Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Immunology ; 171(2): 198-211, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-37884280

RESUMEN

Glioblastoma, isocitrate dehydrogenase-wildtype (GB), is the most common and aggressive primary brain malignancy with poor outcome. Immune checkpoint inhibitors (ICIs) have been tested in GB and, despite disappointing results, the identification of a small subgroup of responders underlies the need to improve our understanding of the tumour microenvironment (TME) immunity. This study aimed to determine whether the expression of selected immune checkpoints on tissue-resident memory T cells (Trm) may predict patient outcome. We conducted a single cohort observational study. Tumour samples were collected from 45 patients with histologically confirmed GB (WHO grade 4) and processed to obtain single-cell suspensions. Patients were assessed for the correlation of Trm phenotype with overall survival (OS) or progression-free survival (PFS) using multiparametric flow cytometry and uni/multivariate analyses. Levels of Trm expressing programmed cell death protein 1 (PD1) and T cell immunoglobulin and mucin domain-containing protein 3 (TIM3) were found to be linked to clinical outcome. Low frequency of Trm expressing PD1 or TIM3 or both markers defined subgroups as independent positive prognostic factors for patient survival. On multivariate analysis, low CD8+CD103+PD1+TIM3+ Trm and Karnofsky performance status (KPS) ≥70 were confirmed to be the most predictive independent factors associated with longer OS (hazard ratios-HR [95%CI]: 0.14 [0.04-0.52] p < 0.001, 0.39 [0.16-0.96] p = 0.04, respectively). The CD8+CD103+ Trm subgroups were also age-related predictors for survival in GB.


Asunto(s)
Glioblastoma , Receptor 2 Celular del Virus de la Hepatitis A , Humanos , Receptor de Muerte Celular Programada 1/metabolismo , Pronóstico , Linfocitos T CD8-positivos , Microambiente Tumoral
2.
Int J Cancer ; 153(5): 1080-1095, 2023 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-37293858

RESUMEN

BRAFV600 mutations are the most common oncogenic alterations in melanoma cells, supporting proliferation, invasion, metastasis and immune evasion. In patients, these aberrantly activated cellular pathways are inhibited by BRAFi whose potent antitumor effect and therapeutic potential are dampened by the development of resistance. Here, by using primary melanoma cell lines, generated from lymph node lesions of metastatic patients, we show that the combination of two FDA-approved drugs, the histone deacetylate inhibitor (HDCAi) romidepsin and the immunomodulatory agent IFN-α2b, reduces melanoma proliferation, long-term survival and invasiveness and overcomes acquired resistance to the BRAFi vemurafenib (VEM). Targeted resequencing revealed that each VEM-resistant melanoma cell line and the parental counterpart are characterized by a distinctive and similar genetic fingerprint, shaping the differential and specific antitumor modulation of MAPK/AKT pathways by combined drug treatment. By using RNA-sequencing and functional in vitro assays, we further report that romidepsin-IFN-α2b treatment restores epigenetically silenced immune signals, modulates MITF and AXL expression and induces both apoptosis and necroptosis in sensitive and VEM-resistant primary melanoma cells. Moreover, the immunogenic potential of drug-treated VEM-resistant melanoma cells results significantly enhanced, given the increased phagocytosis rate of these cells by dendritic cells, which in turn exhibit also a selective down-modulation of the immune checkpoint TIM-3. Overall, our results provide evidence that combined epigenetic-immune drugs can overcome VEM resistance of primary melanoma cells by oncogenic and immune pathways reprogramming, and pave the way for rapidly exploiting this combination to improve BRAFi-resistant metastatic melanoma treatment, also via reinforcement of immune checkpoint inhibitor therapy.


Asunto(s)
Interferón Tipo I , Melanoma , Humanos , Vemurafenib/farmacología , Vemurafenib/uso terapéutico , Resistencia a Antineoplásicos , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas B-raf/genética , Melanoma/tratamiento farmacológico , Melanoma/genética , Melanoma/patología , Línea Celular Tumoral
3.
PLoS One ; 13(1): e0189477, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29320502

RESUMEN

Individuals exposed to Mycobacterium tuberculosis (Mtb) may be infected and remain for the entire life in this condition defined as latent tuberculosis infection (LTBI) or develop active tuberculosis (TB). Among the multiple factors governing the outcome of the infection, dendritic cells (DCs) play a major role in dictating antibacterial immunity. However, current knowledge on the role of the diverse components of human DCs in shaping specific T-cell response during Mtb infection is limited. In this study, we performed a comparative evaluation of peripheral blood circulating DC subsets as well as of monocyte-derived Interferon-α DCs (IFN-DCs) from patients with active TB, subjects with LTBI and healthy donors (HD). The proportion of circulating myeloid BDCA3+ DCs (mDC2) and plasmacytoid CD123+ DCs (pDCs) declined significantly in active TB patients compared to HD, whereas the same subsets displayed a remarkable activation in LTBI subjects. Simultaneously, the differentiation of IFN-DCs from active TB patients resulted profoundly impaired compared to those from LTBI and HD individuals. Importantly, the altered developmental trait of IFN-DCs from active TB patients was associated with down-modulation of IFN-linked genes, marked changes in molecular signaling conveying antigen (Ag) presentation and full inability to induce Ag-specific T cell response. Thus, these data reveal an important role of IFN-α in determining the induction of Mtb-specific immunity.


Asunto(s)
Células Dendríticas/inmunología , Interferón-alfa/inmunología , Tuberculosis Latente/patología , Adulto , Antígenos CD/inmunología , Regulación hacia Abajo , Femenino , Humanos , Tuberculosis Latente/inmunología , Masculino , Persona de Mediana Edad , Mycobacterium tuberculosis/inmunología
4.
Cancer Immunol Res ; 5(7): 604-616, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28615266

RESUMEN

Colorectal cancer results from the progressive accumulation of genetic and epigenetic alterations. IFN signaling defects play an important role in the carcinogenesis process, in which the inability of IFN transcription regulatory factors (IRF) to access regulatory sequences in IFN-stimulated genes (ISG) in tumors and in immune cells may be pivotal. We reported that low-dose combination of two FDA-approved epidrugs, azacytidine (A) and romidepsin (R), with IFNα2 (ARI) hampers the aggressiveness of both colorectal cancer metastatic and stem cells in vivo and triggers immunogenic cell death signals that stimulate dendritic cell (DC) function. Here, we investigated the molecular signals induced by ARI treatment and found that this drug combination increased the accessibility to regulatory sequences of ISGs and IRFs that were epigenetically silenced in both colorectal cancer cells and DCs. Likewise, specific ARI-induced histone methylation and acetylation changes marked epigenetically affected ISG promoters in both metastatic cancer cells and DCs. Analysis by ChIP-seq confirmed such ARI-induced epigenetically regulated IFN signature. The activation of this signal endowed DCs with a marked migratory capability. Our results establish a direct correlation between reexpression of silenced ISGs by epigenetic control and ARI anticancer activity and provide new knowledge for the development of innovative combined therapeutic strategies for colorectal cancer. Cancer Immunol Res; 5(7); 604-16. ©2017 AACR.


Asunto(s)
Carcinogénesis/efectos de los fármacos , Neoplasias Colorrectales/tratamiento farmacológico , Células Dendríticas/efectos de los fármacos , Interferón-alfa/inmunología , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/inmunología , Azacitidina/administración & dosificación , Carcinogénesis/inmunología , Línea Celular Tumoral , Neoplasias Colorrectales/inmunología , Neoplasias Colorrectales/patología , Células Dendríticas/inmunología , Depsipéptidos/administración & dosificación , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/inmunología , Humanos , Factor 7 Regulador del Interferón/genética , Factores Reguladores del Interferón/genética , Interferón-alfa/genética , Interferón gamma/genética , Óxido Nítrico Sintasa de Tipo II/genética , Receptores de Citocinas/genética , Receptores de Interferón , Transducción de Señal/efectos de los fármacos
5.
Sci Rep ; 7(1): 1093, 2017 04 24.
Artículo en Inglés | MEDLINE | ID: mdl-28439087

RESUMEN

Immunotherapy efficacy relies on the crosstalk within the tumor microenvironment between cancer and dendritic cells (DCs) resulting in the induction of a potent and effective antitumor response. DCs have the specific role of recognizing cancer cells, taking up tumor antigens (Ags) and then migrating to lymph nodes for Ag (cross)-presentation to naïve T cells. Interferon-α-conditioned DCs (IFN-DCs) exhibit marked phagocytic activity and the special ability of inducing Ag-specific T-cell response. Here, we have developed a novel microfluidic platform recreating tightly interconnected cancer and immune systems with specific 3D environmental properties, for tracking human DC behaviour toward tumor cells. By combining our microfluidic platform with advanced microscopy and a revised cell tracking analysis algorithm, it was possible to evaluate the guided efficient motion of IFN-DCs toward drug-treated cancer cells and the succeeding phagocytosis events. Overall, this platform allowed the dissection of IFN-DC-cancer cell interactions within 3D tumor spaces, with the discovery of major underlying factors such as CXCR4 involvement and underscored its potential as an innovative tool to assess the efficacy of immunotherapeutic approaches.


Asunto(s)
Rastreo Celular/métodos , Neoplasias del Colon/terapia , Células Dendríticas/inmunología , Inmunoterapia/métodos , Microfluídica/métodos , Modelos Biológicos , Células Cultivadas , Humanos , Factores Inmunológicos/metabolismo , Interferón-alfa/metabolismo , Microscopía/métodos , Resultado del Tratamiento
6.
Oncotarget ; 7(18): 26361-73, 2016 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-27028869

RESUMEN

Epigenetic alterations, including dysregulated DNA methylation and histone modifications, govern the progression of colorectal cancer (CRC). Cancer cells exploit epigenetic regulation to control cellular pathways, including apoptotic and metastatic signals. Since aberrations in epigenome can be pharmacologically reversed by DNA methyltransferase and histone deacetylase inhibitors, epigenetics in combination with standard agents are currently envisaged as a new therapeutic frontier in cancer, expected to overcome drug resistance associated with current treatments. In this study, we challenged this idea and demonstrated that the combination of azacitidine and romidepsin with IFN-α owns a high therapeutic potential, targeting the most aggressive cellular components of CRC, such as metastatic cells and cancer stem cells (CSCs), via tight control of key survival and death pathways. Moreover, the antitumor efficacy of this novel pharmacological approach is associated with induction of signals of immunogenic cell death. Of note, a previously undisclosed key role of IFN-α in inducing both antiproliferative and pro-apoptotic effects on CSCs of CRC was also found. Overall, these findings open a new frontier on the suitability of IFN-α in association with epigenetics as a novel and promising therapeutic approach for CRC management.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Neoplasias Colorrectales/patología , Interferón-alfa/farmacología , Células Madre Neoplásicas/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Azacitidina/farmacología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Depsipéptidos/farmacología , Epigénesis Genética/efectos de los fármacos , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Células Madre Neoplásicas/patología , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Mol Cell Neurosci ; 45(3): 234-44, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20600925

RESUMEN

Fine regulation of the innate immune response following brain injury or infection is important to avoid excessive activation of microglia and its detrimental consequences on neural cell viability and function. To get insights on the molecular networks regulating microglia activation, we analyzed expression, regulation and functional relevance of tumor necrosis factor receptors (TNFR) 2 in cultured mouse microglia. We found that microglia upregulate TNFR2 mRNA and protein and shed large amounts of soluble TNFR2, but not TNFR1, in response to pro-inflammatory stimuli and through activation of TNFR2 itself. By microarray analysis, we demonstrate that TNFR2 stimulation in microglia regulates expression of genes involved in immune processes, including molecules with anti-inflammatory and neuroprotective function like granulocyte colony-stimulating factor, adrenomedullin and IL-10. In addition, we identify IFN-γ as a regulator of the balance between pro- and anti-inflammatory/neuroprotective factors induced by TNFR2 stimulation. These data indicate that, through TNFR2, microglia may contribute to the counter-regulatory response activated in neuropathological conditions.


Asunto(s)
Inflamación/inmunología , Microglía/metabolismo , Receptores Tipo II del Factor de Necrosis Tumoral/metabolismo , Transducción de Señal/fisiología , Animales , Células Cultivadas , Regulación de la Expresión Génica , Factor Estimulante de Colonias de Granulocitos/metabolismo , Interferón gamma/inmunología , Interleucina-10/metabolismo , Ratones , Análisis por Micromatrices , Microglía/citología , Receptores Tipo I de Factores de Necrosis Tumoral/genética , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Receptores Tipo II del Factor de Necrosis Tumoral/genética , Factor de Necrosis Tumoral alfa/metabolismo
8.
Blood ; 115(8): 1554-63, 2010 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-20009034

RESUMEN

The identification of molecules responsible for apoptotic cell (AC) uptake by dendritic cells (DCs) and induction of T-cell immunity against AC-associated antigens is a challenge in immunology. DCs differentiated in the presence of interferon-alpha (IFN-alpha-conditioned DCs) exhibit a marked phagocytic activity and a special attitude in inducing CD8(+) T-cell response. In this study, we found marked overexpression of the scavenger receptor oxidized low-density lipoprotein receptor 1 (LOX-1) in IFN-alpha-conditioned DCs, which was associated with increased levels of genes belonging to immune response families and high competence in inducing T-cell immunity against antigens derived from allogeneic apoptotic lymphocytes. In particular, the capture of ACs by IFN-alpha DCs led to a substantial subcellular rearrangement of major histocompatibility complex class I and class II molecules, along with enhanced cross-priming of autologous CD8(+) T cells and CD4(+) T-cell activation. Remarkably, AC uptake, CD8(+) T-cell cross-priming, and, to a lesser extent, priming of CD4(+) T lymphocytes were inhibited by a neutralizing antibody to the scavenger receptor LOX-1 protein. These results unravel a novel LOX-1-dependent pathway by which IFN-alpha can, under both physiologic and pathologic conditions, render DCs fully competent for presenting AC-associated antigens for cross-priming CD8(+) effector T cells, concomitantly with CD4(+) T helper cell activation.


Asunto(s)
Presentación de Antígeno/inmunología , Apoptosis/efectos de los fármacos , Células Dendríticas/inmunología , Factores Inmunológicos/farmacología , Interferón-alfa/farmacología , Receptores Depuradores de Clase E/inmunología , Transducción de Señal/efectos de los fármacos , Apoptosis/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Células Cultivadas , Humanos , Inmunidad Celular/efectos de los fármacos , Factores Inmunológicos/inmunología , Interferón-alfa/inmunología , Activación de Linfocitos/efectos de los fármacos , Transducción de Señal/inmunología
9.
Clin Cancer Res ; 13(2 Pt 1): 644-53, 2007 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-17255288

RESUMEN

PURPOSE: Immunotherapy is a promising antitumor strategy, which can be successfully combined with current anticancer treatments, as suggested by recent studies showing the paradoxical chemotherapy-induced enhancement of the immune response. The purpose of the present work is to dissect the biological events induced by chemotherapy that cooperate with immunotherapy in the success of the combined treatment against cancer. In particular, we focused on the following: (a) cyclophosphamide-induced modulation of several cytokines, (b) homeostatic proliferation of adoptively transferred lymphocytes, and (c) homing of transferred lymphocytes to secondary lymphoid organs and tumor mass. EXPERIMENTAL DESIGN: Here, we used the adoptive transfer of tumor-immune cells after cyclophosphamide treatment of tumor-bearing mice as a model to elucidate the mechanisms by which cyclophosphamide can render the immune lymphocytes competent to induce tumor rejection. RESULTS: The transfer of antitumor immunity was found to be dependent on CD4(+) T cells and on the cooperation of adoptively transferred cells with the host immune system. Of note, tumor-immune lymphocytes migrated specifically to the tumor only in mice pretreated with cyclophosphamide. Cyclophosphamide treatment also promoted homeostatic proliferation/activation of transferred B and T lymphocytes. Optimal therapeutic responses to the transfer of immune cells were associated with the cyclophosphamide-mediated induction of a "cytokine storm" [including granulocyte macrophage colony-stimulating factor, interleukin (IL)-1beta, IL-7, IL-15, IL-2, IL-21, and IFN-gamma], occurring during the "rebound phase" after drug-induced lymphodepletion. CONCLUSIONS: The ensemble of these data provides a new rationale for combining immunotherapy and chemotherapy to induce an effective antitumor response in cancer patients.


Asunto(s)
Antineoplásicos/farmacología , Linfocitos B/metabolismo , Ciclofosfamida/farmacología , Citocinas/metabolismo , Linfocitos T/metabolismo , Animales , Linfocitos T CD4-Positivos/metabolismo , Línea Celular Tumoral , Proliferación Celular , Sistema Inmunológico , Inmunoterapia/métodos , Interferón gamma/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones SCID , Trasplante de Neoplasias
10.
Vaccine ; 24 Suppl 2: S2-56-7, 2006 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-16823927

RESUMEN

Type I IFN is a cytokine family endowed with multiple biological activities. In recent years, type I IFN has been demonstrated to play a crucial role in innate immunity, in dendritic cell maturation/differentiation and in the priming of primary antibody responses, especially when administered i.m. with a purified influenza vaccine. Due to the increasing interest in mucosal vaccination especially for respiratory infections, we investigated two different IFN-adjuvanted immunization protocols against influenza. In the first one, anesthesized C3H/HeN mice were instilled intranasally with 50 microl of a commercially available influenza vaccine containing 5 mg of hemagglutinin (HA) and a partially purified preparation of mouse IFN-alphal. A single intranasal administration of IFN-adjuvanted vaccine resulted in a full protection of 100% of mice against virus challenge while vaccine alone was only partially effective (40%). From the analysis of the specific antibody response emerged that type I IFN induced a significant increase of antibody titers in all the Ig subclasses with particular effect on IgG2a and IgA. To mimic aerosol administration and to limit vaccine delivery strictly to nasal mucosa, a second method of intranasal vaccination was developed in which mice were left awake and their nostrils moistened along the day of treatment (every 10 min) with six mini-doses (8 microl each) of vaccine +/- IFN-I with dose and timing equivalent to previous immunization protocol. This vaccination schedule prevented mice from pulmonary damage and the concomintant use of type I IFN induced an efficient and long lasting both local and systemic immune response. These findings shed new light on the involvement of type I IFN in the early phases of the immune response and open new and practical perspectives in vaccine research.


Asunto(s)
Adyuvantes Inmunológicos/farmacología , Vacunas contra la Influenza/inmunología , Interferón Tipo I/farmacología , Adyuvantes Inmunológicos/administración & dosificación , Administración Intranasal , Animales , Formación de Anticuerpos , Inmunidad Mucosa , Vacunas contra la Influenza/administración & dosificación , Interferón Tipo I/administración & dosificación , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL
11.
Blood ; 108(2): 609-17, 2006 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-16569763

RESUMEN

Interferon consensus sequence-binding protein (ICSBP)/interferon regulatory factor 8 (IRF-8) is a transcription factor that plays critical roles in the differentiation of defined dendritic-cell (DC) populations and in the immune response to many pathogens. In this study, we show that splenic DCs (s-DCs) from ICSBP(-/-) mice are markedly defective in their ability to capture and to present exogenous antigens (Ags) to naive CD4(+) T lymphocytes. We found that CD8alpha(+) DCs and, to a lesser extent, CD8alpha(-) DCs from ICSBP(-/-) mice are impaired at internalizing Ags, either through a receptor-mediated pathway or by macropinocytosis, in spite of having a more immature phenotype than their wild-type (WT) counterparts. These features reflected a greatly impaired ability of ICSBP(-/-) s-DCs to present injected soluble ovalbumin (OVA) to OVA-specific CD4(+) T cells in vivo. Conversely, bone marrow (BM)-derived DCs from ICSBP(-/-) mice, in keeping with their immature phenotype, exhibited higher endocytic activity than WT cells. However, Ag-loaded ICSBP(-/-) BM-DCs were defective in priming Ag-specific CD4(+) T lymphocytes and failed to induce a contact hypersensitivity (CHS) response when injected into competent WT hosts. Together, these results indicate that, throughout the developmental program of DCs, ICSBP differentially controls Ag uptake and MHC class II (MHC-II) presentation affecting both functions only in differentiated peripheral DCs.


Asunto(s)
Presentación de Antígeno , Linfocitos T CD4-Positivos/inmunología , Células Dendríticas/citología , Factores Reguladores del Interferón/fisiología , Animales , Células Cultivadas , Células Dendríticas/inmunología , Antígenos de Histocompatibilidad Clase II/inmunología , Factores Reguladores del Interferón/deficiencia , Factores Reguladores del Interferón/inmunología , Ratones , Ratones Noqueados , Pinocitosis , Bazo/citología
12.
Vaccine ; 23(23): 2994-3004, 2005 Apr 27.
Artículo en Inglés | MEDLINE | ID: mdl-15811645

RESUMEN

In view of the increasing interest in mucosal vaccination, we investigated whether type I IFN could act as adjuvant of an intranasally administered influenza vaccine. A single intranasal administration of IFNalphabeta-adjuvanted vaccine in anesthetized C3H/HeN mice was capable of protecting the totality of animals against virus challenge, while vaccine alone was only partially effective. To mimic intranasal vaccine administration in man and to limit vaccine delivery strictly to nasal mucosa, we used a second method of vaccination based on vaccine fractionation in six doses and intranasal instillation in non-anesthetized mice. By using this vaccination schedule, IFNalphabeta-adjuvanted vaccine also prevented mice from disease development and induced an efficient long lasting immune response. Further experiments showed that IFNalphabeta increased the percentage of antigen-associated phagocytes in the nasal mucus layer, thus suggesting a new possible mechanism of action for type I IFN as an adjuvant.


Asunto(s)
Adyuvantes Inmunológicos/administración & dosificación , Antígenos Virales/inmunología , Vacunas contra la Influenza/administración & dosificación , Interferón Tipo I/administración & dosificación , Administración Intranasal , Animales , Anticuerpos Antivirales/biosíntesis , Relación Dosis-Respuesta Inmunológica , Inmunidad Mucosa , Inmunoglobulina A Secretora/biosíntesis , Ratones , Ratones Endogámicos C3H , Vacunación
13.
Blood ; 102(7): 2334-7, 2003 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-12805068

RESUMEN

CC chemokine ligand 2 (CCL2) is constitutively expressed at high levels in human peripheral blood monocytes, and its expression is further up-modulated during their differentiation into macrophages as well as in the course of HIV infection. To investigate the role of endogenous CCL2 on HIV replication and macrophage function, CCL2's activity was neutralized by specific antibodies. Infection of monocyte-derived macrophages with laboratory-adapted HIV-1 or primary viral isolates in the continuous presence of anti-CCL2 antibody resulted in significantly lower p24 Gag antigen release with respect to control cultures. Interestingly, CCL2 neutralization did not affect the early steps of the HIV life cycle but resulted in the intracellular accumulation of p24 Gag antigen. Simultaneously, remarkable changes in cell morphology and size occurred in cell cultures maintained in the presence of anti-CCL2 antibody. These results suggest that CCL2 may represent an autocrine factor important for enhancing virion production likely by affecting the macrophage cytoskeleton.


Asunto(s)
Quimiocina CCL2/metabolismo , Infecciones por VIH/inmunología , VIH-1/crecimiento & desarrollo , Macrófagos/virología , Comunicación Autocrina/inmunología , Células Cultivadas , Citoesqueleto/metabolismo , Infecciones por VIH/metabolismo , Humanos , Macrófagos/citología , Monocitos/citología , Virión/crecimiento & desarrollo , Replicación Viral
14.
J Immunol ; 169(6): 3038-45, 2002 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-12218119

RESUMEN

Type I IFNs are modulators of myeloid dendritic cell (DC) development, survival, and functional activities. Here we monitored the signal transduction pathway underlying type I IFN biological activities during in vitro maturation of human monocyte-derived DCs. IFN-inducible tyrosine phosphorylation of STAT family members was severely impaired upon LPS-induced DC maturation. This correlated with a marked reduction of both type I IFN receptor chains occurring as early as 4 h after LPS treatment. The reduced receptor expression was a post-transcriptional event only partially mediated by ligand-induced internalization/degradation. In fact, although an early and transient production of type I IFNs was observed after LPS treatment, its neutralization was not sufficient to completely rescue IFN receptor expression. Notably, neutralization of LPS-induced, endogenous type I IFNs did not interfere with the acquisition of a fully mature surface phenotype, nor did it have a significant effect on the allostimulatory properties of LPS-stimulated DCs. Overall, these data indicate that DCs strictly modulate their responsiveness to type I IFNs as part of their maturation program, underlining the importance of the IFN system in the regulation of DC physiology.


Asunto(s)
Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Regulación hacia Abajo/inmunología , Interferón Tipo I/fisiología , Receptores de Interferón/antagonistas & inhibidores , Receptores de Interferón/biosíntesis , Diferenciación Celular/inmunología , Células Cultivadas , Humanos , Inmunofenotipificación , Interferón Tipo I/antagonistas & inhibidores , Interferón Tipo I/metabolismo , Lipopolisacáridos/antagonistas & inhibidores , Lipopolisacáridos/farmacología , Proteínas de la Membrana , Monocitos/citología , Monocitos/inmunología , Monocitos/metabolismo , Procesamiento Postranscripcional del ARN/inmunología , Receptor de Interferón alfa y beta , Receptores de Interferón/fisiología , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...