Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Immunity ; 55(11): 2103-2117.e10, 2022 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-36323311

RESUMEN

The surface of the central nervous system (CNS) is protected by the meninges, which contain a dense network of meningeal macrophages (MMs). Here, we examined the role of tissue-resident MM in viral infection. MHC-II- MM were abundant neonatally, whereas MHC-II+ MM appeared over time. These barrier macrophages differentially responded to in vivo peripheral challenges such as LPS, SARS-CoV-2, and lymphocytic choriomeningitis virus (LCMV). Peripheral LCMV infection, which was asymptomatic, led to a transient infection and activation of the meninges. Mice lacking macrophages but conserving brain microglia, or mice bearing macrophage-specific deletion of Stat1 or Ifnar, exhibited extensive viral spread into the CNS. Transcranial pharmacological depletion strategies targeting MM locally resulted in several areas of the meninges becoming infected and fatal meningitis. Low numbers of MHC-II+ MM, which is seen upon LPS challenge or in neonates, corelated with higher viral load upon infection. Thus, MMs protect against viral infection and may present targets for therapeutic manipulation.


Asunto(s)
COVID-19 , Coriomeningitis Linfocítica , Animales , Ratones , Lipopolisacáridos , Ratones Endogámicos C57BL , SARS-CoV-2 , Virus de la Coriomeningitis Linfocítica/fisiología , Macrófagos , Meninges
2.
Lab Invest ; 103(12): 100258, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37813278

RESUMEN

Breast cancer is one of the most prominent types of cancers, in which therapeutic resistance is a major clinical concern. Specific subtypes, such as claudin-low and metaplastic breast carcinoma (MpBC), have been associated with high nongenetic plasticity, which can facilitate resistance. The similarities and differences between these orthogonal subtypes, identified by molecular and histopathological analyses, respectively, remain insufficiently characterized. Furthermore, adequate methods to identify high-plasticity tumors to better anticipate resistance are lacking. Here, we analyzed 11 triple-negative breast tumors, including 3 claudin-low and 4 MpBC, via high-resolution spatial transcriptomics. We combined pathological annotations and deconvolution approaches to precisely identify tumor spots, on which we performed signature enrichment, differential expression, and copy number analyses. We used The Cancer Genome Atlas and Cancer Cell Line Encyclopedia public databases for external validation of expression markers. By focusing our spatial transcriptomic analyses on tumor cells in MpBC samples, we bypassed the negative impact of stromal contamination and identified specific markers that are neither expressed in other breast cancer subtypes nor expressed in stromal cells. Three markers (BMPER, POPDC3, and SH3RF3) were validated in external expression databases encompassing bulk tumor material and stroma-free cell lines. We unveiled that existing bulk expression signatures of high-plasticity breast cancers are relevant in mesenchymal transdifferentiated compartments but can be hindered by abundant stromal cells in tumor samples, negatively impacting their clinical applicability. Spatial transcriptomic analyses constitute powerful tools to identify specific expression markers and could thus enhance diagnosis and clinical care of rare high-plasticity breast cancers.


Asunto(s)
Neoplasias de la Mama , Neoplasias de la Mama Triple Negativas , Humanos , Femenino , Neoplasias de la Mama/diagnóstico , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama Triple Negativas/patología , Perfilación de la Expresión Génica , Mama/metabolismo , Transcriptoma , Claudinas/metabolismo , Pronóstico , Proteínas Portadoras/metabolismo , Proteínas Musculares/metabolismo , Moléculas de Adhesión Celular/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo
3.
Int J Cancer ; 151(1): 138-152, 2022 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-35253899

RESUMEN

Beyond their critical role in hemostasis, platelets physically interact with neutrophils to form neutrophil-platelet aggregates (NPAs), enhancing neutrophil effector functions during inflammation. NPAs may also promote disease worsening in various inflammatory diseases. However, characterization of NPAs in cancer remains totally unexplored. Using ImageStreamX (ISX) imaging flow cytometer, we were not only allowed able to detect CD15+ CD14- CD36+ ITGA2B+ NPAs in both healthy donors' (HDs) and cancer patients' bloods, but we also showed that NPAs result from the binding of platelets preferentially to low-density neutrophils (LDNs) as opposed to normal-density neutrophils (NDNs). By reanalyzing two independent public scRNAseq data of whole blood leukocytes from cancer patients and HDs, we could identify a subset of neutrophils with high platelet gene expression that may correspond to NPAs. Moreover, we showed that cancer patients' derived NPAs possessed a distinct molecular signature compared to the other neutrophil subsets, independently of platelet genes. Gene ontology (GO) term enrichment analysis of this NPAs-associated neutrophil transcriptomic signature revealed a significant enrichment of neutrophil degranulation, chemotaxis and trans-endothelial migration GO terms. Lastly, using The Cancer Genome Atlas (TCGA), we could show by multivariate Cox analysis that the NPAs-associated neutrophil transcriptomic signature was associated with a worse patient prognosis in several cancer types. These results suggest that neutrophils from NPAs are systemically primed by platelets empowering them with cancer progression capacities once at tumor site. NPAs may therefore hold clinical utility as novel noninvasive blood prognostic biomarker in cancer patients with solid tumors.


Asunto(s)
Neoplasias , Neutrófilos , Plaquetas , Citometría de Flujo , Humanos , Neoplasias/patología , Neutrófilos/patología , Pronóstico
4.
Proc Natl Acad Sci U S A ; 115(41): 10404-10409, 2018 10 09.
Artículo en Inglés | MEDLINE | ID: mdl-30249647

RESUMEN

Prominent changes in the gut microbiota (referred to as "dysbiosis") play a key role in the development of allergic disorders, but the underlying mechanisms remain unknown. Study of the delayed-type hypersensitivity (DTH) response in mice contributed to our knowledge of the pathophysiology of human allergic contact dermatitis. Here we report a negative regulatory role of the RIG-I-like receptor adaptor mitochondrial antiviral signaling (MAVS) on DTH by modulating gut bacterial ecology. Cohousing and fecal transplantation experiments revealed that the dysbiotic microbiota of Mavs-/- mice conferred a proallergic phenotype that is communicable to wild-type mice. DTH sensitization coincided with increased intestinal permeability and bacterial translocation within lymphoid organs that enhanced DTH severity. Collectively, we unveiled an unexpected impact of RIG-I-like signaling on the gut microbiota with consequences on allergic skin disease outcome. Primarily, these data indicate that manipulating the gut microbiota may help in the development of therapeutic strategies for the treatment of human allergic skin pathologies.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/fisiología , Disbiosis/complicaciones , Microbioma Gastrointestinal/inmunología , Hipersensibilidad/etiología , Intestinos/inmunología , Enfermedades Cutáneas Bacterianas/etiología , Animales , Modelos Animales de Enfermedad , Femenino , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Hipersensibilidad/metabolismo , Hipersensibilidad/patología , Intestinos/microbiología , Intestinos/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , Transducción de Señal , Enfermedades Cutáneas Bacterianas/metabolismo , Enfermedades Cutáneas Bacterianas/patología
5.
Nat Immunol ; 9(9): 1037-46, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18641654

RESUMEN

Tumor necrosis factor receptor 1 (TNFR1) and Toll-like receptors (TLRs) regulate immune and inflammatory responses. Here we show that the TNFR1-associated death domain protein (TRADD) is critical in TNFR1, TLR3 and TLR4 signaling. TRADD deficiency abrogated TNF-induced apoptosis, prevented recruitment of the ubiquitin ligase TRAF2 and ubiquitination of the adaptor RIP1 in the TNFR1 signaling complex, and considerably inhibited but did not completely abolish activation of the transcription factor NF-kappaB and mitogen-activated protein kinases 'downstream' of TNFR1. TRIF-dependent cytokine production induced by the synthetic double-stranded RNA poly(I:C) and lipopolysaccharide was lower in TRADD-deficient mice than in wild-type mice. Moreover, TRADD deficiency inhibited poly(I:C)-mediated RIP1 ubiquitination and activation of NF-kappaB and mitogen-activated protein kinase signaling in fibroblasts but not in bone marrow macrophages. Thus, TRADD is an essential component of TNFR1 signaling and has a critical but apparently cell type-specific function in TRIF-dependent TLR responses.


Asunto(s)
Transducción de Señal , Proteína de Dominio de Muerte Asociada a Receptor de TNF/deficiencia , Factor 1 Asociado a Receptor de TNF/metabolismo , Péptidos y Proteínas Asociados a Receptores de Factores de Necrosis Tumoral/fisiología , Animales , Ratones , Proteínas Quinasas Activadas por Mitógenos/metabolismo , FN-kappa B/metabolismo , Transducción de Señal/efectos de los fármacos , Proteína de Dominio de Muerte Asociada a Receptor de TNF/metabolismo , Péptidos y Proteínas Asociados a Receptores de Factores de Necrosis Tumoral/genética , Ubiquitina/metabolismo
7.
Immunity ; 28(5): 651-61, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18439848

RESUMEN

Upon detection of viral RNA, the helicases RIG-I and/or MDA5 trigger, via their adaptor Cardif (also known as IPS-1, MAVS, or VISA), the activation of the transcription factors NF-kappaB and IRF3, which collaborate to induce an antiviral type I interferon (IFN) response. FADD and RIP1, known as mediators of death-receptor signaling, are implicated in this antiviral pathway; however, the link between death-receptor and antiviral signaling is not known. Here we showed that TRADD, a crucial adaptor of tumor necrosis factor receptor (TNFRI), was important in RIG-like helicase (RLH)-mediated signal transduction. TRADD is recruited to Cardif and orchestrated complex formation with the E3 ubiquitin ligase TRAF3 and TANK and with FADD and RIP1, leading to the activation of IRF3 and NF-kappaB. Loss of TRADD prevented Cardif-dependent activation of IFN-beta, reduced the production of IFN-beta in response to RNA viruses, and enhanced vesicular stomatitis virus replication. Thus, TRADD is not only an essential component of proinflammatory TNFRI signaling, but is also required for RLH-Cardif-dependent antiviral immune responses.


Asunto(s)
ADN Helicasas/metabolismo , Factor 3 Regulador del Interferón/metabolismo , Infecciones por Rhabdoviridae/inmunología , Proteína de Dominio de Muerte Asociada a Receptor de TNF/metabolismo , Vesiculovirus/inmunología , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Proteína de Dominio de Muerte Asociada a Fas/metabolismo , Proteínas Activadoras de GTPasa/metabolismo , Humanos , Factor 3 Regulador del Interferón/inmunología , Interferón Tipo I/inmunología , Interferón Tipo I/metabolismo , Proteínas de la Membrana/metabolismo , Ratones , Ratones Noqueados , Ratones Mutantes , FN-kappa B/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Receptores de Superficie Celular , Infecciones por Rhabdoviridae/virología , Transducción de Señal , Factor 3 Asociado a Receptor de TNF/inmunología , Factor 3 Asociado a Receptor de TNF/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Vesiculovirus/fisiología
8.
J Immunol ; 191(7): 3847-57, 2013 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-23975862

RESUMEN

The inflammasome is a signaling platform that is central to the innate immune responses to bacterial infections. Francisella tularensis is a bacterium replicating within the host cytosol. During F. tularensis subspecies novicida infection, AIM2, an inflammasome receptor sensing cytosolic DNA, activates caspase-1 in an ASC-dependent manner, leading to both pyroptosis and release of the proinflammatory cytokines IL-1ß and IL-18. Activation of this canonical inflammasome pathway is key to limit F. novicida infection. In this study, by comparing the immune responses of AIM2 knockout (KO), ASC(KO), and Casp1(KO) mice in response to F. novicida infection, we observed that IFN-γ levels in the serum of Casp1(KO) mice were much higher than the levels observed in AIM2(KO) and ASC(KO) mice. This difference in IFN-γ production was due to a large production of IFN-γ by NK cells in Casp1(KO) mice that was not observed in ASC(KO) mice. The deficit in IFN-γ production observed in ASC(KO) mice was not due to a reduced Dock2 expression or to an intrinsic defect of ASC(KO) NK cells. We demonstrate that in infected Casp1(KO) mice, IFN-γ production is due to an ASC-dependent caspase-1-independent pathway generating IL-18. Furthermore, we present in vitro data suggesting that the recently described AIM2/ASC/caspase-8 noncanonical pathway is responsible for the caspase-1-independent IL-18 releasing activity. To our knowledge, this study is the first in vivo evidence of an alternative pathway able to generate in a caspase-1-independent pathway bioactive IL-18 to boost the production of IFN-γ, a cytokine critical for the host antibacterial response.


Asunto(s)
Caspasa 1/deficiencia , Proteínas del Citoesqueleto/genética , Francisella , Infecciones por Bacterias Gramnegativas/genética , Infecciones por Bacterias Gramnegativas/metabolismo , Interferón gamma/metabolismo , Interleucina-18/metabolismo , Animales , Proteínas Reguladoras de la Apoptosis , Proteínas Adaptadoras de Señalización CARD , Caspasa 1/genética , Caspasa 8/genética , Caspasa 8/metabolismo , Degranulación de la Célula/inmunología , Diferenciación Celular/inmunología , Proteínas del Citoesqueleto/metabolismo , Modelos Animales de Enfermedad , Francisella/inmunología , Proteínas Activadoras de GTPasa/genética , Proteínas Activadoras de GTPasa/metabolismo , Infecciones por Bacterias Gramnegativas/inmunología , Factores de Intercambio de Guanina Nucleótido , Células HEK293 , Humanos , Inflamasomas/metabolismo , Interleucina-18/sangre , Células Asesinas Naturales/citología , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Subgrupos Linfocitarios/inmunología , Subgrupos Linfocitarios/metabolismo , Ratones , Ratones Noqueados
9.
Immunopharmacol Immunotoxicol ; 37(1): 19-25, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25358107

RESUMEN

The purpose of this study is to define the optimal dose of oral cyclosporine A (CsA) microemulsion in newborn swine for transplantation studies and to describe its pharmacokinetics and acute renal effects in short-term administration. Thirteen neonatal pigs were randomized into four groups: one control and three groups with CsA administration at 4, 8 and 12 mg/kg/d for 15 days (D). Blood samples were collected on D 0, 2, 4, 9 and 14 to determine the changes of the CsA trough concentrations, the creatinine (Cr) and blood urea nitrogen (BUN) serum concentrations. On D 14, blood samples were collected every hour from 1 h to 10 h after CsA administration to determine the area under the curve (AUC). On D 15, kidneys were removed for histological analysis. We observed a stabilization of CsA trough concentrations from D 4 to D 14. On D 14, in the three treated groups, CsA trough concentrations were 687 ± 7, 1200 ± 77 and 2211 ± 1030 ng/ml, respectively; AUC (0-10 h) were 6721 ± 51 ng·h/ml in group 4 mg/kg/d, 13431 ± 988 ng·h/ml in group 8 mg/kg/d and 28264 ± 9430 ng·h/ml in group 12 mg/kg/d. Cr concentrations were not significantly different among the four groups; but compared to control group, BUN concentrations of the three treated groups increased significantly. CsA was well tolerated; neither acute, severe adverse event nor renal histological abnormality was observed. In conclusion, a 15-d course of oral CsA treatment ranged from 4 to 12 mg/kg/d is safe for newborn pigs, which need much lower CsA dose than adult pigs to reach comparable trough level and AUC. As immunosuppressive therapy in newborn pigs, we recommend a CsA dose of 4 mg/kg/d to achieve a trough blood concentration between 400 and 800 ng/ml.


Asunto(s)
Ciclosporina/administración & dosificación , Ciclosporina/efectos adversos , Inmunosupresores/administración & dosificación , Inmunosupresores/efectos adversos , Riñón/efectos de los fármacos , Tolerancia al Trasplante/efectos de los fármacos , Administración Oral , Animales , Animales Recién Nacidos , Área Bajo la Curva , Nitrógeno de la Urea Sanguínea , Creatinina/sangre , Ciclosporina/sangre , Relación Dosis-Respuesta Inmunológica , Femenino , Inmunosupresores/sangre , Riñón/patología , Masculino , Distribución Aleatoria , Porcinos
10.
Gut Microbes ; 15(2): 2249960, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37655966

RESUMEN

Over 90% of epidemic non-bacterial gastroenteritis are caused by human noroviruses (NoVs), which persist in a substantial subset of people allowing their spread worldwide. This has led to a significant number of endemic cases and up to 70,000 children deaths in developing countries. NoVs are primarily transmitted through the fecal-oral route. To date, studies have focused on the influence of the gut microbiota on enteric viral clearance by mucosal immunity. In this study, the use of mouse norovirus S99 (MNoV_S99) and CR6 (MNoV_CR6), two persistent strains, allowed us to provide evidence that the norovirus-induced exacerbation of colitis severity relied on bacterial sensing by nucleotide-binding oligomerization domain 2 (Nod2). Consequently, Nod2-deficient mice showed reduced levels of gravity of Dextran sodium sulfate (DSS)-induced colitis with both viral strains. And MNoV_CR6 viremia was heightened in Nod2-/- mice in comparison with animals hypomorphic for Atg16l1, which are prone to aggravated inflammation under DSS. Accordingly, the infection of macrophages derived from WT mice promoted the phosphorylation of Signal Transducer and Activator of Transcription 1 (STAT1) and NOD2's expression levels. Higher secretion of Tumor Necrosis Factor alpha (TNFα) following NOD2 activation and better viral clearance were measured in these cells. By contrast, reduced levels of pSTAT1 and blunted downstream secretion of TNFα were found in Nod2-deficient macrophages infected by MNoV_S99. Hence, our results uncover a previously unidentified virus-host-bacterial interplay that may represent a novel therapeutic target for treating noroviral origin gastroenteritis that may be linked with susceptibility to several common illnesses such as Crohn's disease.


Asunto(s)
Infecciones por Caliciviridae , Colitis , Gastroenteritis , Microbioma Gastrointestinal , Proteína Adaptadora de Señalización NOD2 , Animales , Ratones , Infecciones por Caliciviridae/inmunología , Colitis/inducido químicamente , Colitis/virología , Gastroenteritis/inmunología , Gastroenteritis/virología , Proteína Adaptadora de Señalización NOD2/metabolismo
11.
Nat Genet ; 55(4): 607-618, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36928603

RESUMEN

Malignant pleural mesothelioma (MPM) is an aggressive cancer with rising incidence and challenging clinical management. Through a large series of whole-genome sequencing data, integrated with transcriptomic and epigenomic data using multiomics factor analysis, we demonstrate that the current World Health Organization classification only accounts for up to 10% of interpatient molecular differences. Instead, the MESOMICS project paves the way for a morphomolecular classification of MPM based on four dimensions: ploidy, tumor cell morphology, adaptive immune response and CpG island methylator profile. We show that these four dimensions are complementary, capture major interpatient molecular differences and are delimited by extreme phenotypes that-in the case of the interdependent tumor cell morphology and adapted immune response-reflect tumor specialization. These findings unearth the interplay between MPM functional biology and its genomic history, and provide insights into the variations observed in the clinical behavior of patients with MPM.


Asunto(s)
Neoplasias Pulmonares , Mesotelioma Maligno , Mesotelioma , Neoplasias Pleurales , Humanos , Mesotelioma Maligno/genética , Mesotelioma Maligno/complicaciones , Mesotelioma/genética , Mesotelioma/patología , Multiómica , Neoplasias Pleurales/genética , Neoplasias Pleurales/patología , Neoplasias Pulmonares/patología , Biomarcadores de Tumor/genética
12.
Proc Natl Acad Sci U S A ; 106(29): 12067-72, 2009 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-19574455

RESUMEN

The ATPase retinoid acid-inducible gene (RIG)-I senses viral RNA in the cytoplasm of infected cells and subsequently activates cellular antiviral defense mechanisms. RIG-I recognizes molecular structures that discriminate viral from host RNA. Here, we show that RIG-I ligands require base-paired structures in conjunction with a free 5'-triphosphate to trigger antiviral signaling. Hitherto unavailable chemically synthesized 5'-triphosphate RNA ligands do not trigger RIG-I-dependent IFN production in cells, and they are unable to trigger the ATPase activity of RIG-I without a base-paired stretch. Consistently, immunostimulatory RNA from cells infected with a virus recognized by RIG-I is sensitive to double-strand, but not single-strand, specific RNases. In vitro, base-paired stretches and the 5'-triphosphate bind to distinct sites of RIG-I and synergize to trigger the induction of signaling competent RIG-I multimers. Strengthening our model of a bipartite molecular pattern for RIG-I activation, we show that the activity of supposedly "single-stranded" 5'-triphosphate RNAs generated by in vitro transcription depends on extended and base-paired by-products inadvertently, but commonly, produced by this method. Together, our findings accurately define a minimal molecular pattern sufficient to activate RIG-I that can be found in viral genomes or transcripts.


Asunto(s)
Emparejamiento Base , ARN/química , ARN/inmunología , Receptores de Ácido Retinoico/metabolismo , Transducción de Señal/inmunología , Virus/inmunología , Adenosina Trifosfatasas/metabolismo , Animales , Sitios de Unión , Línea Celular , ARN Polimerasas Dirigidas por ADN/metabolismo , Humanos , Ligandos , Ratones , Unión Proteica , Multimerización de Proteína , Receptores de Reconocimiento de Patrones/metabolismo , Transcripción Genética , Proteínas Virales/metabolismo
13.
EMBO Rep ; 10(8): 916-22, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19590578

RESUMEN

Detection of viral nucleic acids is central to antiviral immunity. Recently, DAI/ZBP1 (DNA-dependent activator of IRFs/Z-DNA binding protein 1) was identified as a cytoplasmic DNA sensor and shown to activate the interferon regulatory factor (IRF) and nuclear factor-kappa B (NF-kappaB) transcription factors, leading to type-I interferon production. DAI-induced IRF activation depends on TANK-binding kinase 1 (TBK1), whereas signalling pathways and molecular components involved in NF-kappaB activation remain elusive. Here, we report the identification of two receptor-interacting protein (RIP) homotypic interaction motifs (RHIMs) in the DAI protein sequence, and show that these domains relay DAI-induced NF-kappaB signals through the recruitment of the RHIM-containing kinases RIP1 and RIP3. We show that knockdown of not only RIP1, but also RIP3 affects DAI-induced NF-kappaB activation. Importantly, RIP recruitment to DAI is inhibited by the RHIM-containing murine cytomegalovirus (MCMV) protein M45. These findings delineate the DAI signalling pathway to NF-kappaB and suggest a possible new immune modulation strategy of the MCMV.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , FN-kappa B/metabolismo , Proteínas de Complejo Poro Nuclear/metabolismo , Proteínas de Unión al ARN/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Proteínas Adaptadoras del Transporte Vesicular/química , Proteínas Adaptadoras del Transporte Vesicular/genética , Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Animales , Sitios de Unión , Línea Celular , Proteínas de Unión al ADN/genética , Glicoproteínas/genética , Glicoproteínas/metabolismo , Humanos , Immunoblotting , Inmunoprecipitación , Ratones , Proteínas de Complejo Poro Nuclear/química , Proteínas de Complejo Poro Nuclear/genética , Reacción en Cadena de la Polimerasa , Unión Proteica/genética , Unión Proteica/fisiología , Proteínas de Unión al ARN/química , Proteínas de Unión al ARN/genética , Proteína Serina-Treonina Quinasas de Interacción con Receptores/química , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética
14.
J Immunol ; 182(6): 3846-54, 2009 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-19265164

RESUMEN

Most memory CD8 T cell subsets that have been hitherto defined are generated in response to infectious pathogens. In this study, we have characterized the CD8 T cells that survive priming conditions, devoid of pathogen-derived danger signals. In both a TCR-transgenic model and a model of contact hypersensitivity, we show that the priming of naive CD8 T cells under sterile inflammatory conditions generates memory. The corresponding memory CD8 T cells can be identified by their intermediate expression levels of CD44 and CD122. We also show that CD44/122(int) memory CD8 T cells spontaneously develop in wild type mice and that they display intermediate levels of several other memory traits including functional (IFN-gamma secretion capacity, CCL5 messenger stores), phenotypic, and molecular (T-bet and eomesodermin expression levels) features. We finally show that they correspond to an early differentiation stage and can further differentiate in CD44/122(high) memory T cells. Altogether, our results identify a new memory CD8 T cell subset that is generated under sterile inflammatory conditions and involved in the recall contact hypersensitivity reactions that are responsible for allergic contact dermatitis.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Diferenciación Celular/inmunología , Receptores de Hialuranos/fisiología , Memoria Inmunológica/genética , Mediadores de Inflamación/fisiología , Subunidad beta del Receptor de Interleucina-2/fisiología , Activación de Linfocitos/inmunología , Animales , Biomarcadores/metabolismo , Linfocitos T CD8-positivos/trasplante , Diferenciación Celular/genética , Dermatitis por Contacto/genética , Dermatitis por Contacto/inmunología , Receptores de Hialuranos/biosíntesis , Mediadores de Inflamación/metabolismo , Subunidad beta del Receptor de Interleucina-2/biosíntesis , Activación de Linfocitos/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Subgrupos de Linfocitos T/citología , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo
15.
Front Immunol ; 10: 2155, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31616408

RESUMEN

Neutrophils have been extensively described in the pathophysiology of autoimmune and infectious diseases. Accumulating evidence also suggests the important role of neutrophils in cancer progression through their interaction with cancer and immune cells in blood and in the tumor microenvironment (TME). Most studies have described neutrophils as key drivers of cancer progression, due to their involvement in various tumor promoting functions including proliferation, aggressiveness, and dissemination, as well as in immune suppression. However, such studies were focusing on late-stages of tumorigenesis, in which chronic inflammation had already developed. The role of tumor-associated neutrophils (TANs) at early stages of tumor development remains poorly described, though recent findings indicate that early-stage TANs may display anti-tumor properties. Beyond their role at tumor site, evidence supported by NLR retrospective studies and functional analyses suggest that blood neutrophils could also actively contribute to tumorigenesis. Hence, it appears that the phenotype and functions of neutrophils vary greatly during tumor progression, highlighting their heterogeneity. The origin of pro- or anti-tumor neutrophils is generally believed to arise following a change in cell state, from resting to activated. Moreover, the fate of neutrophils may also involve distinct differentiation programs yielding various subsets of pro or anti-tumor neutrophils. In this review, we will discuss the current knowledge on neutrophils heterogeneity across different tissues and their impact on tumorigenesis, as well as neutrophil-based therapeutic strategies that have shown promising results in pre-clinical studies, paving the way for the design of neutrophil-based next generation immunotherapy.


Asunto(s)
Carcinogénesis/inmunología , Inmunoterapia , Neoplasias , Neutrófilos/inmunología , Microambiente Tumoral/inmunología , Animales , Carcinogénesis/patología , Humanos , Inflamación/inmunología , Inflamación/patología , Inflamación/terapia , Neoplasias/inmunología , Neoplasias/patología , Neoplasias/terapia , Neutrófilos/patología
16.
J Clin Invest ; 115(11): 3117-27, 2005 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16276417

RESUMEN

Panton-Valentine leukocidin (PVL) is a pore-forming toxin secreted by Staphylococcus aureus that has recently been associated with necrotizing pneumonia. In the present study, we report that in vitro, PVL induces polymorphonuclear cell death by necrosis or by apoptosis, depending on the PVL concentration. PVL-induced apoptosis was associated with a rapid disruption of mitochondrial homeostasis and activation of caspase-9 and caspase-3, suggesting that PVL-induced apoptosis is preferentially mediated by the mitochondrial pathway. Polymorphonuclear cell exposure to PVL leads to mitochondrial localization of the toxin, whereas Bax, 1 of the 2 essential proapoptotic members of the Bcl-2 family, was still localized in the cytosol. Addition of PVL to isolated mitochondria induced the release of the apoptogenic proteins cytochrome c and Smac/DIABLO. Therefore, we suggest that PVL, which belongs to the pore-forming toxin family, could act at the mitochondrion level by creating pores in the mitochondrial outer membrane. Furthermore, LukS-PV, 1 of the 2 components of PVL, was detected in lung sections of patients with necrotizing pneumonia together with DNA fragmentation, suggesting that PVL induces apoptosis in vivo and thereby is directly involved in the pathophysiology of necrotizing pneumonia.


Asunto(s)
Apoptosis/efectos de los fármacos , Leucocidinas/farmacología , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Neutrófilos/citología , Neutrófilos/efectos de los fármacos , Proteína X Asociada a bcl-2/fisiología , Proteínas Reguladoras de la Apoptosis , Toxinas Bacterianas , Membrana Celular/fisiología , Células Cultivadas , Citocromos c/metabolismo , Exotoxinas , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Cinética , Pulmón/microbiología , Pulmón/patología , Mitocondrias/fisiología , Proteínas Mitocondriales/metabolismo , Necrosis , Neutrófilos/microbiología , Neumonía Estafilocócica/microbiología , Neumonía Estafilocócica/patología , Staphylococcus aureus/fisiología
17.
Trends Cancer ; 3(7): 491-505, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28718404

RESUMEN

Disruption of endoplasmic reticulum (ER) homeostasis results in ER stress and activation of the unfolded protein response (UPR). This response alleviates cell stress, and is activated in both tumor cells and tumor infiltrating immune cells. The UPR plays a dual function in cancer biology, acting as a barrier to tumorigenesis at the premalignant stage, while fostering cancer maintenance in established tumors. In infiltrating immune cells, the UPR has been involved in both immunosurveillance and immunosuppressive functions. This review aims to decipher the role of the UPR at different stages of tumorigenesis and how the UPR shapes the balance between immunosurveillance and immune escape. This knowledge may improve existing UPR-targeted therapies and the design of novel strategies for cancer treatment.


Asunto(s)
Estrés del Retículo Endoplásmico/inmunología , Monitorización Inmunológica , Neoplasias/diagnóstico , Transducción de Señal/inmunología , Respuesta de Proteína Desplegada/inmunología , Animales , Antineoplásicos/uso terapéutico , Apoptosis , Carcinogénesis/inmunología , Transformación Celular Neoplásica/inmunología , Retículo Endoplásmico/metabolismo , Humanos , Inmunidad Celular/inmunología , Estadificación de Neoplasias , Neoplasias/tratamiento farmacológico , Neoplasias/inmunología , Neoplasias/patología , Escape del Tumor/inmunología , Respuesta de Proteína Desplegada/efectos de los fármacos
19.
Front Immunol ; 7: 116, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27092138

RESUMEN

Although several tolerance induction protocols have been successfully implemented in adult renal transplantation, no tolerance induction approach has, as yet, been defined for solid organ transplantations in young infants. Pediatric transplant recipients have a pressing demand for the elaboration of tolerance induction regimens. Indeed, since they display a longer survival time, they are exposed to a higher level of risks linked to long-term immunosuppression (IS) and to chronic rejection. Interestingly, central tolerance induction may be of great interest in newborns, because of their immunological immaturity and the important role of the thymus at this early stage in life. The present review aims to clarify mechanisms and strategies of tolerance induction in these immunologically premature recipients. We first introduce the discovery and mechanisms of neonatal tolerance in murine experimental models and subsequently analyze tolerance induction in human newborn infants. Hematopoietic mixed chimerism in neonates is also discussed based on in utero hematopoietic stem cell (HSC) transplant studies. Then, we review the recent advances in tolerance induction approaches in adults, including the infusion of HSCs associated with less toxic conditioning regimens, regulatory T cells/facilitating cells/mesenchymal stem cells transplantation, costimulatory blockade, and thymus manipulation. Finally, IS withdrawal in pediatric solid organ transplant is discussed. In conclusion, the establishment of transplant tolerance induction in infants is promising and deserves further investigations. Future studies could focus on the selection of patients, on less toxic conditioning regimens, and on biomarkers for IS minimization or withdrawal.

20.
Transpl Immunol ; 39: 74-83, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27561237

RESUMEN

Lymphodepletive agents play important role in different clinical applications or experimental transplant studies. In order to facilitate preclinical pediatric transplant studies, we have developed the rabbit anti-pig thymocyte globulin (pATG) and studied its effects in neonatal swines. In vitro assays showed that pATG can bind to lymphocytes and neutrophils in a dose-dependent manner and lyse peripheral blood mononuclear cells by apoptosis and complement-dependent cytotoxicity. In vivo, pATG as a monotherapy was administered at different doses (2.5, 5, 20, 40 and 80mg/kg) in newborn pigs. Results showed that pATG induced a dose-dependent but transient T-cell depletion in peripheral blood. Lymphodepletion was also observed in lymph nodes, spleen and thymus. Pharmacokinetic studies showed dose-related cell-bound pATG on lymphocytes, as well as the presence of free pATG in the serum. Both cell-bound and free pATG levels decreased gradually after administration. Interestingly, adjuvant mycophenolate mofetil (MMF) given at 1g/m2/day for 1week successfully maintained pATG-induced T-cell depletion. In conclusion, pATG administration can cause transient T-cell depletion in neonatal pigs and this effect can be maintained by MMF. Therefore, we have developed an original immunosuppressive regimen that can be used for transplantation studies in swine model.


Asunto(s)
Suero Antilinfocítico/uso terapéutico , Depleción Linfocítica/métodos , Linfocitos/inmunología , Neutrófilos/inmunología , Adyuvantes Inmunológicos/uso terapéutico , Animales , Animales Recién Nacidos , Citotoxicidad Celular Dependiente de Anticuerpos , Apoptosis , Células Cultivadas , Proteínas del Sistema Complemento/metabolismo , Relación Dosis-Respuesta Inmunológica , Estudios de Factibilidad , Humanos , Modelos Animales , Ácido Micofenólico/uso terapéutico , Conejos , Porcinos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA