Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Nephrol Dial Transplant ; 32(12): 2132-2141, 2017 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-29045706

RESUMEN

BACKGROUND: Kidney injury molecule-1 (KIM-1) and neutrophil gelatinase-associated lipocalin (NGAL) are promising biomarkers for monitoring delayed graft function (DGF) after kidney transplantation. Here we investigated localization and distribution of KIM-1 and NGAL staining in renal allograft biopsies and studied their association with histological features, functional DGF (fDGF) and the tubular function slope (TFS), a functioning proximal tubular epithelial cell (PTEC) marker. METHODS: Day 10 protocol biopsies of 64 donation after circulatory death recipients were stained for KIM-1 and NGAL and the positive area was quantified using ImageJ software. Biopsies were scored according to Banff and acute tubular necrosis (ATN) criteria. A 99mtechnetium-mercaptoacetyltriglycine (99mTc-MAG3)-renography was performed to calculate TFS. RESULTS: KIM-1 staining was located on the brush border of tubular epithelial cells (TECs) and correlated with denudation, while NGAL was present more focally in a cytoplasmic distribution. KIM-1 and NGAL staining were not correlated and no co-localization was observed. Quantitative stainings were not associated with fDGF, but KIM-1 tended to be higher in patients with prolonged fDGF (≥21 days; P = 0.062). No correlation was observed between the quantitative tissue stainings and urinary KIM-1 or NGAL. Quantitative KIM-1 staining was inversely correlated with the TFS (Spearman's ρ = -0.53; P < 0.001), whereas NGAL was not. The latter finding might be because cortical NGAL staining is dependent on filtration and subsequent reabsorption by functioning PTECs. Staining of NGAL was indeed restricted to PTECs, as shown by co-localization with a PTEC-specific lectin. CONCLUSIONS: KIM-1 and NGAL staining showed different localization and distribution. Quantitative KIM-1 staining was inversely correlated with functioning PTECs.


Asunto(s)
Biomarcadores/metabolismo , Moléculas de Adhesión Celular/metabolismo , Funcionamiento Retardado del Injerto/diagnóstico , Células Epiteliales/patología , Receptor Celular 1 del Virus de la Hepatitis A/metabolismo , Trasplante de Riñón/efectos adversos , Túbulos Renales Proximales/patología , Anciano , Animales , Biopsia , Funcionamiento Retardado del Injerto/metabolismo , Células Epiteliales/metabolismo , Femenino , Humanos , Túbulos Renales Proximales/lesiones , Túbulos Renales Proximales/metabolismo , Lipocalina 2/metabolismo , Masculino , Persona de Mediana Edad , Ratas , Ratas Endogámicas Lew , Coloración y Etiquetado , Trasplante Homólogo
2.
Eur J Immunol ; 45(6): 1736-47, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25820702

RESUMEN

IL-35 is a cytokine of the IL-12 family, existing as a heterodimer of IL-12p35 and Ebi3. IL-35 has anti-inflammatory properties and is produced by regulatory T cells in humans and mice, where it is required for optimal suppression of immune responses. Distinct from other IL-12 cytokines, the expression of IL-35 has not been described in antigen-presenting cells. In view of the immune-regulatory properties of IL-35, we investigated the expression, regulation, and function of IL-12p35 and Ebi3 in human monocyte-derived dendritic cells and tolerogenic DCs (tolDCs). These tolDCs do not produce IL-12p70 or the homodimer IL-12p40. We demonstrate that tolDCs completely lack transcriptional expression of IL-12p40. However, tolDCs maintain mRNA expression of IL-12p35 and Ebi3. Using intracellular flow cytometry and Western blot analysis, we show that tolDCs produce Ebi3 and IL-12p35, and both can be enhanced upon stimulation with IFN-γ, LPS, or CD40L. tolDCs supernatants have the capacity to suppress T-cell activation. Using IL12A silencing, we demonstrate that IL-12p35 is required for tolDCs to reach their full suppressive potential. Taken together, our results indicate that tolDCs produce IL-35, providing an additional novel mechanism by which tolDCs elicit their tolerogenic potential.


Asunto(s)
Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Tolerancia Inmunológica , Interleucinas/biosíntesis , Células Presentadoras de Antígenos/inmunología , Células Presentadoras de Antígenos/metabolismo , Antígenos B7/metabolismo , Antígeno B7-2/metabolismo , Antígeno B7-H1/metabolismo , Células Dendríticas/efectos de los fármacos , Dexametasona/farmacología , Expresión Génica , Humanos , Tolerancia Inmunológica/efectos de los fármacos , Interleucina-12/biosíntesis , Subunidad p35 de la Interleucina-12/genética , Subunidad p35 de la Interleucina-12/metabolismo , Interleucina-27/genética , Interleucina-27/metabolismo , Interleucinas/genética , Interleucinas/metabolismo , Lipopolisacáridos/inmunología , Antígenos de Histocompatibilidad Menor , Fenotipo
3.
Cell Immunol ; 284(1-2): 1-8, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23916683

RESUMEN

Reactive oxygen species (ROS) produced by macrophages have recently been shown to have immunosuppressive properties and induce regulatory T cells. Here we investigated the ROS producing capacity of well-defined human Mph2 subsets and studied the contribution of ROS in the Mph-T cell interaction. Mph were generated from monocytes using M-CSF (Mph2), IL-4 (Mph2a), or IL-10 (Mph2c). Upon PMA stimulation, Mph2 and Mph2c showed a high ROS producing capacity, whereas this was low for Mph2a. Mph2 and Mph2c displayed a reduced T cell stimulatory capacity compared to Mph2a. Addition of the ROS inhibitor DPI decreased the T cell proliferation and IFN-γ production. When testing directly on Mph, DPI dose-dependently decreased the IL-10 and IL-12p40 production of CD40L-stimulated Mph2 subsets. In conclusion, the ROS producing capacity is different among human Mph type-2 subsets. In all cases, DPI suppressed T cell proliferation and cytokine production, indicating a ROS-dependent mechanism of T cell activation.


Asunto(s)
Macrófagos/inmunología , Especies Reactivas de Oxígeno/inmunología , Linfocitos T Reguladores/inmunología , Citometría de Flujo , Humanos , Activación de Linfocitos/inmunología , Factor Estimulante de Colonias de Macrófagos/inmunología , Macrófagos/metabolismo , Compuestos Onio/farmacología , Reacción en Cadena de la Polimerasa , ARN Mensajero/química , ARN Mensajero/genética , Especies Reactivas de Oxígeno/metabolismo
4.
Proc Natl Acad Sci U S A ; 107(41): 17686-91, 2010 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-20861446

RESUMEN

The phagocyte NAPDH-oxidase complex consists of several phagocyte oxidase (phox) proteins, generating reactive oxygen species (ROS) upon activation. ROS are involved in the defense against microorganisms and also in immune regulation. Defective ROS formation leads to chronic granulomatous disease (CGD) with increased incidence of autoimmunity and disturbed resolution of inflammation. Because regulatory T cells (Tregs) suppress autoimmune T-cell responses and are crucial in down-regulating immune responses, we hypothesized that ROS deficiency may lead to decreased Treg induction. Previously, we showed that in p47(phox)-mutated mice, reconstitution of macrophages (Mph) with ROS-producing capacity was sufficient to protect the mice from arthritis. Now, we present evidence that Mph-derived ROS induce Tregs. In vitro, we showed that Mph ROS-dependently induce Treg, using an NADPH-oxidase inhibitor. This finding was confirmed genetically: rat or human CGD Mph with mutated p47(phox) or gp91(phox) displayed hampered Treg induction and T-cell suppression. However, basal Treg numbers in these subjects were comparable to those in controls, indicating a role for ROS in induction of peripheral Tregs. Induction of allogeneic delayed-type hypersensitivity with p47(phox)-mutated Mph confirmed the importance of Mph-derived ROS in Treg induction in vivo. We conclude that NAPDH oxidase activity in Mph is important for the induction of Tregs to regulate T cell-mediated inflammation.


Asunto(s)
Macrófagos/metabolismo , NADPH Oxidasas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Linfocitos T Reguladores/inmunología , Animales , Cartilla de ADN/genética , Citometría de Flujo , Enfermedad Granulomatosa Crónica/inmunología , Humanos , Glicoproteínas de Membrana/inmunología , NADPH Oxidasa 2 , NADPH Oxidasas/antagonistas & inhibidores , NADPH Oxidasas/inmunología , Ratas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Estadísticas no Paramétricas , Linfocitos T Reguladores/citología , Linfocitos T Reguladores/metabolismo
5.
J Immunol ; 185(12): 7252-61, 2010 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-21076069

RESUMEN

The plastic role of dendritic cells (DCs) in the regulation of immune responses has made them interesting targets for immunotherapy, but also for pathogens or tumors to evade immunity. Functional alterations of DCs are often ascribed to manipulation of canonical NF-κB activity. However, though this pathway has been linked to murine myeloid DC biology, a detailed analysis of its importance in human myeloid DC differentiation, survival, maturation, and function is lacking. The myeloid DC subsets include interstitial DCs and Langerhans cells. In this study, we investigated the role of canonical NF-κB in human myeloid DCs generated from monocytes (monocyte-derived DCs [mo-DCs]) or CD34(+) progenitors (CD34-derived myeloid DCs [CD34-mDCs]). Inhibition of NF-κB activation during and after mo-DC, CD34-interstitial DC, or CD34-Langerhans cell differentiation resulted in apoptosis induction associated with caspase 3 activation and loss of mitochondrial transmembrane potential. Besides regulating survival, canonical NF-κB activity was required for the acquisition of a DC phenotype. Despite phenotypic differences, however, Ag uptake, costimulatory molecule and CCR7 expression, as well as T cell stimulatory capacity of cells generated under NF-κB inhibition were comparable to control DCs, indicating that canonical NF-κB activity during differentiation is redundant for the development of functional APCs. However, both mo-DC and CD34-mDC functionality were reduced by NF-κB inhibition during activation. In conclusion, canonical NF-κB activity is essential for the development and function of mo-DCs as well as CD34-mDCs. Insight into the role of this pathway may help in understanding how pathogens and tumors escape immunity and aid in developing novel treatment strategies aiming to interfere with human immune responses.


Asunto(s)
Antígenos CD34 , Diferenciación Celular/inmunología , Células de Langerhans/inmunología , Células Mieloides/inmunología , FN-kappa B/inmunología , Antígenos/inmunología , Antígenos/metabolismo , Apoptosis/inmunología , Caspasa 3/inmunología , Caspasa 3/metabolismo , Células Cultivadas , Regulación de la Expresión Génica/inmunología , Humanos , Células de Langerhans/citología , Células de Langerhans/metabolismo , Células Mieloides/citología , Células Mieloides/metabolismo , FN-kappa B/metabolismo , Receptores CCR7/biosíntesis , Receptores CCR7/inmunología , Células Madre/citología , Células Madre/inmunología , Células Madre/metabolismo
6.
J Leukoc Biol ; 96(2): 313-24, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24782489

RESUMEN

Uptake of apoptotic cells by DCs is considered to contribute to induction and maintenance of immunological tolerance. TolDCs are sought after as cellular therapy in transplantation and autoimmunity and can be generated in vitro using GCs. In this study, we investigated how uptake of dead cells affects the production and expression of different members of the IL-12 family by immature DCs or TolDCs. We show that compared to regular immature DCs, TolDCs display elevated levels of PS-recognizing bridge molecule receptors αvß5 and CD36, and have enhanced phagocytic abilities with accelerated uptake of apoptotic cells. We confirm that apoptotic cell uptake results in diminished production of IL-12p40 and IL-12p70 by DCs. We now show that this also results in increased expression of IL-12p35 and Ebi3. TolDCs completely lack expression of IL-12p40 yet have enhanced levels of Ebi3 and IL-12p35. Uptake by TolDCs of apoptotic or necrotic cells does not affect the expression of Ebi3/IL-12p35 and also does not increase IL-12p40. This is distinct from the culture of immature DCs with necrotic cells, which is sufficient to induce IL-12p40 secretion. Conversely, ingestion of apoptotic cells by DCs leads to increased expression of IL-12p35 and Ebi3 without affecting IL-12p40. In conclusion, we have shown that uptake of apoptotic versus necrotic cells by DCs differentially regulates members of the IL-12 family. Apoptotic cells favor expression of Ebi3 and IL-12p35, and we propose that differential regulation of the IL-12 family is an additional mechanism in determining the immune response to dying cells.


Asunto(s)
Apoptosis/inmunología , Células Dendríticas/inmunología , Regulación de la Expresión Génica/inmunología , Interleucina-12/inmunología , Fagocitosis/inmunología , Células Cultivadas , Humanos , Necrosis/inmunología
7.
Mol Immunol ; 49(3): 549-57, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22047959

RESUMEN

Macrophages have been demonstrated to suppress T cell responses by producing reactive oxygen species (ROS) leading to the subsequent induction of T regulatory cells in a ROS-dependent manner. Macrophages may therefore be instrumental in downregulating T cell responses in situations of exacerbated immune responses. Here we investigated the effect of immunosuppressive drugs on ROS production by macrophage subsets and the subsequent effects on T cell activation. Macrophage types 1 and 2 were differentiated with GM-CSF or M-CSF, in presence or absence of dexamethasone, cyclosporine A, FK506, rapamycin, or mycophenolic acid. The ROS producing capacity of fully differentiated Mph was highest in anti-inflammatory Mph2 and not affected by exposure to immunosuppressive drugs. However, presence of rapamycin during Mph2 differentiation decreased the ROS production of these cells. In contrast, other immunosuppressive drugs, with dexamethasone being the most potent, increased the ROS producing capacity of Mph2. Intriguingly although the ROS producing ability of Mph1 was unaffected, dexamethasone strongly increased the ROS producing capabilities of dendritic cells. Both at the mRNA and protein level we found that dexamethasone enhanced the expression of NOX2 protein p47(phox). Functionally, dexamethasone further enhanced the capacity of Mph2 to suppress T cell mediated IFN-γ and IL-4 production. In vivo, only in rats with normal ROS production (congenic DA.Ncf1(E3/E3)) it was observed that dexamethasone injection resulted in long-lasting upregulation of ROS production by macrophages and induced higher levels of Treg in a ROS-dependent manner. In conclusion, we show that the anti-inflammatory drug dexamethasone increases the ROS producing capacity of macrophages.


Asunto(s)
Dexametasona/farmacología , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Especies Reactivas de Oxígeno/metabolismo , Linfocitos T/inmunología , Animales , Células Cultivadas , NADPH Oxidasas/inmunología , NADPH Oxidasas/metabolismo , Unión Proteica , Ratas
8.
Am J Transplant ; 5(9): 2114-25, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16095490

RESUMEN

Graft-infiltrating dendritic cells (DC) and alloreactive T lymphocytes play a critical role in renal allograft rejection. Renal proximal tubular epithelial cells (TEC) are considered as active players in the attraction of leukocytes during renal inflammatory responses. Macrophage inflammatory protein (MIP)-3alpha/CCL20 is a major chemokine expressed by epithelial cells that attracts immature DC. In the present study, we present evidence that also the transplanted kidney can be a major source of MIP-3alpha/CCL20. Renal transplant recipients with rejection showed significantly increased excretion of urinary MIP-3alpha/CCL20 that correlated with transplant function. The tubular staining for MIP-3alpha/CCL20 in renal biopsies of patients with rejection as well as in vitro studies with primary human TEC indicated that TEC might be responsible for the increased urinary MIP-3alpha/CCL20. Furthermore, MIP-3alpha/CCL20 produced by activated TEC was highly potent in the attraction of CD1a+CD34+-derived DC precursors. These data suggest a role for MIP-3alpha/CCL20 in amplification of the immune response during renal allograft rejection by attraction of CCR6+ inflammatory cells, which may include DC, to the site of inflammation.


Asunto(s)
Quimiocinas CC/biosíntesis , Células Dendríticas/citología , Trasplante de Riñón/métodos , Proteínas Inflamatorias de Macrófagos/biosíntesis , Trasplante Homólogo/métodos , Adulto , Antígenos CD1/biosíntesis , Antígenos CD34/biosíntesis , Biopsia , Ligando de CD40/biosíntesis , Movimiento Celular , Quimiocina CCL20 , Quimiocina CCL5/biosíntesis , Quimiocinas/metabolismo , Factores Quimiotácticos , Células Dendríticas/metabolismo , Relación Dosis-Respuesta a Droga , Ensayo de Inmunoadsorción Enzimática , Células Epiteliales/patología , Femenino , Citometría de Flujo , Regulación de la Expresión Génica , Rechazo de Injerto , Humanos , Inmunohistoquímica , Inflamación , Interleucina-1/biosíntesis , Riñón/patología , Túbulos Renales/patología , Leucocitos/citología , Masculino , Persona de Mediana Edad , Modelos Estadísticos , Receptores CCR6 , Receptores de Quimiocina/biosíntesis , Factores de Tiempo
9.
Kidney Int ; 68(5): 2091-102, 2005 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16221208

RESUMEN

BACKGROUND: Renal tubular epithelial cells (TECs) play an active role in renal inflammation. Previous studies have demonstrated the capacity of TECs to modulate T-cell responses both positively and negatively. Recently, new costimulatory molecules [inducible T cell costimulator-L (ICOS-L) and B7-H1] have been described, which appear to be involved in peripheral T-cell activation. METHODS: We characterized expression and regulation of costimulatory molecules on primary human TECs and the TEC line human kidney-2 (HK-2) with reverse transcription-polymerase chain reaction (RT-PCR) and flow cytometry. Immunohistochemistry was performed on human kidney biopsies. The capacity of TECs to modulate T-cell activation was studied in TEC/T-cell cultures. RESULTS: We demonstrate that TECs express ICOS-L and B7-H1 in vitro and in vivo. Stimulation with interferon-gamma (IFN-gamma) resulted in increased expression of B7-H1, whereas ICOS-L expression was marginally increased upon stimulation with CD40L, with no effect of interleukin (IL-1), IL-17, or tumor necrosis factor-alpha (TNF-alpha). Furthermore, we show that TECs are able to costimulate T cells that have received signal-1 using alphaCD3 antibodies, inducing strong IL-10 production, which was partially mediated by ICOS-L. In contrast, B7-H1 appeared to be involved in inhibition of proliferation and cytokine synthesis. In addition, TECs were able to alter the cytokine profile of fully activated T cells, which were incubated with alphaCD3 and alphaCD28 antibodies, resulting in low IFN-gamma and high IL-10 production. This activity appeared to be independent of ICOS-L and B7-H1. CONCLUSION: Interaction of tubular epithelial cells and kidney infiltrating T cells via ICOS-L and B7-H1 may change the balance of positive and negative signals to the T cells, leading to IL-10 production and limitation of local immune responses.


Asunto(s)
Antígeno B7-1/genética , Túbulos Renales/citología , Túbulos Renales/inmunología , Glicoproteínas de Membrana/genética , Péptidos/genética , Proteínas/genética , Linfocitos T/citología , Linfocitos T/inmunología , Antígenos CD , Antígeno B7-H1 , Comunicación Celular/inmunología , Línea Celular Transformada , Células Epiteliales/citología , Células Epiteliales/fisiología , Expresión Génica/inmunología , Humanos , Ligando Coestimulador de Linfocitos T Inducibles , Interferón gamma/metabolismo , Interleucina-10/metabolismo , Activación de Linfocitos/fisiología , Transducción de Señal/inmunología , Linfocitos T/metabolismo
10.
J Immunol ; 172(10): 6482-9, 2004 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-15128841

RESUMEN

Sanglifehrin A (SFA) is a recently developed immunosuppressant that belongs to the family of immunophilin-binding ligands. SFA is a cyclophilin A-binding immunosuppressive drug with a novel, but unidentified, mechanism of action. Several reports exist about the effect of SFA on T cells, but its effect on the initiators of the immune response, i.e., dendritic cells (DCs), is relatively unknown. Therefore, we examined the effect of SFA on the differentiation and function of human monocyte-derived DCs. Unlike the well-known cyclophilin A-binding immunosuppressant cyclosporin A, which did not affect DC phenotype, differentiation of DCs in the presence of SFA resulted in CD14-CD1a DCs with normal DC morphology, viability, and a proper capacity to activate allogeneic T cells. However, DCs generated in the presence of SFA demonstrated reduced macropinocytosis and lectin-mediated endocytosis, which was in line with a decreased expression of C-type lectins, including mannose receptor, C1qRP, DC-ASGPR, and especially, DC-SIGN. In contrast, FcalphaRI (CD89) and FcgammaRII (CD32) were increased by SFA. The explicit effect of SFA on the expression of Ag uptake receptors and Ag capture by DCs makes SFA unique among immunophilin-binding immunosuppressive drugs.


Asunto(s)
Ciclofilinas/metabolismo , Células Dendríticas/efectos de los fármacos , Células Dendríticas/metabolismo , Endocitosis/efectos de los fármacos , Endocitosis/inmunología , Fluoresceína-5-Isotiocianato/análogos & derivados , Inmunosupresores/farmacología , Lactonas/farmacología , Receptores de Antígenos/metabolismo , Compuestos de Espiro/farmacología , Antígenos CD/biosíntesis , Diferenciación Celular/inmunología , Células Cultivadas , Ciclosporina/metabolismo , Ciclosporina/farmacología , Células Dendríticas/citología , Células Dendríticas/inmunología , Dextranos/antagonistas & inhibidores , Dextranos/metabolismo , Fluoresceína-5-Isotiocianato/metabolismo , Humanos , Inmunofenotipificación , Lactonas/metabolismo , Lectinas Tipo C/antagonistas & inhibidores , Lectinas Tipo C/biosíntesis , Activación de Linfocitos/efectos de los fármacos , Activación de Linfocitos/inmunología , Prueba de Cultivo Mixto de Linfocitos , Manosa/antagonistas & inhibidores , Manosa/metabolismo , Receptores de Antígenos/biosíntesis , Receptores Fc/biosíntesis , Receptores de IgG/biosíntesis , Albúmina Sérica/antagonistas & inhibidores , Albúmina Sérica/metabolismo , Compuestos de Espiro/metabolismo , Linfocitos T/inmunología , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/inmunología
11.
Blood ; 101(4): 1439-45, 2003 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-12393532

RESUMEN

The longevity of dendritic cells (DCs) is a critical regulatory factor influencing the outcome of immune responses. Recently, we demonstrated that the immunosuppressive drug rapamycin (Rapa) specifically induces apoptosis in DCs but not in other myeloid cell types. The present study unraveled the mechanism used by Rapa to induce apoptosis in human monocyte-derived DCs. Our data demonstrate that granulocyte-macrophage colony-stimulating factor (GM-CSF) preserves DC survival specifically via the phosphatidylinositol-3 lipid kinase/mammalian target of rapamycin (PI3K/mTOR) signaling pathway, which is abrogated by Rapa at the level of mTOR. Disruption of this GM-CSF signaling pathway induced loss of mitochondrial membrane potential, phosphatidyl-serine exposure, and nuclear changes. Apoptosis of these nonproliferating DCs was preceded by an up-regulation of the cell cycle inhibitor p27(KIP1). Overexpression of p27(KIP1) in DCs using adenoviral gene transduction revealed that apoptosis is directly regulated by p27(KIP1). Furthermore, both overexpression of p27(KIP1) and disruption of the GM-CSF/PI3K/mTOR signaling pathway decreased the expression of the antiapoptotic protein mcl-1. This mTOR/p27(KIP1)/mcl-1 survival seems unique for DCs and may provide novel opportunities to influence immune responses by specific interference with the life span of these cells.


Asunto(s)
Apoptosis/efectos de los fármacos , Proteínas de Ciclo Celular/genética , Células Dendríticas/efectos de los fármacos , Factor Estimulante de Colonias de Granulocitos y Macrófagos/fisiología , Inmunosupresores/farmacología , Proteínas Proto-Oncogénicas c-bcl-2 , Sirolimus/farmacología , Proteínas Supresoras de Tumor/genética , Adenoviridae/genética , Proteínas de Ciclo Celular/fisiología , Supervivencia Celular , Inhibidor p27 de las Quinasas Dependientes de la Ciclina , Células Dendríticas/citología , Células Dendríticas/fisiología , Inhibidores Enzimáticos/farmacología , Expresión Génica , Factor Estimulante de Colonias de Granulocitos y Macrófagos/administración & dosificación , Humanos , Monocitos/fisiología , Proteína 1 de la Secuencia de Leucemia de Células Mieloides , Proteínas de Neoplasias/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Proteínas Quinasas/metabolismo , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR , Transfección , Proteínas Supresoras de Tumor/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA