Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Cancer Res ; 78(10): 2513-2523, 2018 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-29510993

RESUMEN

Glycolysis and fatty acid synthesis are highly active in cancer cells through cytosolic citrate metabolism, with intracellular citrate primarily derived from either glucose or glutamine via the tricarboxylic acid cycle. We show here that extracellular citrate is supplied to cancer cells through a plasma membrane-specific variant of the mitochondrial citrate transporter (pmCiC). Metabolomic analysis revealed that citrate uptake broadly affected cancer cell metabolism through citrate-dependent metabolic pathways. Treatment with gluconate specifically blocked pmCiC and decreased tumor growth in murine xenografts of human pancreatic cancer. This treatment altered metabolism within tumors, including fatty acid metabolism. High expression of pmCiC was associated with invasion and advanced tumor stage across many human cancers. These findings support the exploration of extracellular citrate transport as a novel potential target for cancer therapy.Significance: Uptake of extracellular citrate through pmCiC can be blocked with gluconate to reduce tumor growth and to alter metabolic characteristics of tumor tissue. Cancer Res; 78(10); 2513-23. ©2018 AACR.


Asunto(s)
Proteínas de Transporte de Anión/antagonistas & inhibidores , Proteínas de Transporte de Anión/metabolismo , Proliferación Celular/efectos de los fármacos , Ácido Cítrico/metabolismo , Gluconatos/farmacología , Proteínas Mitocondriales/antagonistas & inhibidores , Proteínas Mitocondriales/metabolismo , Neoplasias Pancreáticas/patología , Neoplasias de la Próstata/patología , Animales , Línea Celular Tumoral , Células Epiteliales/metabolismo , Ácidos Grasos/biosíntesis , Glucólisis/fisiología , Humanos , Masculino , Ratones , Transportadores de Anión Orgánico , Próstata/citología , Próstata/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/genética
2.
J Biomed Sci ; 24(1): 78, 2017 Oct 16.
Artículo en Inglés | MEDLINE | ID: mdl-29037191

RESUMEN

BACKGROUND: Improving the neuronal yield from in vitro cultivated neural progenitor cells (NPCs) is an essential challenge in transplantation therapy in neurological disorders. In this regard, Ascorbic acid (AA) is widely used to expand neurogenesis from NPCs in cultures although the mechanisms of its action remain unclear. Neurogenesis from NPCs is regulated by the redox-sensitive WNT/ß-catenin signaling pathway. We therefore aimed to investigate how AA interacts with this pathway and potentiates neurogenesis. METHODS: Effects of 200 µM AA were compared with the pro-neurogenic reagent and WNT/ß-catenin signaling agonist lithium chloride (LiCl), and molecules with antioxidant activities i.e. N-acetyl-L-cysteine (NAC) and ruthenium red (RuR), in differentiating neural progenitor ReNcell VM cells. Cells were supplemented with reagents for two periods of treatment: a full period encompassing the whole differentiation process versus an early short period that is restricted to the cell fate commitment stage. Intracellular redox balance and reactive oxygen species (ROS) metabolism were examined by flow cytometry using redox and ROS sensors. Confocal microscopy was performed to assess cell viability, neuronal yield, and levels of two proteins: Nucleoredoxin (NXN) and the WNT/ß-catenin signaling component Dishevelled 2 (DVL2). TUBB3 and MYC gene responses were evaluated by quantitative real-time PCR. DVL2-NXN complex dissociation was measured by fluorescence resonance energy transfer (FRET). RESULTS: In contrast to NAC which predictably exhibited an antioxidant effect, AA treatment enhanced ROS metabolism with no cytotoxic induction. Both drugs altered ROS levels only at the early stage of the differentiation as no changes were held beyond the neuronal fate commitment stage. FRET studies showed that AA treatment accelerated the redox-dependent release of the initial pool of DVL2 from its sequestration by NXN, while RuR treatment hampered the dissociation of the two proteins. Accordingly, AA increased WNT/ß-catenin signaling output i.e. MYC mRNA level, whereas RuR attenuated it. Moreover, AA improved neurogenesis as much as LiCl as both TUBB3-positive cell yield and TUBB3 mRNA level increased, while NAC or RuR attenuated neurogenesis. Markedly, the neurogenesis outputs between the short and the full treatment with either NAC or AA were found unchanged, supporting our model that neuronal yield is altered by events taking place at the early phase of differentiation. CONCLUSIONS: Our findings demonstrate that AA treatment elevates ROS metabolism in a non-lethal manner prior to the NPCs commitment to their neuronal fate. Such effect stimulates the redox-sensitive DVL2 activation and WNT/ß-catenin signaling response that would enhance the ensuing neuronal cell differentiation.


Asunto(s)
Ácido Ascórbico/metabolismo , Diferenciación Celular , Células-Madre Neurales/efectos de los fármacos , Neurogénesis/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Vía de Señalización Wnt , Humanos , Células-Madre Neurales/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa
4.
J Hepatol ; 64(1): 128-34, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26341825

RESUMEN

BACKGROUND & AIMS: NKp46(+) cells are major effector cells in the pathogenesis of hepatic ischemia reperfusion injury (IRI). Nevertheless, the precise role of unconventional subsets like the IL-22-producing NKp46(+) cells (NK22) remains unknown. The purpose of this study was to examine the role of NK22 cells in IRI in transplantation, particularly with respect to regulation by the transcription factor ROR-gamma-t (RORγt). METHODS: To explore the role of NK22 cells in IRI in the absence of adaptive immunity, B6.RORγt-(gfp/wt)-reporter and B6.RORγt-(gfp/gfp)-knockout (KO) mice on a Rag KO background underwent 90min partial warm ischemia, followed by 24h of reperfusion. RESULTS: Rag KO mice that possess fully functional NKp46(+) cells, and Rag-common-γ-chain-double-KO (Rag-γc-DKO) mice that lack T, B and NKp46(+) cells, were used as controls. We found that Rag-γc-DKO mice lacking NK22 cells show more severe levels of hepatocellular damage (GPT, histological injury) when compared to both Rag-RORγt-reporter and Rag KO mice that possess NK22 cells. Importantly, Rag-RORγt-reporter and Rag KO mice undergoing IRI expressed high protein levels of both IL-22 and GFP (RORγt), suggesting a protective role for RORγt(+) NK22 cells in IRI. Therefore, we tested the hypothesis that RORγt critically protects from IRI through the induction of hepatic NK22 cells by studying Rag-Rorγt-DKO mice under IRI conditions. We found that the lack of RORγt(+) NK22 cells in Rag-Rorγt-DKO mice significantly enhanced IR-induced hepatocellular injury, a phenotype that could be reversed upon adoptive transfer of Rag-Rorγt-reporter NK22 cells into DKO mice. CONCLUSIONS: RORγt(+) NK22 cells play an important protective role in IRI in mice.


Asunto(s)
Antígenos Ly/fisiología , Interleucinas/biosíntesis , Hígado/irrigación sanguínea , Receptor 1 Gatillante de la Citotoxidad Natural/fisiología , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/fisiología , Daño por Reperfusión/prevención & control , Animales , Antígenos Ly/análisis , Proteínas de Homeodominio/fisiología , Interferón gamma/biosíntesis , Células Asesinas Naturales/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptor 1 Gatillante de la Citotoxidad Natural/análisis , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/análisis , Daño por Reperfusión/inmunología , Interleucina-22
5.
Cell Mol Neurobiol ; 35(8): 1203-16, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26022602

RESUMEN

Apoptosis is an essential physiological process accompanying the development of the central nervous system and human neurogenesis. However, the time scale and the underlying molecular mechanisms are yet poorly understood. Due to this fact, we investigated the functionality and general inducibility of apoptosis in the human neural ReNcell VM progenitor cell line during differentiation and also after exposure to staurosporine (STS) and ultraviolet B (UVB) irradiation. Transmission light microscopy, flow cytometry, and Western-/Immunoblot analysis were performed to compare proliferating and differentiating, in addition to STS- and UVB-treated cells. In particular, from 24 to 72 h post-initiation of differentiation, G0/G1 cell cycle arrest, increased loss of apoptotic cells, activation of pro-apoptotic BAX, Caspase-3, and cleavage of its substrate PARP were observed during cell differentiation and, to a higher extent, after treatment with STS and UVB. We conclude that redundant or defective cells are eliminated by apoptosis, while otherwise fully differentiated cells were less responsive to apoptosis induction by STS than proliferating cells, likely as a result of reduced APAF-1 expression, and increased levels of BCL-2. These data provide the evidence that apoptotic mechanisms in the neural ReNcell VM progenitor cell line are not only functional, but also inducible by external stimuli like growth factor withdrawal or treatment with STS and UVB, which marks this cell line as a suitable model to investigate apoptosis signaling pathways in respect to the differentiation processes of human neural progenitor cells in vitro.


Asunto(s)
Apoptosis/fisiología , Diferenciación Celular/fisiología , Células-Madre Neurales/fisiología , Neurogénesis/fisiología , Neuronas/fisiología , Transducción de Señal/fisiología , Línea Celular , Línea Celular Transformada , Humanos
6.
Transplantation ; 99(2): 391-9, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25606781

RESUMEN

BACKGROUND: Natural killer (NK) cells play a dichotomous role in alloimmune responses because they are known to promote both allograft survival and rejection. The aim of this study was to investigate the role of functionally distinct NK cell subsets in alloimmunity with the hypothesis that this dichotomy is explained by the functional heterogeneity of distinct NK cell subsets. METHODS: Because T-bet controls thematuration of NK cells from CD27high to terminally differentiated CD27low NK cells, we used Rag−/−T-bet−/− mice that lackmature CD27low NK cells to study the distinct roles of CD27low versus CD27high NK cells in a model of Tcell­mediated skin transplant rejection under costimulatory blockade conditions. RESULTS: We found that T cell­reconstituted Rag1−/− recipients (possessing CD27low NK cells) show significantly prolonged allograft survival on costimulatory blockade when compared to Rag1−/−T-bet−/− mice (lacking CD27low NK cells), indicating that CD27low but not CD27high NK cells enhance allograft survival. Critically, Rag1−/−T-bet−/− recipients showed strikingly increased alloreactive memory CD8+ Tcell responses, as indicated by increased CD8+ Tcell proliferation and interferon-γ production. Therefore, we speculated that CD27low NK cells directly regulate alloreactive CD8+ Tcell responses under costimulatory blockade conditions. To test this, we adoptively transferred CD27low NK cells into Rag1−/−T-bet−/− skin transplant recipients and found that the CD27low NK cells restore better allograft survival by inhibiting the proliferation of alloreactive interferon-γ+CD8+ T cells. CONCLUSIONS: In summary, mature CD27low NK cells promote allograft survival under costimulatory blockade conditions by regulating alloreactive memory CD8+ T-cell responses.


Asunto(s)
Linfocitos T CD8-positivos/metabolismo , Rechazo de Injerto/prevención & control , Supervivencia de Injerto , Células Asesinas Naturales/metabolismo , Trasplante de Piel/efectos adversos , Proteínas de Dominio T Box/metabolismo , Miembro 7 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/deficiencia , Traslado Adoptivo , Animales , Linfocitos T CD8-positivos/inmunología , Proliferación Celular , Genotipo , Rechazo de Injerto/genética , Rechazo de Injerto/inmunología , Rechazo de Injerto/metabolismo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Memoria Inmunológica , Interferón gamma/metabolismo , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/trasplante , Activación de Linfocitos , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , Transducción de Señal , Proteínas de Dominio T Box/deficiencia , Proteínas de Dominio T Box/genética , Proteínas de Dominio T Box/inmunología , Factores de Tiempo , Miembro 7 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/inmunología
7.
J Biol Chem ; 289(40): 27937-51, 2014 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-25124032

RESUMEN

Emerging evidence suggests that reactive oxygen species (ROS) can stimulate the Wnt/ß-catenin pathway in a number of cellular processes. However, potential sources of endogenous ROS have not been thoroughly explored. Here, we show that growth factor depletion in human neural progenitor cells induces ROS production in mitochondria. Elevated ROS levels augment activation of Wnt/ß-catenin signaling that regulates neural differentiation. We find that growth factor depletion stimulates the release of Ca(2+) from the endoplasmic reticulum stores. Ca(2+) subsequently accumulates in the mitochondria and triggers ROS production. The inhibition of mitochondrial Ca(2+) uptake with simultaneous growth factor depletion prevents the rise in ROS metabolism. Moreover, low ROS levels block the dissociation of the Wnt effector Dishevelled from nucleoredoxin. Attenuation of the response amplitudes of pathway effectors delays the onset of the Wnt/ß-catenin pathway activation and results in markedly impaired neuronal differentiation. Our findings reveal Ca(2+)-mediated ROS metabolic cues that fine-tune the efficiency of cell differentiation by modulating the extent of the Wnt/ß-catenin signaling output.


Asunto(s)
Calcio/metabolismo , Diferenciación Celular , Mitocondrias/metabolismo , Células-Madre Neurales/citología , Especies Reactivas de Oxígeno/metabolismo , Vía de Señalización Wnt , beta Catenina/metabolismo , Humanos , Células-Madre Neurales/metabolismo , Proteínas Wnt/metabolismo , beta Catenina/genética
8.
J Immunol ; 192(4): 1954-61, 2014 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-24415778

RESUMEN

We studied the developmental and functional mechanisms behind NK cell-mediated antitumor responses against metastatic colorectal carcinoma (CRC) in mice. In particular, we focused on investigating the significance of T-box transcription factors and the immunotherapeutic relevance of IL-15 in the development and function of tumor-reactive NK cells. Pulmonary CRC metastases were experimentally seeded via an adoptive i.v. transfer of luciferase-expressing CT26 CRC cells that form viewable masses via an in vivo imaging device; genetically deficient mice were used to dissect the antitumor effects of developmentally different NK cell subsets. IL-15 precomplexed to IL-15 receptor-α was used in immunotherapy experiments. We found that mice deficient for the T-box transcription factor T-bet lack terminally differentiated antitumor CD27(low)KLRG1(+) NK cells, leading to a terminal course of rapid-onset pulmonary CRC metastases. The importance of this NK cell subset for effective antitumor immunity was shown by adoptively transferring purified CD27(low)KLRG1(+) NK cells into T-bet-deficient mice and, thereby, restoring immunity against lung metastasis formation. Importantly, immunity to metastasis formation could also be restored in T-bet-deficient recipients by treating mice with IL-15 precomplexed to IL-15 receptor-α, which induced the development of eomesodermin(+)KLRG1(+) NK cells from existing NK cell populations. Thus, contingent upon their T-bet-dependent development and activation status, NK cells can control metastatic CRC in mice, which is highly relevant for the development of immunotherapeutic approaches in the clinic.


Asunto(s)
Neoplasias Colorrectales/patología , Células Asesinas Naturales/inmunología , Neoplasias Pulmonares/secundario , Receptores Inmunológicos/metabolismo , Proteínas de Dominio T Box/genética , Traslado Adoptivo , Animales , Diferenciación Celular/inmunología , Células Cultivadas , Neoplasias Colorrectales/inmunología , Neoplasias Colorrectales/terapia , Proteínas de Homeodominio/genética , Inmunoterapia , Interferón gamma/genética , Interleucina-15/metabolismo , Células Asesinas Naturales/citología , Lectinas Tipo C , Neoplasias Pulmonares/prevención & control , Neoplasias Pulmonares/terapia , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Perforina , Proteínas Citotóxicas Formadoras de Poros/genética , Receptores de Interleucina-15/metabolismo , Proteínas Recombinantes de Fusión/uso terapéutico , Proteínas de Dominio T Box/deficiencia , Proteínas de Dominio T Box/metabolismo , Miembro 7 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/metabolismo
9.
J Immunol ; 191(8): 4440-6, 2013 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-24058178

RESUMEN

Although Th1, Th2, and Th17 cells are thought to be major effector cells in adaptive alloimmune responses, their respective contribution to allograft rejection remains unclear. To precisely address this, we used mice genetically modified for the Th1 and Th17 hallmark transcription factors T-bet and RORγt, respectively, which allowed us to study the alloreactive role of each subset in an experimental transplant setting. We found that in a fully mismatched heterotopic mouse heart transplantation model, T cells deficient for T-bet (prone to Th17 differentiation) versus RORγt (prone to Th1 differentiation) rejected allografts at a more accelerated rate, indicating a predominance of Th17- over Th1-driven alloimmunity. Importantly, T cells doubly deficient for both T-bet and RORγt differentiated into alloreactive GATA-3-expressing Th2 cells, which promptly induced allograft rejection characterized by a Th2-type intragraft expression profile and eosinophilic infiltration. Mechanistically, Th2-mediated allograft rejection was contingent on IL-4, as its neutralization significantly prolonged allograft survival by reducing intragraft expression of Th2 effector molecules and eosinophilic allograft infiltration. Moreover, under IL-4 neutralizing conditions, alloreactive double-deficient T cells upregulated Eomesodermin (Eomes) and IFN-γ, but not GATA-3. Thus, in the absence of T-bet and RORγt, Eomes may salvage Th1-mediated alloimmunity that underlies IL-4 neutralization-resistant allograft rejection. We summarize that, whereas Th17 cells predictably promote allograft rejection, IL-4-producing GATA-3(+) Th2 cells, which are generally thought to protect allogeneic transplants, may actually be potent facilitators of organ transplant rejection in the absence of T-bet and RORγt. Moreover, Eomes may rescue Th1-mediated allograft rejection in the absence of IL-4, T-bet, and RORγt.


Asunto(s)
Aloinjertos/inmunología , Rechazo de Injerto/inmunología , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Proteínas de Dominio T Box/metabolismo , Células Th2/inmunología , Traslado Adoptivo , Animales , Diferenciación Celular , Eosinófilos/inmunología , Factor de Transcripción GATA3/biosíntesis , Trasplante de Corazón/efectos adversos , Interferón gamma/biosíntesis , Interleucina-4/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/deficiencia , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/genética , Proteínas de Dominio T Box/biosíntesis , Proteínas de Dominio T Box/deficiencia , Proteínas de Dominio T Box/genética
10.
Chembiochem ; 14(12): 1444-9, 2013 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-23843347

RESUMEN

Highly cytotoxic C7-modified colchicine analogues, exemplified by tubuloclustin, promote microtubule disassembly followed by the formation of very stable tubulin clusters, both in vitro and in cells. The proposed mechanism of action of tubuloclustin and its analogues, beyond that of colchicine, includes additional specific interactions with the α-tubulin subunit.


Asunto(s)
Adamantano/análogos & derivados , Colchicina/análogos & derivados , Colchicina/farmacología , Tubulina (Proteína)/metabolismo , Adamantano/química , Adamantano/farmacología , Animales , Células Cultivadas , Colchicina/química , Citotoxinas/química , Citotoxinas/farmacología , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Células HeLa , Humanos , Ratones , Microscopía Electrónica de Transmisión , Estructura Molecular , Unión Proteica/efectos de los fármacos
11.
Arthritis Res Ther ; 15(2): R49, 2013 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-23566364

RESUMEN

INTRODUCTION: An NZB-derived genetic locus (Sle2c2) that suppresses autoantibody production in a mouse model of induced systemic lupus erythematosus contains a polymorphism in the gene encoding the G-CSF receptor. This study was designed to test the hypothesis that the Sle2c2 suppression is associated with an impaired G-CSF receptor function that can be overcome by exogenous G-CSF. METHODS: Leukocytes from B6.Sle2c2 and B6 congenic mice, which carry a different allele of the G-CSF receptor, were compared for their responses to G-CSF. Autoantibody production was induced with the chronic graft-versus-host-disease (cGVHD) model by adoptive transfer of B6.bm12 splenocytes. Different treatment regimens varying the amount and frequency of G-CSF (Neulasta®) or carrier control were tested on cGVHD outcomes. Autoantibody production, immune cell activation, and reactive oxygen species (ROS) production were compared between the two strains with the various treatments. In addition, the effect of G-CSF treatment was examined on the production autoantibodies in the B6.Sle1.Sle2.Sle3 (B6.TC) spontaneous model of lupus. RESULTS: B6.Sle2c2 and B6 leukocytes responded differently to G-CSF. G-CSF binding by B6.Sle2c2 leukocytes was reduced as compared to B6, which was associated with a reduced expansion in response to in vivo G-CSF treatment. G-CSF in vivo treatment also failed to mobilize bone-marrow B6.Sle2c2 neutrophils as it did for B6 neutrophils. In contrast, the expression of G-CSF responsive genes indicated a higher G-CSF receptor signaling in B6.Sle2c2 cells. G-CSF treatment restored the ability of B6.Sle2c2 mice to produce autoantibodies in a dose-dependent manner upon cGVHD induction, which correlated with restored CD4+ T cells activation, as well as dendritic cell and granulocyte expansion. Steady-state ROS production was higher in B6.Sle2c2 than in B6 mice. cGVHD induction resulted in a larger increase in ROS production in B6 than in B6.Sle2c2 mice, and this difference was eliminated with G-CSF treatment. Finally, a low dose G-CSF treatment accelerated the production of anti-dsDNA IgG in young B6.TC mice. CONCLUSION: The different in vivo and in vitro responses of B6.Sle2c2 leukocytes are consistent with the mutation in the G-CSFR having functional consequences. The elimination of Sle2c2 suppression of autoantibody production by exogenous G-CSF indicates that Sle2c2 corresponds to a loss of function of G-CSF receptor. This result was corroborated by the increased anti-dsDNA IgG production in G-CSF-treated B6.TC mice, which also carry the Sle2c2 locus. Overall, these results suggest that the G-CSF pathway regulates the production of autoantibodies in murine models of lupus.


Asunto(s)
Formación de Anticuerpos/inmunología , Autoanticuerpos/inmunología , Factor Estimulante de Colonias de Granulocitos/inmunología , Lupus Eritematoso Sistémico/inmunología , Transducción de Señal/inmunología , Traslado Adoptivo , Animales , Formación de Anticuerpos/genética , Modelos Animales de Enfermedad , Citometría de Flujo , Factor Estimulante de Colonias de Granulocitos/genética , Lupus Eritematoso Sistémico/genética , Ratones , Ratones Congénicos , Ratones Endogámicos NZB , Ratones Mutantes , Transducción de Señal/genética
12.
Transplantation ; 95(1): 54-62, 2013 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-23222894

RESUMEN

BACKGROUND: Mammalian target of rapamycin (mTOR) inhibitors possess anticancer properties potentially useful in reducing posttransplantation malignancy. Besides controlling tumor-sensitive proliferative and angiogenic effects, mTOR influences transcription factors T-bet and Eomesodermin (Eomes) in CD8 cytotoxic T cells (Tc), which are key in rejecting tumors, and allografts. METHODS: To study the role of mTOR in tumor and transplant immunity in an antigen-specific way, we used T-cell receptor transgenic B6.OTI recipients, B6.OVA.TG donors, and OVA-B16F10 melanoma cells. For tracking color-coded OTI-Tc cells associated with antitumor and alloimmunity in vivo, CD8-OTI transgenic reporter mice were created by crossbreeding DsRed-expressing B6.Nagy mice with B6.OTI mice. RESULTS: The role of mTOR in regulating the differentiation and function of alloreactive Tc cells in vitro was explored by stimulating OTI-Tc cells with ovalbumin-transgenic antigen-presenting cells in the presence of rapamycin or tacrolimus. Rapamycin, but not tacrolimus, induced a pro-antitumor phenotypic shift from CD62LCD44 effector memory Tc cells to CD62LCD44 central memory Tc cells, which featured up-regulated levels of T-bet and Eomes and preserved levels of interferon-γ and perforin. For future investigations, an in vivo model was established whereby DsRedOTI-Tc cells adoptively transferred into B6 mice bearing either a ovalbumin-transgenic mouse skin transplant or OVA-B16F10 tumor could be traced by fluorescence-activated cell sorting analysis as effector or memory Tc cells in transplant and tumor tissues. CONCLUSION: mTOR, but not calcineurin, inhibition spares antitumoral memory Tc cells by distinctively regulating T-bet and Eomes. This finding is now testable in a new tumor transplant model, which incorporates DsRedOTI-Tc cell tracing, opening the way to study the differential effects of immunosuppressants in posttransplantation malignancy.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Memoria Inmunológica/inmunología , Inmunosupresores/farmacología , Neoplasias/inmunología , Trasplante Homólogo/inmunología , Animales , Linfocitos T CD8-positivos/efectos de los fármacos , Inhibidores de la Calcineurina , Interleucina-12/fisiología , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Sirolimus/farmacología , Proteínas de Dominio T Box/análisis , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/fisiología
13.
J Med Chem ; 54(20): 7138-49, 2011 Oct 27.
Artículo en Inglés | MEDLINE | ID: mdl-21916509

RESUMEN

A series of 4-azapodophyllotoxin derivatives with modified rings B and E have been synthesized using allylpolyalkoxybenzenes from parsley seed oil. The targeted molecules were evaluated in vivo in a phenotypic sea urchin embryo assay for antimitotic and tubulin destabilizing activity. The most active compounds identified by the in vivo sea urchin embryo assay featured myristicin-derived ring E. These molecules were determined to be more potent than podophyllotoxin. Cytotoxic effects of selected molecules were further confirmed and evaluated by conventional assays with A549 and Jurkat human leukemic T-cell lines including cell growth inhibition, cell cycle arrest, cellular microtubule disruption, and induction of apoptosis. The ring B modification yielded 6-OMe substituted molecule as the most active compound. Finally, in Jurkat cells, compound induced caspase-dependent apoptosis mediated by the apical caspases-2 and -9 and not caspase-8, implying the involvement of the intrinsic caspase-9-dependent apoptotic pathway.


Asunto(s)
Antimitóticos/síntesis química , Compuestos Aza/síntesis química , Petroselinum/química , Podofilotoxina/análogos & derivados , Podofilotoxina/síntesis química , Animales , Antimitóticos/farmacología , Apoptosis/efectos de los fármacos , Compuestos Aza/farmacología , Caspasa 2/metabolismo , Caspasa 9/metabolismo , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Embrión no Mamífero/efectos de los fármacos , Embrión no Mamífero/fisiología , Humanos , Microtúbulos/efectos de los fármacos , Microtúbulos/ultraestructura , Extractos Vegetales/química , Aceites de Plantas/química , Podofilotoxina/farmacología , Erizos de Mar/efectos de los fármacos , Erizos de Mar/embriología , Semillas/química , Estereoisomerismo , Relación Estructura-Actividad , Moduladores de Tubulina/síntesis química , Moduladores de Tubulina/farmacología
14.
Environ Health Perspect ; 118(10): 1363-9, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20457553

RESUMEN

BACKGROUND: Nanomaterials are extensively used in industry and daily life, but little is known about possible health effects. An intensified research regarding toxicity of nanomaterials is urgently needed. Several studies have demonstrated that nanoparticles (NPs; diameter < 100 nm) can be transported to the central nervous system; however, interference of NPs with the electrical activity of neurons has not yet been shown. OBJECTIVES/METHODS: We investigated the acute electrophysiological effects of carbon black (CB), hematite (Fe2O3), and titanium dioxide (TiO2) NPs in primary murine cortical networks on microelectrode array (MEA) neurochips. Uptake of NPs was studied by transmission electron microscopy (TEM), and intracellular formation of reactive oxygen species (ROS) was studied by flow cytometry. RESULTS: The multiparametric assessment of electrical activity changes caused by the NPs revealed an NP-specific and concentration-dependent inhibition of the firing patterns. The number of action potentials and the frequency of their patterns (spike and burst rates) showed a significant particle-dependent decrease and significant differences in potency. Further, we detected the uptake of CB, Fe2O3, and TiO2 into glial cells and neurons by TEM. Additionally, 24 hr exposure to TiO2 NPs caused intracellular formation of ROS in neuronal and glial cells, whereas exposure to CB and Fe2O3 NPs up to a concentration of 10 µg/cm2 did not induce significant changes in free radical levels. CONCLUSION: NPs at low particle concentrations are able to exhibit a neurotoxic effect by disturbing the electrical activity of neuronal networks, but the underlying mechanisms depend on the particle type.


Asunto(s)
Microelectrodos , Nanopartículas , Red Nerviosa , Potenciales de Acción , Animales , Ratones , Microscopía Electrónica de Transmisión
15.
Eur J Med Chem ; 45(5): 1683-97, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20110137

RESUMEN

A series of novel 1,3,4-oxadiazole derivatives based on structural and electronic overlap with combretastatins have been designed and synthesized. Initially, we tested all new compounds in vivo using the phenotypic sea urchin embryo assay to yield a number of agents with anti-proliferative, anti-mitotic, and microtubule destabilizing activities. The experimental data led to identification of 1,3,4-oxadiazole derivatives with isothiazole (5-8) and phenyl (9-12) pharmacophores featuring activity profiles comparable to that of combretastatins, podophyllotoxin and nocodazole. Cytotoxic effects of the two lead molecules, namely 6 and 12, were further confirmed and evaluated by conventional assays with the A549 human cancer cell line including cell proliferation, cell cycle arrest at the G2/M phase, cellular microtubule distribution, and finally in vitro microtubule assembly with purified tubulin. The modeling results using 3D similarity (ROCS) and docking (FRED) correlated well with the observed activity of the molecules. Docking data suggested that the most potent molecules are likely to target the colchicine binding site.


Asunto(s)
Antineoplásicos/farmacología , Microtúbulos/efectos de los fármacos , Mitosis/efectos de los fármacos , Oxadiazoles/farmacología , Erizos de Mar/efectos de los fármacos , Erizos de Mar/embriología , Animales , Antineoplásicos/síntesis química , Antineoplásicos/química , Bioensayo , Ciclo Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Microtúbulos/metabolismo , Modelos Moleculares , Estructura Molecular , Oxadiazoles/síntesis química , Oxadiazoles/química , Fenotipo , Erizos de Mar/metabolismo , Relación Estructura-Actividad , Células Tumorales Cultivadas
16.
Biochim Biophys Acta ; 1763(4): 402-12, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16713449

RESUMEN

This study focused on the cell activating capacity of extremely low frequency magnetic fields (ELF-MF) on human umbilical cord blood-derived monocytes. Our results confirm the previous findings of cell activating capacity of ELF-MF (1.0 mT) in human monocytes, which was detected as an increased ROS release. Furthermore, gene expression profiling (whole-genome cDNA array Human Unigene RZPD-2) was performed to achieve a comprehensive view of involved genes during the cell activation process after 45 min ELF-MF exposure. Our results indicate the alteration of 986 genes involved in metabolism, cellular physiological processes, signal transduction and immune response. Significant regulations could be analyzed for 5 genes (expression >2- or <0.5-fold): IL15RA (Interleukin 15 receptor, alpha chain), EPS15R (Epidermal growth factor receptor pathway substrate 15 - like 1), DNMT3A (Hypothetical protein MGC16121), DNMT3A (DNA (cytosine-5) methyltransferase 3 alpha), and one gene with no match to known genes, DKFZP586J1624. Real-time RT-PCR analysis of the kinetic of the expression of IL15RA, and IL10RA during 45 min ELF-MF exposure indicates the regulation of cell activation via the alternative pathway, whereas the delayed gene expression of FOS, IL2RA and the melatonin synthesizing enzyme HIOMT suggests the suppression of inflammatory processes. Accordingly, we suggest that ELF-MF activates human monocytes via the alternative pathway.


Asunto(s)
Campos Electromagnéticos , Perfilación de la Expresión Génica , Monocitos/metabolismo , Células Cultivadas , ADN Metiltransferasa 3A , Sangre Fetal/citología , Sangre Fetal/metabolismo , Humanos , Cinética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...