Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Transfusion ; 50(3): 547-55, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19906035

RESUMEN

BACKGROUND: Numerous mechanisms have been proposed to explain the beneficial action of intravenous immune globulin (IVIG) in autoimmune and systemic inflammatory disorders. Among others' data, an in vitro increase of intracellular TGF-beta expression when culturing CD4+ T lymphocytes in the presence of IVIG has been reported. As IVIG infusion involves administration of soluble contaminants likewise all hemoderivative preparations, we hypothesized that, besides several other immunomodulatory proposed mechanisms, the clinical effects of IVIG therapy might be, at least partly, due to contaminating soluble HLA Class I (sHLA-I) molecules capable to exert pleiotropic immunomodulatory effects among which TGF-beta(1) modulation. STUDY DESIGN AND METHODS: Ex vivo and in vitro transcriptional and posttranscriptional modulation of TGF-beta(1) in CD8+ T lymphocytes and neutrophils after IVIG infusion was analyzed. RESULTS: Ex vivo analysis of cells drawn from 10 enrolled IVIG recipients pointed out a significant increase of TGF-beta(1) mRNA and intracellular TGF-beta(1) molecules in both leukotypes. In vitro comparable results were obtained incubating CD8+ T lymphocytes and neutrophils from healthy donors with IVIG. The immunodepletion of sHLA-I and/or soluble Fas ligand (sFasL) abolished TGF-beta(1) modulation in both leukotypes. Coculture with human immunoglobulin (Ig)M monoclonal antibody or chimeric IgG (MabThera, Roche), whose manufacturing excludes "contamination," did not exert any mRNA modulation. Finally, IgM or MabThera plus purified sHLA-I molecules enhanced TGF-beta(1) mRNA in both white blood cells to levels comparable to those obtained with IVIG incubation. CONCLUSION: On the whole, these data lead us to speculate that the ability of IVIG administration to modulate TGF-beta(1) might be related to the immunomodulatory activities of sHLA-I and sFasL molecules on activated CD8+ T lymphocytes and neutrophils.


Asunto(s)
Linfocitos T CD8-positivos/metabolismo , Antígenos de Histocompatibilidad Clase I , Inmunoglobulinas Intravenosas , Factores Inmunológicos , Neutrófilos/metabolismo , Transcripción Genética/efectos de los fármacos , Factor de Crecimiento Transformador beta1/biosíntesis , Anticuerpos Monoclonales/farmacología , Células Cultivadas , Proteína Ligando Fas/administración & dosificación , Proteína Ligando Fas/farmacología , Femenino , Antígenos de Histocompatibilidad Clase I/administración & dosificación , Antígenos de Histocompatibilidad Clase I/farmacología , Humanos , Inmunoglobulinas Intravenosas/administración & dosificación , Inmunoglobulinas Intravenosas/farmacología , Factores Inmunológicos/administración & dosificación , Factores Inmunológicos/farmacología , Activación de Linfocitos/efectos de los fármacos , Masculino , ARN Mensajero/biosíntesis
2.
Int J Gynecol Cancer ; 20(9): 1474-81, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21119362

RESUMEN

OBJECTIVES: In cervical cancer, increased cytokeratin 18 (CK18) filament expression is associated with disease progression. However, it may also provide resistance to cytokine-induced apoptosis. The present study tested whether CK18 expression influences susceptibility to cytokine-induced apoptosis. METHODS: The cervical cancer cell lines C-4II (high CK18 expression), ME-180 (low CK18 expression), and 2 subtypes of HeLa cells containing or lacking CK18 expression (CK18+ and CK18- cells, respectively) were exposed to vehicle (control), Fas ligand (FasL) (50 ng/mL), or tumor necrosis factor α (TNF-α; 10 ng/mL) without/with cycloheximide (CHX; 2.5 µg/mL) to test the hypothesis that diminished CK18 expression increases susceptibility to cytokine-induced apoptosis. RESULTS: Flow cytometric analysis of cell death via TUNEL staining revealed that cytokine-induced apoptosis was 2-fold greater in ME-180 cells than C-4II cells in response to FasL+CHX or TNF-α+CHX (P < 0.05). Similarly, there was a higher incidence of FasL-induced apoptosis in CK18- HeLa cells (23% and 91% apoptotic for FasL and FasL+CHX, respectively) than CK18+ HeLa cells (1% and 11%, respectively; P < 0.05). Surprisingly, TNF-α had no effect on either CK18+ or CK18- HeLa cells (P > 0.05). Caspase 3 activity was greater in CK18- HeLa cells than in CK18+ HeLa cells at 8 and 18 hours after FasL treatment (P < 0.05), an effect abrogated by the caspase 8 inhibitor IETD-fmk (P < 0.05). CONCLUSIONS: Cervical cancer cells with diminished CK18 expression are more susceptible to cytokine-induced apoptosis, particularly in response to FasL treatment. These observations suggest that relative CK18 expression is an important factor when considering therapeutic strategies to enhance immune cell-mediated death of cervical cancer cells.


Asunto(s)
Apoptosis/efectos de los fármacos , Carcinoma/patología , Citocinas/farmacología , Queratina-18/metabolismo , Neoplasias del Cuello Uterino/patología , Apoptosis/fisiología , Carcinoma/genética , Carcinoma/metabolismo , Caspasa 3/metabolismo , Muerte Celular/efectos de los fármacos , Muerte Celular/genética , Citocinas/administración & dosificación , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/genética , Evaluación Preclínica de Medicamentos , Resistencia a Antineoplásicos/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Proteína Ligando Fas/administración & dosificación , Proteína Ligando Fas/farmacología , Femenino , Células HeLa , Humanos , Queratina-18/antagonistas & inhibidores , Queratina-18/genética , Queratina-18/fisiología , ARN Interferente Pequeño/administración & dosificación , ARN Interferente Pequeño/farmacología , Células Tumorales Cultivadas , Neoplasias del Cuello Uterino/genética , Neoplasias del Cuello Uterino/metabolismo
3.
Mol Cancer Ther ; 19(2): 513-524, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31871267

RESUMEN

Cytotoxic T-lymphocyte antigen 4 (CTLA4)-FasL, a homo-hexameric signal converter protein, is capable of inducing robust apoptosis in malignant cells of the B-cell lineage expressing its cognate B7 and Fas targets, while sparing nonmalignant ones. This fusion protein's striking proapoptotic efficacy stems from its complementary abilities to coordinately activate apoptotic signals and abrogate antiapoptotic ones. A limiting factor in translating FasL or Fas receptor agonists into the clinic has been lethal hepatotoxicity. Here, we establish CTLA4-FasL's in vivo efficacy in multiple murine and xenograft models, for both systemic and subcutaneous tumors. Significantly, good laboratory practice (GLP) toxicology studies in mice indicate that CTLA4-FasL given repeatedly at doses up to five times the effective dose was well-tolerated and resulted in no significant adverse events. An equivalent single dose of CTLA4-FasL administered to nonhuman primates was also well-tolerated, albeit with a moderate dose-dependent leukopenia that was completely reversible. Interestingly, monkey peripheral blood mononuclear cells were more sensitive to CTLA4-FasL-induced apoptosis when tested in vitro. In both species, there was short-term elevation in serum levels of IL6, IL2, and IFNγ, although this was not associated with clinical signs of proinflammatory cytokine release, and further, this cytokine elevation could be completely prevented by dexamethasone premedication. Liver toxicity was not observed in either species, as confirmed by serum liver enzyme levels and histopathologic assessment. In conclusion, CTLA4-FasL emerges from animal model studies as an effective and safe agent for targeted FasL-mediated treatment of B7-expressing aggressive B-cell lymphomas.


Asunto(s)
Antígeno CTLA-4/administración & dosificación , Proteína Ligando Fas/administración & dosificación , Proteínas Recombinantes de Fusión/farmacología , Secuencia de Aminoácidos , Animales , Antígeno CTLA-4/inmunología , Proteína Ligando Fas/efectos adversos , Proteína Ligando Fas/inmunología , Proteína Ligando Fas/farmacocinética , Femenino , Humanos , Células Jurkat , Macaca fascicularis , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Primates , Distribución Aleatoria , Proteínas Recombinantes de Fusión/efectos adversos , Proteínas Recombinantes de Fusión/inmunología , Proteínas Recombinantes de Fusión/farmacocinética , Ensayos Antitumor por Modelo de Xenoinjerto
4.
PLoS One ; 13(7): e0201087, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30024959

RESUMEN

Fas/Fas ligand system was shown to be related to insulin resistance and type 2 diabetes mellitus (T2DM). However, the role of soluble Fas ligand (sFasL) in functioning of immune cells in type 2 diabetes mellitus (T2DM) has not been studied yet. The aim of the present study was to determine in vitro effects of sFasL on neutrophil activation and apoptosis. We demonstrate here that sFasL exhibited proinflammatory effect and induced mRNA levels of caspase-1, NF-κB, IL-1ß and CD18 expression. At the same time, sFasL induced reactive oxygen species (ROS) production. Activation of caspase-1 activity abolished sFasL-dependent apoptosis, and suppressed Fas expression and mRNA levels of caspase-3 in neutrophils from T2DM patients. Collectively, our findings identify a novel proinflammatory role of sFasL in T2DM neutrophils that is dependent of caspase activity. Thus, sFasL enhances inflammatory response of neutrophils from T2DM patients without increasing apoptosis suggesting its triggering role in T2DM inflammation.


Asunto(s)
Diabetes Mellitus Tipo 2/sangre , Diabetes Mellitus Tipo 2/inmunología , Proteína Ligando Fas/metabolismo , Activación Neutrófila , Neutrófilos/inmunología , Adulto , Apoptosis/fisiología , Células Cultivadas , Proteína Ligando Fas/administración & dosificación , Femenino , Humanos , Interleucina-1beta/metabolismo , Interleucina-8/metabolismo , Masculino , Persona de Mediana Edad , ARN Mensajero/metabolismo
5.
Cancer Lett ; 354(2): 355-64, 2014 Nov 28.
Artículo en Inglés | MEDLINE | ID: mdl-25199763

RESUMEN

Fas and PI3K/Akt signaling pathways pivotally impact on cancer cell death and survival respectively and are considered as promising targets for innovative anticancer therapies. To better characterize the combination effect of PI3K/Akt inhibitors and Fas agonists and understand the profile of the interaction between PI3K/Akt and Fas signaling, we qualitatively and quantitatively evaluated the combination effect of PI3K/Akt inhibitors LY294002, Akt inhibitor VIII and FasL. At the concentration that can block cell cycle progression and DNA synthesis but not elicit apoptosis, these inhibitors potentiate FasL to induce apoptosis. At higher concentrations, when the PI3K/Akt inhibitors induce apoptosis, they synergize FasL to induce apoptosis. In addition, PI3K/Akt inhibition significantly facilitates the Fas-mediated apoptotic signaling. Understanding the combination effects between PI3K/Akt inhibition and Fas activation not only leads to rational design of effective combination therapy of PI3K/Akt inhibitors but also improve our knowledge about the impact of PI3K-Akt pathway on Fas signaling and the potential modulation of innate immune system by PI3K-Akt-targeting drugs in anticancer treatment.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Neoplasias del Colon/tratamiento farmacológico , Proteína Ligando Fas/farmacología , Inhibidores de las Quinasa Fosfoinosítidos-3 , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Receptor fas/metabolismo , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Cromonas/administración & dosificación , Cromonas/farmacología , Neoplasias del Colon/enzimología , Neoplasias del Colon/patología , Sinergismo Farmacológico , Proteína Ligando Fas/administración & dosificación , Proteína Ligando Fas/metabolismo , Técnicas de Inactivación de Genes , Células HCT116 , Humanos , Morfolinas/administración & dosificación , Morfolinas/farmacología , Inhibidores de Proteínas Quinasas/administración & dosificación , Proteínas Proto-Oncogénicas c-akt/deficiencia , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Interferente Pequeño/administración & dosificación , ARN Interferente Pequeño/genética , Proteínas Recombinantes/farmacología , Transducción de Señal , Transfección
6.
Urol Oncol ; 31(8): 1676-1682, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22717623

RESUMEN

OBJECTIVE: Intravesical bacillus Calmette-Guerin (BCG) is the gold standard for high-grade non-muscle-invasive bladder cancer (NMIBC); however, some patients do not respond to initial therapy while others relapse and/or progress. Therefore, combination strategies that can enhance the efficacy and sustainability of BCG are needed. Herein, we explore the efficacy of lenalidomide, a thalidomide derivative with immunomodulatory effects, in combination with BCG, both in vitro and in vivo. MATERIALS AND METHODS: We explored the outcomes of lenalidomide in combination with BCG in vivo using the MBT-2 cell line implanted in C3H immunocompetent mice. Apoptosis, cell proliferation, and microvessel density were measured by immunohistochemistry. In vitro, we performed Western blotting for cell cycle and apoptosis regulatory proteins and a chromatin condensation assay to evaluate TNF-α and FasL in combination with lenalidomide. RESULTS: In the mouse model, combination therapy with BCG and lenalidomide resulted in a statistically significant decrease in tumor size compared with the control group. IHC demonstrated a nonsignificant increase in apoptosis in the combination condition and no effect on cellular proliferation. Microvessel density was decreased in all treated conditions. In vitro, caspase-3 activation and chromatin condensation studies demonstrated increased cell death in the combinations of lenalidomide and TNF-α. CONCLUSIONS: The immunomodulatory molecule lenalidomide augments the response to BCG in an in vivo mouse model. This provides the rationale for studying the combination in patients with high grade NMIBC.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Proliferación Celular/efectos de los fármacos , Microvasos/efectos de los fármacos , Neoplasias de la Vejiga Urinaria/tratamiento farmacológico , Animales , Apoptosis/efectos de los fármacos , Vacuna BCG/administración & dosificación , Vacuna BCG/farmacología , Western Blotting , Caspasa 3/metabolismo , Línea Celular Tumoral , Sinergismo Farmacológico , Proteína Ligando Fas/administración & dosificación , Proteína Ligando Fas/farmacología , Femenino , Inmunohistoquímica , Lenalidomida , Ratones , Ratones Endogámicos C3H , Microvasos/metabolismo , Microvasos/patología , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Talidomida/administración & dosificación , Talidomida/análogos & derivados , Talidomida/farmacología , Carga Tumoral/efectos de los fármacos , Factor de Necrosis Tumoral alfa/administración & dosificación , Factor de Necrosis Tumoral alfa/farmacología , Neoplasias de la Vejiga Urinaria/metabolismo , Neoplasias de la Vejiga Urinaria/patología
7.
Invest Ophthalmol Vis Sci ; 51(7): 3560-6, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20164456

RESUMEN

PURPOSE. Current studies suggest that the immune system plays a critical role in blinding eye disorders. The eye is an immune-privileged site, and FasL expression is a major part of that mechanism because Fas/FasL interactions regulate inflammation and neovascularization, preventing damage to delicate ocular structures. These studies were undertaken to test the idea that modulating immune privilege might be an effective therapeutic approach to pathogenic angiogenesis in the eye. METHODS. C57BL/6 mice or FasL-defective B6-gld mice were laser treated to induce choroidal neovascularization (CNV). Mice were injected with cytotoxic FasL in the vitreous cavity or were treated with oral doxycycline in the drinking water. They were evaluated for CNV 7 days later. In some experiments eye tissue was harvested and evaluated for FasL expression, macrophage influx by immunohistochemistry, and release of sFasL. RESULTS. Injection of cytotoxic FasL successfully prevented neovascularization in a mouse model of CNV. Oral doxycycline increased functional FasL in the eye and substantially inhibited neovascularization. Doxycycline treatment increased FasL expression on the RPE cells and reduced circulating and tissue-associated sFasL. Treatment was ineffective in B6-gld mice, demonstrating that CNV inhibition was mediated by FasL. CONCLUSIONS. Targeting immune privilege using cytotoxic molecules or by increasing expression of the proapoptotic protein FasL may be a viable approach to treating neovascular eye disease.


Asunto(s)
Neovascularización Coroidal/prevención & control , Doxiciclina/administración & dosificación , Proteína Ligando Fas/administración & dosificación , Administración Oral , Animales , Antígeno CD11b/metabolismo , Neovascularización Coroidal/sangre , Neovascularización Coroidal/etiología , Modelos Animales de Enfermedad , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/metabolismo , Ensayo de Inmunoadsorción Enzimática , Proteína Ligando Fas/sangre , Técnica del Anticuerpo Fluorescente Indirecta , Expresión Génica , Sistema Inmunológico/efectos de los fármacos , Inyecciones , Ratones , Ratones Endogámicos C57BL , Epitelio Pigmentado de la Retina/metabolismo , Ligando Inductor de Apoptosis Relacionado con TNF/uso terapéutico , Cuerpo Vítreo
8.
Cell Mol Immunol ; 6(3): 167-74, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19567199

RESUMEN

The prognosis of hepatocellular carcinoma (HCC) is poor, even with the combined treatment of curative resection and adjuvant chemoradiotherapy. To solve this problem, many biologic therapies have been investigated. Fas ligand (FasL, CD95L) is mainly expressed in activated T lymphocytes and natural killer (NK) cells, and plays a central role in both cell-mediated immunity and immune downregulation. Several studies have shown that FasL is expressed in HCC. In the present report, we prepared recombinant human pET-22b(+)/FasL protein and investigated the effect of FasL on HCC cells in vitro and on tumor growth in a murine HCC tumor model. The well-known cytotoxic chemotherapeutic reagent adriamycin (ADM) served as a control. We found that FasL effectively suppressed the viability of H22 tumor cells and significantly induced the apoptosis of H22 cells. The apoptotic levels of cells treated with FasL-ADM were significantly higher than those treated with FasL or ADM alone, and the FasL-ADM combination resulted in a more than additive effect on tumor growth delay in this model. The results suggested that combined treatment of FasL and other chemotherapeutic agents may be a new approach to improve the efficacy of chemotherapy for HCC.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Doxorrubicina/farmacología , Proteína Ligando Fas/farmacología , Neoplasias Hepáticas Experimentales/tratamiento farmacológico , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Doxorrubicina/administración & dosificación , Sinergismo Farmacológico , Proteína Ligando Fas/administración & dosificación , Proteína Ligando Fas/genética , Femenino , Citometría de Flujo , Humanos , Hígado/efectos de los fármacos , Hígado/patología , Neoplasias Hepáticas Experimentales/patología , Masculino , Ratones , Ratones Endogámicos BALB C , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/farmacología , Resultado del Tratamiento , Carga Tumoral/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA