Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 70
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Nat Immunol ; 20(8): 1012-1022, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31263276

RESUMEN

The plasma membrane tetraspan molecule MS4A4A is selectively expressed by macrophage-lineage cells, but its function is unknown. Here we report that MS4A4A was restricted to murine and human mononuclear phagocytes and was induced during monocyte-to-macrophage differentiation in the presence of interleukin 4 or dexamethasone. Human MS4A4A was co-expressed with M2/M2-like molecules in subsets of normal tissue-resident macrophages, infiltrating macrophages from inflamed synovium and tumor-associated macrophages. MS4A4A interacted and colocalized with the ß-glucan receptor dectin-1 in lipid rafts. In response to dectin-1 ligands, Ms4a4a-deficient macrophages showed defective signaling and defective production of effector molecules. In experimental models of tumor progression and metastasis, Ms4a4a deficiency in macrophages had no impact on primary tumor growth, but was essential for dectin-1-mediated activation of macrophages and natural killer (NK) cell-mediated metastasis control. Thus, MS4A4A is a tetraspan molecule selectively expressed in macrophages during differentiation and polarization, essential for dectin-1-dependent activation of NK cell-mediated resistance to metastasis.


Asunto(s)
Células Asesinas Naturales/inmunología , Lectinas Tipo C/metabolismo , Macrófagos/inmunología , Proteínas de la Membrana/metabolismo , Metástasis de la Neoplasia/inmunología , Neoplasias/inmunología , Animales , Diferenciación Celular/inmunología , Linaje de la Célula , Dexametasona/farmacología , Humanos , Interleucina-4/metabolismo , Activación de Linfocitos/inmunología , Activación de Macrófagos/inmunología , Macrófagos/citología , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Metástasis de la Neoplasia/prevención & control , Neoplasias/patología
2.
Immunity ; 53(2): 303-318.e5, 2020 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-32579887

RESUMEN

Granulocyte-monocyte progenitors (GMPs) have been previously defined for their potential to generate various myeloid progenies such as neutrophils and monocytes. Although studies have proposed lineage heterogeneity within GMPs, it is unclear if committed progenitors already exist among these progenitors and how they may behave differently during inflammation. By combining single-cell transcriptomic and proteomic analyses, we identified the early committed progenitor within the GMPs responsible for the strict production of neutrophils, which we designate as proNeu1. Our dissection of the GMP hierarchy led us to further identify a previously unknown intermediate proNeu2 population. Similar populations could be detected in human samples. proNeu1s, but not proNeu2s, selectively expanded during the early phase of sepsis at the expense of monocytes. Collectively, our findings help shape the neutrophil maturation trajectory roadmap and challenge the current definition of GMPs.


Asunto(s)
Células Precursoras de Granulocitos/citología , Monocitos/citología , Mielopoyesis/fisiología , Neutrófilos/citología , Animales , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Análisis de la Célula Individual
3.
Nat Immunol ; 17(1): 34-40, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26681460

RESUMEN

Macrophages have protective roles in immunity to pathogens, tissue development, homeostasis and repair following damage. Maladaptive immunity and inflammation provoke changes in macrophage function that are causative of disease. Despite a historical wealth of knowledge about macrophages, recent advances have revealed unknown aspects of their development and function. Following development, macrophages are activated by diverse signals. Such tissue microenvironmental signals together with epigenetic changes influence macrophage development, activation and functional diversity, with consequences in disease and homeostasis. We discuss here how recent discoveries in these areas have led to a multidimensional concept of macrophage ontogeny, activation and function. In connection with this, we also discuss how technical advances facilitate a new roadmap for the isolation and analysis of macrophages at high resolution.


Asunto(s)
Activación de Macrófagos/inmunología , Macrófagos , Animales , Humanos
4.
Immunity ; 48(2): 364-379.e8, 2018 02 20.
Artículo en Inglés | MEDLINE | ID: mdl-29466759

RESUMEN

Neutrophils are specialized innate cells that require constant replenishment from proliferative bone marrow (BM) precursors as a result of their short half-life. Although it is established that neutrophils are derived from the granulocyte-macrophage progenitor (GMP), the differentiation pathways from GMP to functional mature neutrophils are poorly defined. Using mass cytometry (CyTOF) and cell-cycle-based analysis, we identified three neutrophil subsets within the BM: a committed proliferative neutrophil precursor (preNeu) which differentiates into non-proliferating immature neutrophils and mature neutrophils. Transcriptomic profiling and functional analysis revealed that preNeu require the C/EBPε transcription factor for their generation from the GMP, and their proliferative program is substituted by a gain of migratory and effector function as they mature. preNeus expand under microbial and tumoral stress, and immature neutrophils are recruited to the periphery of tumor-bearing mice. In summary, our study identifies specialized BM granulocytic populations that ensure supply under homeostasis and stress responses.


Asunto(s)
Células de la Médula Ósea/fisiología , Neutrófilos/fisiología , Animales , Células de la Médula Ósea/inmunología , Proteínas Potenciadoras de Unión a CCAAT/fisiología , Linaje de la Célula , Movimiento Celular , Proliferación Celular , Células Cultivadas , Perfilación de la Expresión Génica , Humanos , Ratones , Neoplasias Experimentales/inmunología , Neutrófilos/inmunología
5.
Immunity ; 43(3): 435-49, 2015 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-26377897

RESUMEN

Immune cells play a key role in host defense against infection and cancer. Upon encountering danger signals, these cells undergo activation leading to a modulation in their immune functions. However, recent studies reveal that immune cells upon activation also show distinct metabolic changes that impact their immune functions. Such metabolic reprogramming and its functional effects are well known for cancer cells. Given that immune cells have emerged as crucial players in cancer progression, it is important to understand whether immune cells also undergo metabolic reprogramming in tumors and how this might affect their contribution in cancer progression. This emerging aspect of tumor-associated immune cells is reviewed here, discussing metabolic reprogramming of different immune cell types, the key pathways involved, and its impact on tumor progression.


Asunto(s)
Sistema Inmunológico/inmunología , Sistema Inmunológico/metabolismo , Neoplasias/inmunología , Neoplasias/metabolismo , Animales , Progresión de la Enfermedad , Metabolismo Energético/genética , Metabolismo Energético/inmunología , Regulación Neoplásica de la Expresión Génica/inmunología , Humanos , Sistema Inmunológico/patología , Redes y Vías Metabólicas/genética , Redes y Vías Metabólicas/inmunología , Modelos Inmunológicos , Neoplasias/genética
6.
Immunity ; 42(3): 484-98, 2015 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-25746953

RESUMEN

Sepsis is characterized by a dysregulated inflammatory response to infection. Despite studies in mice, the cellular and molecular basis of human sepsis remains unclear and effective therapies are lacking. Blood monocytes serve as the first line of host defense and are equipped to recognize and respond to infection by triggering an immune-inflammatory response. However, the response of these cells in human sepsis and their contribution to sepsis pathogenesis is poorly understood. To investigate this, we performed a transcriptomic, functional, and mechanistic analysis of blood monocytes from patients during sepsis and after recovery. Our results revealed the functional plasticity of monocytes during human sepsis, wherein they transited from a pro-inflammatory to an immunosuppressive phenotype, while enhancing protective functions like phagocytosis, anti-microbial activity, and tissue remodeling. Mechanistically, hypoxia inducible factor-1α (HIF1α) mediated this functional re-programming of monocytes, revealing a potential mechanism for their therapeutic targeting to regulate human sepsis.


Asunto(s)
Reprogramación Celular/inmunología , Subunidad alfa del Factor 1 Inducible por Hipoxia/inmunología , Huésped Inmunocomprometido , Quinasas Asociadas a Receptores de Interleucina-1/inmunología , Sepsis/inmunología , Inmunidad Adaptativa , Convalecencia , Citocinas/genética , Citocinas/inmunología , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Inmunidad Innata , Quinasas Asociadas a Receptores de Interleucina-1/genética , Monocitos/inmunología , Monocitos/patología , Fagocitosis , Sepsis/genética , Sepsis/patología , Transducción de Señal , Transcriptoma/inmunología
7.
Proc Natl Acad Sci U S A ; 118(25)2021 06 22.
Artículo en Inglés | MEDLINE | ID: mdl-34155144

RESUMEN

Aberrant inflammation, such as that associated with inflammatory bowel disease (IBD), is fueled by the inordinate activity of RelA/NF-κB factors. As such, the canonical NF-κB module mediates controlled nuclear activation of RelA dimers from the latent cytoplasmic complexes. What provokes pathological RelA activity in the colitogenic gut remains unclear. The noncanonical NF-κB pathway typically promotes immune organogenesis involving Nfkb2 gene products. Because NF-κB pathways are intertwined, we asked whether noncanonical signaling aggravated inflammatory RelA activity. Our investigation revealed frequent engagement of the noncanonical pathway in human IBD. In a mouse model of experimental colitis, we established that Nfkb2-mediated regulations escalated the RelA-driven proinflammatory gene response in intestinal epithelial cells, exacerbating the infiltration of inflammatory cells and colon pathologies. Our mechanistic studies clarified that cell-autonomous Nfkb2 signaling supplemented latent NF-κB dimers, leading to a hyperactive canonical RelA response in the inflamed colon. In sum, the regulation of latent NF-κB dimers appears to link noncanonical Nfkb2 signaling to RelA-driven inflammatory pathologies and may provide for therapeutic targets.


Asunto(s)
Inflamación/patología , Intestinos/patología , Subunidad p52 de NF-kappa B/metabolismo , FN-kappa B/metabolismo , Multimerización de Proteína , Transducción de Señal , Factor de Transcripción ReIA/metabolismo , Animales , Colitis/metabolismo , Colitis/patología , Progresión de la Enfermedad , Células Epiteliales/metabolismo , Homeostasis , Humanos , Inflamación/metabolismo , Enfermedades Inflamatorias del Intestino/metabolismo , Enfermedades Inflamatorias del Intestino/patología , Receptor beta de Linfotoxina/metabolismo , Ratones Endogámicos C57BL , Modelos Biológicos , Subunidad p52 de NF-kappa B/deficiencia , Células del Estroma/metabolismo
8.
Immunity ; 41(5): 815-29, 2014 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-25453823

RESUMEN

Monocytes and macrophages are major components of the tumor microenvironment, but their contributions to human cancer are poorly understood. We used molecular profiling combined with functional assays to investigate the role of these cells in human renal cell carcinoma (RCC). Blood monocytes from RCC patients displayed a tumor-promoting transcriptional profile that supported functions like angiogenesis and invasion. Induction of this protumor phenotype required an interleukin-1 receptor (IL-1R)-dependent mechanism. Indeed, targeting of IL-1-IL-1R axis in a human RCC xenograft model abrogated the protumor phenotype of tumor-associated macrophages (TAMs) and reduced tumor growth in vivo. Supporting this, meta-analysis of gene expression from human RCC tumors showed IL1B expression to correlate with myelomonocytic markers, protumor genes, and tumor staging. Analyzing RCC patient tumors confirmed the protumor phenotype of TAMs. These data provide direct evidence for a tumor-promoting role of monocytes and macrophages in human cancer and indicate IL-1-IL-1R as a possible therapeutic target.


Asunto(s)
Carcinoma de Células Renales/inmunología , Interleucina-1beta/inmunología , Macrófagos/inmunología , Monocitos/inmunología , Receptores de Interleucina-1/inmunología , Animales , Proliferación Celular/genética , Citocinas/biosíntesis , Citocinas/inmunología , Perfilación de la Expresión Génica , Humanos , Inflamación/inmunología , Proteína Antagonista del Receptor de Interleucina 1/farmacología , Interleucina-1beta/antagonistas & inhibidores , Interleucina-1beta/biosíntesis , Interleucina-1beta/genética , Ratones , Ratones Noqueados , Ratones SCID , Factor 88 de Diferenciación Mieloide , Trasplante de Neoplasias , Neovascularización Patológica , Receptores de Interleucina-1/antagonistas & inhibidores , Receptores de Interleucina-1/genética , Factor de Transcripción ReIA/genética , Trasplante Heterólogo , Células Tumorales Cultivadas
9.
Immunity ; 41(1): 14-20, 2014 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-25035950

RESUMEN

Description of macrophage activation is currently contentious and confusing. Like the biblical Tower of Babel, macrophage activation encompasses a panoply of descriptors used in different ways. The lack of consensus on how to define macrophage activation in experiments in vitro and in vivo impedes progress in multiple ways, including the fact that many researchers still consider there to be only two types of activated macrophages, often termed M1 and M2. Here, we describe a set of standards encompassing three principles-the source of macrophages, definition of the activators, and a consensus collection of markers to describe macrophage activation-with the goal of unifying experimental standards for diverse experimental scenarios. Collectively, we propose a common framework for macrophage-activation nomenclature.


Asunto(s)
Activación de Macrófagos/inmunología , Macrófagos/inmunología , Terminología como Asunto , Animales , Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Guías como Asunto , Humanos , Factor Estimulante de Colonias de Macrófagos/inmunología , Ratones , Investigación
10.
Int J Cancer ; 151(3): 435-449, 2022 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-35415893

RESUMEN

Pancreatic ductal adenocarcinoma (PDAC) is a highly lethal and debilitating disease with limited therapeutic options. The aim of this clinical study was to evaluate the safety, efficacy and pharmacokinetics of a novel regimen comprised of metronomic oxaliplatin (O), chronomodulated capecitabine (X) and UGT1A1 genotype-guided dosing of irinotecan (IRI) [OXIRI] as well as its immunomodulatory effects. Thirty-six patients were enrolled into either dose-escalation or expansion cohorts. In the dose escalation phase, capecitabine doses (2000, 2650, 3500 and 4500 mg/day) were administered at midnight on days 1 to 14 while oxaliplatin and irinotecan were intravenously infused at fixed doses of 50 and 75 mg/m2 respectively on days 1, 8 in a 21-day cycle. The maximum tolerated dose of capecitabine was 2650 mg/day and the most common grade 3 adverse events were neutropenia (30.6%) and diarrhea (13.9%). No grade 4 toxicity was observed. UGT1A1-genotype directed dosing resulted in similar exposure levels of irinotecan, SN-38 and SN-38G in all patients. Objective response rate was 22.2%. Median overall survival and progression-free survival were 8.1 and 5.2 months, respectively. Exploratory immunoprofiling by flow cytometry and quantitative spatial localization analysis of infiltrated immune cells performed on biopsy and plasma samples revealed significant declines in CCL22, CCL2 and TNFα levels at end of first cycle and an active host immune response. Our study showed that OXIRI was well-tolerated and exhibited good efficacy, with immunomodulatory effects. It may be considered as an alternative to FOLFIRINOX in patients intolerant to the latter.


Asunto(s)
Adenocarcinoma , Neoplasias Pancreáticas , Adenocarcinoma/patología , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Camptotecina , Capecitabina/uso terapéutico , Fluorouracilo/uso terapéutico , Humanos , Inmunidad , Irinotecán , Oxaliplatino , Neoplasias Pancreáticas/patología , Resultado del Tratamiento , Neoplasias Pancreáticas
11.
J Clin Immunol ; 42(2): 214-229, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34716845

RESUMEN

The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants of concern (VOCs) that have become dominant as the pandemic progresses bear the ORF8 mutation together with multiple spike mutations. A 382-nucleotide deletion (Δ382) in the ORF7b and ORF8 regions has been associated with milder disease phenotype and less systemic inflammation in COVID-19 patients. However, its impact on host immunity against SARS-CoV-2 remains undefined. Here, RNA-sequencing was performed to elucidate whole blood transcriptomic profiles and identify contrasting immune signatures between patients infected with either wildtype or Δ382 SARS-CoV-2 variant. Interestingly, the immune landscape of Δ382 SARS-CoV-2 infected patients featured an increased adaptive immune response, evidenced by enrichment of genes related to T cell functionality, a more robust SARS-CoV-2-specific T cell immunity, as well as a more rapid antibody response. At the molecular level, eukaryotic initiation factor 2 signaling was found to be upregulated in patients bearing Δ382, and its associated genes were correlated with systemic levels of T cell-associated and pro-inflammatory cytokines. This study provides more in-depth insight into the host-pathogen interactions of ORF8 with great promise as a therapeutic target to combat SARS-CoV-2 infection.


Asunto(s)
Inmunidad Adaptativa/inmunología , COVID-19/inmunología , SARS-CoV-2/inmunología , Citocinas/inmunología , Interacciones Huésped-Patógeno/inmunología , Humanos , Inflamación/inmunología , Mutación/inmunología , Pandemias/prevención & control , Linfocitos T/inmunología
12.
Nat Immunol ; 11(10): 889-96, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20856220

RESUMEN

Plasticity is a hallmark of cells of the myelomonocytic lineage. In response to innate recognition or signals from lymphocyte subsets, mononuclear phagocytes undergo adaptive responses. Shaping of monocyte-macrophage function is an essential component of resistance to pathogens, tissue damage and repair. The orchestration of myelomonocytic cell function is a key element that links inflammation and cancer and provides a paradigm for macrophage plasticity and function. A better understanding of the molecular basis of myelomonocytic cell plasticity will open new vistas in immunopathology and therapeutic intervention.


Asunto(s)
Subgrupos Linfocitarios/inmunología , Macrófagos/inmunología , Neoplasias/inmunología , Animales , Citocinas/biosíntesis , Humanos , Inflamación/inmunología , Activación de Macrófagos , Macrófagos/metabolismo , Monocitos/inmunología
13.
Immunity ; 38(3): 408-10, 2013 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-23521881

RESUMEN

Monocyte-macrophage differentiation under pathological conditions is poorly understood. In the present issue of Immunity, Egawa et al. (2013) report how basophils drive the differentiation of inflammatory monocytes into M2 macrophages, thereby regulating allergic skin inflammation.

14.
Immunol Rev ; 280(1): 102-111, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-29027220

RESUMEN

Studies in the last 20 years have given us a remarkable insight into the functional and phenotypic diversity of macrophages which reflects their integral role in host defence, homeostasis and pathogenesis. Mouse genetics, transcriptomic and epigenetic studies have provided an ontogenic and molecular perspective to the phenotypic diversity of these cells. Recently, metabolic studies have revealed the crucial role of metabolism and metabolites in shaping the phenotype and function of macrophages. Evidence pertaining to this aspect will be reviewed here.


Asunto(s)
Inflamación/metabolismo , Activación de Macrófagos , Macrófagos/inmunología , Animales , Diferenciación Celular , Citocinas/metabolismo , Glucólisis , Homeostasis , Humanos , Inmunidad , Hierro/metabolismo , Metabolismo de los Lípidos , Microbiota , Fosforilación Oxidativa , Fenotipo
15.
Immunity ; 33(3): 375-86, 2010 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-20832340

RESUMEN

Monocytes are effectors of the inflammatory response to microbes. Human CD14(+) monocytes specialize in phagocytosis and production of reactive oxygen species and secrete inflammatory cytokines in response to a broad range of microbial cues. Here, we have characterized the functions of human monocytes that lack CD14 (CD14(dim)) and express CD16. CD14(dim) monocytes were genetically distinct from natural killer cells. Gene expression analyses indicated similarities with murine patrolling Gr1(dim) monocytes, and they patrolled the endothelium of blood vessels after adoptive transfer, in a lymphocyte function-associated antigen-1-dependent manner. CD14(dim) monocytes were weak phagocytes and did not produce ROS or cytokines in response to cell-surface Toll-like receptors. Instead, they selectively produced TNF-α, IL-1ß, and CCL3 in response to viruses and immune complexes containing nucleic acids, via a proinflammatory TLR7-TLR 8-MyD88-MEK pathway. Thus, CD14(dim) cells are bona fide monocytes involved in the innate local surveillance of tissues and the pathogenesis of autoimmune diseases.


Asunto(s)
Receptores de Lipopolisacáridos/fisiología , Monocitos/fisiología , Ácidos Nucleicos/fisiología , Receptor Toll-Like 7/fisiología , Receptor Toll-Like 8/fisiología , Virus/inmunología , Animales , Presentación de Antígeno , Citocinas/biosíntesis , Proteínas Ligadas a GPI , Antígenos HLA-DR/análisis , Humanos , Lupus Eritematoso Sistémico/inmunología , Ratones , Factor 88 de Diferenciación Mieloide/fisiología , Especies Reactivas de Oxígeno/metabolismo , Receptores de IgG/análisis
16.
Pediatr Res ; 85(4): 477-483, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30659270

RESUMEN

BACKGROUND: A lack of consensus exists as to the timing of kidney biopsy in children with steroid-dependent nephrotic syndrome (SDNS) where minimal change disease (MCD) predominates. This study aimed at examining the applicability of a biomarker-assisted risk score model to select SDNS patients at high risk of focal segmental glomerulosclerosis (FSGS) for biopsy. METHODS: Fifty-five patients with SDNS and biopsy-proven MCD (n = 40) or FSGS (n = 15) were studied. A risk score model was developed with variables consisting of age, sex, eGFR, suPAR levels and percentage of CD8+ memory T cells. Following multivariate regression analysis, total risk score was calculated as sum of the products of odds ratios and corresponding variables. Predictive cut-off point was determined using receiver operator characteristics (ROC) curve analysis. RESULTS: Plasma suPAR levels in FSGS patients were significantly higher, while percentage of CD45RO+CD8+CD3+ was significantly lower than in MCD patients and controls. ROC analysis suggests the risk score model with threshold score of 16.7 (AUC 0.84, 95% CI 0.72-0.96) was a good predictor of FSGS on biopsy. The 100% PPV cut-off was >24.0, while the 100% NPV was <13.3. CONCLUSION: A suPAR and CD8+ memory T cell percentage-based risk score model was developed to stratify SDNS patients for biopsy and for predicting FSGS.


Asunto(s)
Antiinflamatorios/uso terapéutico , Biopsia , Glomeruloesclerosis Focal y Segmentaria/patología , Riñón/patología , Síndrome Nefrótico/patología , Prednisolona/uso terapéutico , Adolescente , Estudios de Casos y Controles , Niño , Preescolar , Femenino , Glomeruloesclerosis Focal y Segmentaria/etiología , Humanos , Subgrupos Linfocitarios , Masculino , Síndrome Nefrótico/complicaciones , Síndrome Nefrótico/etiología , Receptores del Activador de Plasminógeno Tipo Uroquinasa/sangre , Medición de Riesgo
18.
Int Immunol ; 28(5): 223-32, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26567289

RESUMEN

The toll-like receptors (TLRs) are important innate receptors recognizing potentially pathogenic material. However, they also play a significant role in the development of Alzheimer's disease, cancer, autoimmunity and the susceptibility to viral infections. Macrophages are essential for an effective immune response to foreign material and the resolution of inflammation. In these studies, we examined the impact of different TLR ligands on macrophage cell function. We demonstrate that stimulation of all TLRs tested increases the phagocytosis of apoptotic cells by macrophages. TLR7 and TLR9 ligation decreased the levels of the surface co-expression molecules CD86 and MHCII, which was associated with a concomitant reduction in antigen presentation and proliferation of T cells. This down-regulation in macrophage function was not due to an increase in cell death. In fact, exposure to TLR7 or TLR9 ligands promoted cell viability for up to 9 days, in contrast to TLR3 or TLR4. Additionally, macrophages exposed to TLR7/TLR9 ligands had a significantly lower ratio of Il-12/Il-10 mRNA expression compared with those treated with the TLR4 ligand, LPS. Taken together, these data demonstrate that TLR7/TLR9 ligands push the macrophage into a phagocytic long-lived cell, with a decreased capacity of antigen presentation and reminiscent of the M2 polarized state.


Asunto(s)
Presentación de Antígeno , Macrófagos/inmunología , Glicoproteínas de Membrana/agonistas , Glicoproteínas de Membrana/inmunología , Receptor Toll-Like 7/agonistas , Receptor Toll-Like 7/inmunología , Receptor Toll-Like 9/agonistas , Receptor Toll-Like 9/inmunología , Animales , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/inmunología , Antígenos de Histocompatibilidad Clase II/genética , Antígenos de Histocompatibilidad Clase II/inmunología , Interleucina-10/genética , Interleucina-10/inmunología , Interleucina-12/genética , Interleucina-12/inmunología , Ligandos , Lipopolisacáridos/toxicidad , Glicoproteínas de Membrana/genética , Ratones , Ratones Noqueados , Fagocitosis/efectos de los fármacos , Fagocitosis/genética , Receptor Toll-Like 7/genética , Receptor Toll-Like 9/genética
19.
Proc Natl Acad Sci U S A ; 111(38): 13870-5, 2014 Sep 23.
Artículo en Inglés | MEDLINE | ID: mdl-25189770

RESUMEN

Metastatic spread is the leading cause of cancer mortality. Breast cancer (BCa) metastatic recurrence can happen years after removal of the primary tumor. Here we show that Ubc13, an E2 enzyme that catalyzes K63-linked protein polyubiquitination, is largely dispensable for primary mammary tumor growth but is required for metastatic spread and lung colonization by BCa cells. Loss of Ubc13 inhibited BCa growth and survival only at metastatic sites. Ubc13 was dispensable for transforming growth factor ß (TGFß)-induced SMAD activation but was required for activation of non-SMAD signaling via TGFß-activating kinase 1 (TAK1) and p38, whose activity controls expression of numerous metastasis promoting genes. p38 activation restored metastatic activity to Ubc13-deficient cells, and its pharmacological inhibition attenuated BCa metastasis in mice, suggesting it is a therapeutic option for metastatic BCa.


Asunto(s)
Neoplasias de la Mama/enzimología , Quinasas Quinasa Quinasa PAM/metabolismo , Sistema de Señalización de MAP Quinasas , Proteínas de Neoplasias/metabolismo , Enzimas Ubiquitina-Conjugadoras/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Línea Celular Tumoral , Femenino , Xenoinjertos , Humanos , Quinasas Quinasa Quinasa PAM/genética , Ratones , Ratones Endogámicos NOD , Ratones SCID , Metástasis de la Neoplasia , Proteínas de Neoplasias/genética , Trasplante de Neoplasias , Enzimas Ubiquitina-Conjugadoras/genética , Proteínas Quinasas p38 Activadas por Mitógenos/genética
20.
J Immunol ; 191(8): 4375-82, 2013 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-24048896

RESUMEN

TLRs play a pivotal role in the recognition of bacteria and viruses. Members of the family recognize specific pathogen sequences to trigger both MyD88 and TRIF-dependent pathways to stimulate a plethora of cells. Aberrant activation of these pathways is known to play a critical role in the development of autoimmunity and cancer. However, how these pathways are entirely regulated is not fully understood. In these studies, we have identified Annexin-A1 (ANXA1) as a novel regulator of TLR-induced IFN-ß and CXCL10 production. We demonstrate that in the absence of ANXA1, mice produce significantly less IFN-ß and CXCL10, and macrophages and plasmacytoid dendritic cells have a deficiency in activation following polyinosinic:polycytidylic acid administration in vivo. Furthermore, a deficiency in activation is observed in macrophages after LPS and polyinosinic:polycytidylic acid in vitro. In keeping with these findings, overexpression of ANXA1 resulted in enhanced IFN-ß and IFN-stimulated responsive element promoter activity, whereas silencing of ANXA1 impaired TLR3- and TLR4-induced IFN-ß and IFN-stimulated responsive element activation. In addition, we show that the C terminus of ANXA1 directly associates with TANK-binding kinase 1 to regulate IFN regulatory factor 3 translocation and phosphorylation. Our findings demonstrate that ANXA1 plays an important role in TLR activation, leading to an augmentation in the type 1 IFN antiviral cytokine response.


Asunto(s)
Anexina A1/metabolismo , Interferón beta/biosíntesis , Proteínas Serina-Treonina Quinasas/metabolismo , Receptor Toll-Like 3/metabolismo , Receptor Toll-Like 4/metabolismo , Transporte Activo de Núcleo Celular , Animales , Anexina A1/biosíntesis , Anexina A1/genética , Línea Celular , Quimiocina CXCL10/biosíntesis , Células Dendríticas/metabolismo , Activación Enzimática , Células HEK293 , Humanos , Factor 3 Regulador del Interferón/metabolismo , Lipopolisacáridos , Activación de Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Fosforilación , Poli I-C/farmacología , Transducción de Señal/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA