH in parental NB cell lines and cell subclones that exhibit drug resistance to vincristine (VCR), doxorubicin (DOX) and cisplatin. Due to poor solubility of APH in water, gamma-cyclodextrin (gamma-CD) inclusion complexes of APH were used for systemic treatment of xenotransplanted parental and VCR-resistant UKF-NB-3 tumours. APH and its gamma-CD inclusion complexes inhibited growth of parental and drug-resistant NB cells at equimolar doses in vitro. Growth of VCR-sensitive and -resistant NB tumors was inhibited at equal doses in a dose-dependent fashion in vivo. These results indicate that the specific cytotoxic activity of APH against NB cells in vitro and in vivo is independent of cellular mechanisms facilitating drug resistance to conventional chemotherapeutic drugs. Hence, taking into account our previous findings that APH acts synergistically with VCR and DOX, APH might be an additive tool for the therapy of NB and is suitable for evaluation in clinical studies of NB treatment protocols.


Asunto(s)
Afidicolina/uso terapéutico , Supervivencia Celular/efectos de los fármacos , Ciclodextrinas/farmacología , Inhibidores Enzimáticos/uso terapéutico , Neuroblastoma/tratamiento farmacológico , Células Tumorales Cultivadas/efectos de los fármacos , Animales , Antineoplásicos/uso terapéutico , Afidicolina/análogos & derivados , Peso Corporal/efectos de los fármacos , Portadores de Fármacos , Resistencia a Antineoplásicos , Femenino , Humanos , Ratones , Ratones Desnudos , Neuroblastoma/patología , Células Tumorales Cultivadas/patología
11.
J Immunol ; 165(8): 4405-13, 2000 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-11035078

RESUMEN

Human CMV (HCMV) retinitis frequently leads to blindness in iatrogenically immunosuppressed patients and in the end stage of AIDS. Despite the general proinflammatory potential of HCMV, virus infection is associated with a rather mild cellular inflammatory response in the retina. To investigate this phenomenon, the influence of HCMV (strains AD169 or Hi91) infection on C-X-C chemokine secretion, ICAM-1 expression, and neutrophil recruitment in cultured human retinal pigment epithelial (RPE) cells was studied. Supernatants from infected cultures contained enhanced levels of IL-8 and melanoma growth-stimulating activity/Gro alpha and induced neutrophil chemotaxis compared with supernatants from uninfected RPE cells. Despite HCMV-induced ICAM-1 expression on RPE cells, binding of activated neutrophils to HCMV-infected RPE cells and subsequent transepithelial penetration were significantly reduced. Reduced neutrophil adhesion to infected RPE cells correlated with HCMV-induced up-regulation of constitutive Fas ligand (FasL) expression. Functional blocking of FasL on RPE cells with the neutralizing mAbs NOK-1 and NOK-2 or of the Fas receptor on neutrophils with mAbB-D29 prevented the HCMV-induced impairment of neutrophil/RPE interactions. Fas-FasL-dependent impairment of neutrophil binding had occurred by 10 min after neutrophil/RPE coculture without apoptotic signs. Neutrophil apoptosis was first detected after 4 h. Treatment of neutrophils with a specific inhibitor of caspase-8 suppressed apoptosis, whereas it did not prevent impaired neutrophil binding to infected RPE. The current results suggest a novel role for FasL in the RPE regulation of neutrophil binding. This may be an important feature of virus escape mechanisms and for sustaining the immune-privileged character of the retina during HCMV ocular infection.


Asunto(s)
Apoptosis/inmunología , Citomegalovirus/inmunología , Glicoproteínas de Membrana/biosíntesis , Neutrófilos/inmunología , Epitelio Pigmentado Ocular/inmunología , Epitelio Pigmentado Ocular/virología , Regulación hacia Arriba/inmunología , Receptor fas/metabolismo , Adulto , Anticuerpos Bloqueadores/farmacología , Adhesión Celular/inmunología , Células Cultivadas , Quimiotaxis/inmunología , Citomegalovirus/genética , Proteína Ligando Fas , Humanos , Mediadores de Inflamación/metabolismo , Ligandos , Glicoproteínas de Membrana/antagonistas & inhibidores , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/inmunología , Neutrófilos/citología , Neutrófilos/virología , Epitelio Pigmentado Ocular/citología , Epitelio Pigmentado Ocular/patología
12.
Anticancer Drugs ; 11(6): 479-85, 2000 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-11001389

RESUMEN

Disseminated neuroblastoma diseases are still indicated by a poor outcome despite treatment regimens including radiation therapy and high-dose chemotherapy with stem cell rescue. Therefore, new substances and treatment regimens are of interest. Aphidicolin (APH), a tetracyclic diterpene antibiotic produced by Cephalosporium aphidicola, has a specific toxicity for neuroblastoma cells. Furthermore, it was shown to enhance the effects of X-ray radiation and chemotherapy on malignant cells. To find new substances, 20 APH derivatives were tested for their anti-neuroblastoma efficacy in vitro in UKF-NB-2 cells. Five derivatives had antitumoral activity in neuroblastoma cells. A relationship between the structure and the antitumoral efficacy showed that the hydroxyl groups at C-3 and C-18 are essential for the antitumoral effects. Furthermore, antitumoral effects of APH in combination with doxorubicin and vincristine, both part of commonly used treatment regimens for disseminated neuroblastoma diseases, were tested in the neuroblastoma cell line UKF-NB-2. APH was found to act synergistically with vincristine and synergistically to additive with doxorubicin depending on the molecular ratio of the substances in combination. This may offer the chance to use APH and its derivatives as additional tools in the treatment of neuroblastomas.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Antineoplásicos/farmacología , Afidicolina/farmacología , Supervivencia Celular/efectos de los fármacos , Doxorrubicina/farmacología , Inhibidores Enzimáticos/farmacología , Neuroblastoma/tratamiento farmacológico , Células Tumorales Cultivadas/efectos de los fármacos , Vincristina/farmacología , Afidicolina/análogos & derivados , Sinergismo Farmacológico , Humanos , Estructura Molecular , Neuroblastoma/patología , Inhibidores de la Síntesis del Ácido Nucleico , Relación Estructura-Actividad , Sales de Tetrazolio/metabolismo , Tiazoles/metabolismo , Células Tumorales Cultivadas/patología
13.
J Infect Dis ; 182(3): 643-51, 2000 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-10950755

RESUMEN

In fibroblasts, infection with human cytomegalovirus (HCMV) inhibits expression of the extracellular matrix proteins thrombospondin-1 and -2 (TSP-1 and TSP-2). These effects may depend on expression of HCMV immediate-early (IE) genes, which are activated by cellular transcription factor NF-kappaB. The influence of HCMV infection on TSP-1 and TSP-2 expression and the ability of different antiviral drugs to prevent these cellular changes in permissive cultures of human retinal glial cells were observed. Ganciclovir inhibited only HCMV late antigen (LA) expression, whereas antisense oligonucleotide ISIS 2922 and peptide SN50, inhibitors of HCMV IE expression and NF-kappaB activity, respectively, inhibited both IE and LA expression. ISIS 2922 and SN50, but not ganciclovir, prevented down-modulation of TSP-1 and TSP-2. The results showed that HCMV-induced down-modulation of TSP-1 and TSP-2 in retinal glial cells is prevented by inhibition of HCMV IE expression. These findings may be relevant to pathogenesis and treatment of HCMV retinitis.


Asunto(s)
Antivirales/farmacología , Retinitis por Citomegalovirus/metabolismo , Neuroglía/metabolismo , Retina/metabolismo , Trombospondina 1/biosíntesis , Trombospondinas/biosíntesis , Células Cultivadas , Regulación hacia Abajo , Electroforesis en Gel de Poliacrilamida , Citometría de Flujo , Ganciclovir/farmacología , Humanos , Proteínas Inmediatas-Precoces/metabolismo , FN-kappa B/metabolismo , Neuroglía/efectos de los fármacos , Oligodesoxirribonucleótidos Antisentido/farmacología , Péptidos/farmacología , Retina/efectos de los fármacos , Replicación Viral/efectos de los fármacos
14.
Tissue Antigens ; 55(5): 412-21, 2000 May.
Artículo en Inglés | MEDLINE | ID: mdl-10885561

RESUMEN

Previously, experimental in vivo results showed that the productively and persistently human cytomegalovirus (HCMV)-infected neuroblastoma cell line UKF-NB-4AD169 exhibits a more malignant phenotype than the non-infected variant UKF-NB-4. To prove the assumption that enhanced malignancy may be due to enhanced invasive potential of the infected cells we studied interactions of both lines with monolayers of cultured endothelial cells. UKF-NB-4AD169 cells adhered to and transmigrated through endothelial monolayer to a significantly higher extent compared with UKF-NB4. Furthermore, the adhesion of UKF-NB-4AD169 but not of UKF-NB4 resulted in focal disruption of the monolayer integrity which facilitates tumor cell transmigration. Blocking antibodies directed against the beta1 integrin chain as well as beta1alpha5 on the tumor cells specifically inhibited adhesion in a concentration-dependent manner. When UKF-NB-4 were pretreated with a beta1 integrin activating antibody, focal disruption of the endothelial integrity also occurred. These findings lead us to suggest that HCMV infection activates beta1alpha5 in the host neuroblastoma cell which in turn enables these cells to tightly adhere to endothelial cells. In the presence of the protease inhibitor phenantroline, beta1alpha5-mediated adhesion was not impaired whereas UKF-NB4AD169-mediated endothelial monolayer permeabilization was dose dependently inhibited. We conclude that human cytomegalovirus infection contributes to augmented neuroblastoma invasiveness via adhesion of activated beta1alpha5 and subsequent matrix digestion by proteases.


Asunto(s)
Infecciones por Citomegalovirus/patología , Citomegalovirus , Neuroblastoma/patología , Receptores de Fibronectina/metabolismo , Anticuerpos Bloqueadores/farmacología , Anticuerpos Monoclonales/farmacología , Adhesión Celular/fisiología , Comunicación Celular/fisiología , Movimiento Celular/fisiología , Selectina E/análisis , Células Epiteliales/química , Células Epiteliales/citología , Células Epiteliales/virología , Citometría de Flujo , Humanos , Inmunofenotipificación , Molécula 1 de Adhesión Intercelular/análisis , Glicoproteínas de Membrana/análisis , Invasividad Neoplásica , Neuroblastoma/virología , Receptores de Fibronectina/análisis , Receptores de Fibronectina/inmunología , Células Tumorales Cultivadas/química , Células Tumorales Cultivadas/citología , Células Tumorales Cultivadas/virología , Cordón Umbilical/citología , Molécula 1 de Adhesión Celular Vascular/análisis
15.
Anticancer Drugs ; 11(5): 369-76, 2000 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-10912953

RESUMEN

Bovine seminal ribonuclease (BS-RNase) is a protein with a number of biological effects. It shows antitumoral, aspermatogenic, antiembryonic, immunosuppressive and antiviral properties. The cytotoxic effects appear to be specific for tumor cells as non-malignant cells seem to be unaffected in vitro. Unfortunately, the in vivo application of BS-RNase so far was successful only when it was administered intratumorally. Therefore, the objective of the present investigation was to improve the properties of BS-RNase by attachment to nanoparticles made of polylactic acid (PLA-NP) using an adsorption method. This preparation was tested in vitro against leukemia (MOLT-4) and lymphoma (H9) cell lines sensitive and resistant to cytarabine. No difference between the nanoparticle preparation and pure BS-RNase was found in these tests. To examine the in vivo effects, the preparations were tested for their aspermatogenic and antiembryonal efficacy compared to the pure BS-RNase as a rapid test for antitumoral activity. The aspermatogenic and antiembryonal effects were enhanced by the nanoparticle preparation. Consequently, BS-RNase loaded adsorptively to PLA-NP holds promise for the in vivo use as an antitumoral agent. Further research will investigate the efficacy of this preparations in an in vivo tumor model.


Asunto(s)
Antineoplásicos/farmacología , Endorribonucleasas/farmacología , Leucemia/tratamiento farmacológico , Linfoma/tratamiento farmacológico , Células Tumorales Cultivadas/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Bovinos , División Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Portadores de Fármacos , Fibroblastos/efectos de los fármacos , Humanos , Ácido Láctico , Masculino , Ratones , Ratones Endogámicos ICR , Microesferas , Poliésteres , Polímeros , Espermatozoides/efectos de los fármacos , Testículo/efectos de los fármacos , Sales de Tetrazolio/metabolismo , Tiazoles/metabolismo
16.
Anticancer Res ; 20(2A): 853-9, 2000.
Artículo en Inglés | MEDLINE | ID: mdl-10810366

RESUMEN

BACKGROUND: Bovine seminal ribonuclease (BS-RNase) exerts selective cytotoxicity toward different types of tumor cells. In the present study, we tested the effects of BS-RNase on cultured neuroblastoma (NB) cells resistant to chemotherapeutic agents. The selectivity of the antitumoral activity of BS-RNase was evaluated using cultures of CD34+ hematopoietic stem cells. MATERIALS AND METHODS: Human NB cell lines including IMR-32, UKF-NB-2 and UKF-NB-3 were selected for resistance against vincristine, doxorubicin or cisplatin by exposure to increasing concentrations of the respective drug. The cytotoxicity of the drugs to NB cells was evaluated using a clonogenic assay in a methylcellulose medium. Peripheral blood progenitor cells were obtained from adult healthy donors by positive selection using specific anti-CD34+ antibodies. The toxicity of BS-RNase to CD34+ cells was assessed in the direct clonogenic assay using methylcellulose medium or in ex vivo expansion culture supplemented with hematopoietic growth factors. RESULTS: In the clonogenic assay it was shown that BS-RNase completely inhibits growth of both parental NB cells and their sublines resistant to chemotherapeutic drugs at concentrations (up to 50 micrograms/ml) which have no significant influence on the growth of colony-forming units, granulocyte macrophage and erythroid burst-forming units. Moreover, BS-RNase had no effect on the ex vivo expansion of total hematopoietic cells or of colony-forming cells from CD34+ progenitors. CONCLUSIONS: BS-RNase is a highly efficient agent against NB cells resistant to chemotherapeutic drugs. The lack of toxicity to hematopoietic progenitor cells suggests that BS-RNase is also likely to have tolerable hematopoietic toxicity.


Asunto(s)
Antineoplásicos/toxicidad , Supervivencia Celular/efectos de los fármacos , Resistencia a Múltiples Medicamentos , Endorribonucleasas/toxicidad , Células Madre Hematopoyéticas/efectos de los fármacos , Neuroblastoma/patología , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/genética , Adulto , Animales , Antígenos CD34/sangre , Bovinos , Células Cultivadas , Cisplatino/toxicidad , Doxorrubicina/toxicidad , Genes MDR/efectos de los fármacos , Células Madre Hematopoyéticas/citología , Humanos , Cinética , Neuroblastoma/genética , Células Tumorales Cultivadas , Vincristina/toxicidad
17.
J Med Virol ; 60(3): 313-23, 2000 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-10630964

RESUMEN

Human cytomegalovirus (HCMV) infection is associated with excessive proinflammatory immune responses such as cytokine/chemokine production or upregulation of adhesion molecules on the host cells. It is assumed that these features of HCMV-related immunopathology can not be treated effectively with currently available anti HCMV drugs. In the present study the efficacy of ganciclovir (GCV), foscarnet (PFA), cidofovir (HPMPC), and ISIS 2922, an antisense oligonucleotide complementary to HCMV immediate-early (IE) mRNA, was investigated on HCMV-induced secretion and functional activity of the C-X-C chemokine IL-8 and the expression of the intercellular adhesion molecule-1 (ICAM-1). As compared with mock-infected cells IL-8 production was increased up to 9-fold and ICAM-1 expression up to 4-fold in HCMV-infected fibroblasts. Treatment of infected cells with GCV (40 microM), PFA (200 microM) or HPMPC (2 microM) suppressed completely virus replication as demonstrated by quantification of late (L) antigen expression and infectious virus production. These drugs, however, failed to inhibit IE antigen expression and did not prevent HCMV-induced upregulation of IL-8 and ICAM-1. In contrast, ISIS 2922 (1 microM) suppressed both IE and L antigen expression by 99% and inhibited infectious virus production by 10(4)-fold. Moreover, ISIS 2922 significantly suppressed HCMV-induced upregulation of both IL-8 and ICAM-1 expression on the transcriptional and on the protein level. Our results indicate that ISIS 2922 but not inhibitors of HCMV DNA prevents HCMV-induced upregulation of IL-8 and ICAM-1, both hallmarks of inflammatory processes. Thus, inhibition of HCMV IE expression with ISIS 2922 may be an important strategy for the treatment of HCMV-related immunopathogenesis.


Asunto(s)
Antivirales/farmacología , Infecciones por Citomegalovirus/metabolismo , Molécula 1 de Adhesión Intercelular/metabolismo , Interleucina-8/metabolismo , Oligonucleótidos Antisentido/farmacología , Organofosfonatos , Tionucleótidos/farmacología , Células Cultivadas , Quimiotaxis de Leucocito , Cidofovir , Citosina/análogos & derivados , Citosina/farmacología , Fibroblastos/metabolismo , Fibroblastos/virología , Foscarnet/farmacología , Ganciclovir/farmacología , Humanos , Inmunohistoquímica , Molécula 1 de Adhesión Intercelular/efectos de los fármacos , Neutrófilos/fisiología , Compuestos Organofosforados/farmacología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Regulación hacia Arriba
18.
Int J Oncol ; 15(5): 1001-9, 1999 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-10536185

RESUMEN

Bovine seminal ribonuclease (BS-RNase) is a homologue of RNase A with specific antitumor activity. The cytotoxic action of this agent was examined in human neuroblastoma (NB) cell lines (SK-N-SH and UKF-NB-4) possessing the multidrug resistance (MDR) phenotype and NB cell lines (IMR-32, UKF-NB-1, UKF-NB-2 and UKF-NB-3) without MDR. Although MDR cells expressed large amounts of mdr-1 mRNA, contained functional P-glycoprotein and had 20- to 105-fold lower sensitivities to doxorubicin and vincristine than cells with non-MDR phenotypes, BS-RNase was equally toxic to all NB cells at concentrations employed (0.2 to 100 microg/ml). BS-RNase showed high selectivity for NB cells and was non-toxic to normal fibroblasts and epithelial cells. Ultrastructural investigation and annexin V assay showed that BS-RNase is a powerful inductor of apoptosis. The antitumoral effects of BS-RNase were also demonstrated in vivo using established subcutaneous xenografts in athymic (nude) mice of the MDR-1-bearing UKF-NB-4 cell line. Intratumoral injections (12.5 mg/kg) of BS-RNase over four weeks resulted in complete tumor regression and absence of tumor regrowth over a two-week observation period after cessation of treatment. The results show that BS-RNase selectively kills NB cells by inducing apoptosis and that this agent is active against mdr-1 expressing cells both in vitro and in vivo. BS-RNase fulfills important criteria for a candidate antitumor agent in NB patients with advanced disease.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/genética , Antineoplásicos/toxicidad , Apoptosis/fisiología , Neoplasias Encefálicas/patología , Resistencia a Múltiples Medicamentos , Neuroblastoma/patología , Ribonucleasas/toxicidad , Semen/enzimología , Animales , Apoptosis/efectos de los fármacos , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/ultraestructura , Bovinos , Supervivencia Celular/efectos de los fármacos , Humanos , Masculino , Ratones , Ratones Desnudos , Neuroblastoma/tratamiento farmacológico , Neuroblastoma/genética , Neuroblastoma/ultraestructura , Ribonucleasas/uso terapéutico , Transcripción Genética , Trasplante Heterólogo
19.
Am J Pathol ; 155(1): 285-92, 1999 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-10393860

RESUMEN

Thrombospondin-1 (TSP-1) is a potent inhibitor of angiogenesis. It has been shown that promoter sequences of the TSP-1 gene can be transactivated by the wild-type tumor suppressor protein p53. As human cytomegalovirus (HCMV) infection inactivates wild-type p53 of various cell types, we investigated whether HCMV infection is associated with reduced TSP-1 production. We found, in conjunction with accumulated p53, that TSP-1 mRNA and protein expression was significantly reduced in HCMV-infected cultured human fibroblasts. To determine whether the observed TSP-1 suppression depends on p53 inactivation, the p53-defective astrocytoma cell line U373MG was infected with HCMV. In these cells TSP-1 expression was also significantly reduced by HCMV infection whereas expression of the p53 mutant variant remained unaltered. In both cell lines the decreased expression of TSP-1 mRNA occurred early after infection (4 hours), indicating that HCMV inhibits TSP-1 transcription during the immediate-early phase of infection before HCMV DNA replication. Inhibition of HCMV DNA synthesis by ganciclovir did not influence TSP-1 reduction whereas the antisense oligonucleotide ISIS 2922, complementary to HCMV immediate-early mRNA, completely prevented the HCMV-mediated TSP-1 suppression. These findings strongly suggest a novel role for HCMV in the modulation of angiogenesis due to p53-independent down-regulation of TSP-1 expression.


Asunto(s)
Infecciones por Citomegalovirus/metabolismo , Trombospondina 1/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Antivirales/farmacología , Astrocitoma/metabolismo , Fibroblastos/metabolismo , Ganciclovir/farmacología , Humanos , ARN Mensajero/metabolismo , Tionucleótidos/farmacología , Trombospondina 1/genética , Células Tumorales Cultivadas
20.
Intervirology ; 42(5-6): 419-24, 1999.
Artículo en Inglés | MEDLINE | ID: mdl-10702726

RESUMEN

We observed the effects of antiviral therapy on CMV and/or oxidative-stress-induced stimulation of proinflammatory molecules including interleukin-8 (IL-8), melanoma growth stimulatory activity-alpha (GRO-alpha) and intercellular adhesion molecule 1 (ICAM-1) using human foreskin fibroblasts. Ganciclovir, foscarnet or cidofovir completely suppressed virus replication, as demonstrated by CMV late (L) antigen production. These drugs did not influence CMV immediate-early (IE) antigen expression and had no effects on CMV-induced cellular changes in IL-8, GRO-alpha and ICAM-1 levels. Phosphorothioate oligonucleotide (ISIS 2922) suppressed both CMV IE and L antigen by 99%. ISIS 2922 completely suppressed CMV-induced upregulation of both chemokines and ICAM-1. Induction of oxidative stress by H(2)O(2) upregulated IL-8 expression. Oxidative stress and CMV infection showed synergistic effects on IL-8 expression. ISIS 2922 only partially inhibited the upregulation of IL-8 in infected cells treated with H(2)O(2), whereas cotreatment with ISIS 2922 and antioxidants inhibited the upregulation almost completely. The results showed that inhibition of CMV IE expression alone or in combination with antioxidants is promising for the treatment of CMV disease.


Asunto(s)
Antígenos Virales/biosíntesis , Antioxidantes/uso terapéutico , Antivirales/uso terapéutico , Infecciones por Citomegalovirus/tratamiento farmacológico , Infecciones por Citomegalovirus/inmunología , Proteínas Inmediatas-Precoces/biosíntesis , Antígenos Virales/genética , Citocinas/metabolismo , Citomegalovirus/genética , Citomegalovirus/inmunología , Citomegalovirus/metabolismo , Infecciones por Citomegalovirus/virología , Humanos , Proteínas Inmediatas-Precoces/genética , Inflamación , Oxidación-Reducción , Estrés Oxidativo , Tionucleótidos/uso terapéutico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA