Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 130
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Annu Rev Immunol ; 32: 25-50, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24215318

RESUMEN

CD47 is a broadly expressed membrane protein that interacts with the myeloid inhibitory immunoreceptor SIRPα (also termed CD172a or SHPS-1). SIRPα is the prototypic member of the SIRP paired receptor family of closely related SIRP proteins. Engagement of SIRPα by CD47 provides a downregulatory signal that inhibits host cell phagocytosis, and CD47 therefore functions as a "don't-eat-me" signal. Here, we discuss recent structural analysis of CD47-SIRPα interactions and implications of this for the function and evolution of SIRPα and paired receptors in general. Furthermore, we review the proposed roles of CD47-SIRPα interactions in phagocytosis, (auto)immunity, and host defense, as well as its potential significance as a therapeutic target in cancer and inflammation and for improving graft survival in xenotransplantation.


Asunto(s)
Antígenos de Diferenciación/metabolismo , Antígeno CD47/metabolismo , Receptores Inmunológicos/metabolismo , Animales , Antígenos de Diferenciación/química , Antígenos de Diferenciación/genética , Enfermedades Autoinmunes/tratamiento farmacológico , Enfermedades Autoinmunes/etiología , Antígeno CD47/química , Antígeno CD47/genética , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Enfermedades Hematológicas/tratamiento farmacológico , Enfermedades Hematológicas/etiología , Humanos , Sinapsis Inmunológicas/inmunología , Sinapsis Inmunológicas/metabolismo , Terapia Molecular Dirigida , Familia de Multigenes , Neoplasias/tratamiento farmacológico , Neoplasias/etiología , Fagocitosis/efectos de los fármacos , Fagocitosis/inmunología , Unión Proteica/efectos de los fármacos , Receptores Inmunológicos/química , Receptores Inmunológicos/genética , Transducción de Señal , Linfocitos T/inmunología , Linfocitos T/metabolismo
2.
Immunol Rev ; 314(1): 280-301, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36331258

RESUMEN

Tumor-targeting monoclonal antibodies are available for a number of cancer cell types (over)expressing the corresponding tumor antigens. Such antibodies can limit tumor progression by different mechanisms, including direct growth inhibition and immune-mediated mechanisms, in particular complement-dependent cytotoxicity, antibody-dependent cellular phagocytosis, and antibody-dependent cellular cytotoxicity (ADCC). ADCC can be mediated by various types of immune cells, including neutrophils, the most abundant leukocyte in circulation. Neutrophils express a number of Fc receptors, including Fcγ- and Fcα-receptors, and can therefore kill tumor cells opsonized with either IgG or IgA antibodies. In recent years, important insights have been obtained with respect to the mechanism(s) by which neutrophils engage and kill antibody-opsonized cancer cells and these findings are reviewed here. In addition, we consider a number of additional ways in which neutrophils may affect cancer progression, in particular by regulating adaptive anti-cancer immunity.


Asunto(s)
Neoplasias , Neutrófilos , Humanos , Citotoxicidad Celular Dependiente de Anticuerpos , Receptores Fc , Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Monoclonales/farmacología , Receptores de IgG/metabolismo
3.
Immunity ; 43(4): 622-4, 2015 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-26488810

RESUMEN

Whereas dendritic cells (DCs) can be activated in many ways to trigger immunity, hardly anything is known about the mechanisms that counterbalance DC activation. Cyster and colleagues now demonstrate that the "self" molecule CD47 on erythrocytes critically restricts splenic DC activation.


Asunto(s)
Inmunidad Adaptativa , Antígeno CD47/sangre , Células Dendríticas/inmunología , Eritrocitos/inmunología , Receptores Inmunológicos/inmunología , Autotolerancia/inmunología , Bazo/inmunología , Animales
4.
Blood ; 135(24): 2171-2181, 2020 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-32128589

RESUMEN

Megakaryoblastic leukemia 1 (MKL1) promotes the regulation of essential cell processes, including actin cytoskeletal dynamics, by coactivating serum response factor. Recently, the first human with MKL1 deficiency, leading to a novel primary immunodeficiency, was identified. We report a second family with 2 siblings with a homozygous frameshift mutation in MKL1. The index case died as an infant from progressive and severe pneumonia caused by Pseudomonas aeruginosa and poor wound healing. The younger sibling was preemptively transplanted shortly after birth. The immunodeficiency was marked by a pronounced actin polymerization defect and a strongly reduced motility and chemotactic response by MKL1-deficient neutrophils. In addition to the lack of MKL1, subsequent proteomic and transcriptomic analyses of patient neutrophils revealed actin and several actin-related proteins to be downregulated, confirming a role for MKL1 as a transcriptional coregulator. Degranulation was enhanced upon suboptimal neutrophil activation, whereas production of reactive oxygen species was normal. Neutrophil adhesion was intact but without proper spreading. The latter could explain the observed failure in firm adherence and transendothelial migration under flow conditions. No apparent defect in phagocytosis or bacterial killing was found. Also, monocyte-derived macrophages showed intact phagocytosis, and lymphocyte counts and proliferative capacity were normal. Nonhematopoietic primary fibroblasts demonstrated defective differentiation into myofibroblasts but normal migration and F-actin content, most likely as a result of compensatory mechanisms of MKL2, which is not expressed in neutrophils. Our findings extend current insight into the severe immune dysfunction in MKL1 deficiency, with cytoskeletal dysfunction and defective extravasation of neutrophils as the most prominent features.


Asunto(s)
Citoesqueleto de Actina/metabolismo , Mutación del Sistema de Lectura , Neutrófilos/fisiología , Enfermedades de Inmunodeficiencia Primaria/genética , Enfermedades de Inmunodeficiencia Primaria/metabolismo , Transactivadores/deficiencia , Transactivadores/genética , Citoesqueleto de Actina/química , Movimiento Celular/genética , Movimiento Celular/fisiología , Consanguinidad , Femenino , Fibroblastos/metabolismo , Perfilación de la Expresión Génica , Trasplante de Células Madre Hematopoyéticas , Humanos , Lactante , Masculino , Linaje , Polimerizacion , Enfermedades de Inmunodeficiencia Primaria/terapia , Proteómica , Factores de Transcripción/metabolismo
5.
Immunol Rev ; 276(1): 145-164, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28258703

RESUMEN

Immune checkpoint inhibitors, including those targeting CTLA-4/B7 and the PD-1/PD-L1 inhibitory pathways, are now available for clinical use in cancer patients, with other interesting checkpoint inhibitors being currently in development. Most of these have the purpose to promote adaptive T cell-mediated immunity against cancer. Here, we review another checkpoint acting to potentiate the activity of innate immune cells towards cancer. This innate immune checkpoint is composed of what has become known as the 'don't-eat me' signal CD47, which is a protein broadly expressed on normal cells and often overexpressed on cancer cells, and its counter-receptor, the myeloid inhibitory immunoreceptor SIRPα. Blocking CD47-SIRPα interactions has been shown to promote the destruction of cancer cells by phagocytes, including macrophages and neutrophils. Furthermore, there is growing evidence that targeting of the CD47-SIRPα axis may also promote antigen-presenting cell function and thereby stimulate adaptive T cell-mediated anti-cancer immunity. The development of CD47-SIRPα checkpoint inhibitors and the potential side effects that these may have are discussed. Collectively, this identifies the CD47-SIRPα axis as a promising innate immune checkpoint in cancer, and with data of the first clinical studies with CD47-SIRPα checkpoint inhibitors expected within the coming years, this is an exciting and rapidly developing field.


Asunto(s)
Anticuerpos Bloqueadores/uso terapéutico , Antígenos de Diferenciación/metabolismo , Antígenos de Neoplasias/metabolismo , Antígeno CD47/metabolismo , Inmunoterapia/métodos , Células Mieloides/metabolismo , Neoplasias/terapia , Receptores Inmunológicos/metabolismo , Linfocitos T/inmunología , Animales , Presentación de Antígeno , Antígenos de Diferenciación/inmunología , Antígenos de Neoplasias/inmunología , Antígeno CD47/inmunología , Ensayos Clínicos como Asunto , Humanos , Inmunidad Innata , Activación de Linfocitos , Neoplasias/inmunología , Fagocitosis , Receptores Inmunológicos/inmunología
6.
Semin Immunol ; 28(2): 94-108, 2016 04.
Artículo en Inglés | MEDLINE | ID: mdl-26976825

RESUMEN

Neutrophils play a critical role in the host defense against infection, and they are able to perform a variety of effector mechanisms for this purpose. However, there are also a number of pathological conditions, including autoimmunity and cancer, in which the activities of neutrophils can be harmful to the host. Thus the activities of neutrophils need to be tightly controlled. As in the case of other immune cells, many of the neutrophil effector functions are regulated by a series of immunoreceptors on the plasma membrane. Here, we review what is currently known about the functions of the various individual immunoreceptors and their signaling in neutrophils. While these immunoreceptors allow for the recognition of a diverse range of extracellular ligands, such as cell surface structures (like proteins, glycans and lipids) and extracellular matrix components, they commonly signal via conserved ITAM or ITIM motifs and their associated downstream pathways that depend on the phosphorylation of tyrosine residues in proteins and/or inositol lipids. This allows for a balanced homeostatic regulation of neutrophil effector functions. Given the number of available immunoreceptors and their fundamental importance for neutrophil behavior, it is perhaps not surprising that pathogens have evolved means to evade immune responses through some of these pathways. Inversely, some of these receptors evolved to specifically recognize these pathogens. Finally, some interactions mediated by immunoreceptors in neutrophils have been identified as promising targets for therapeutic intervention.


Asunto(s)
Neutrófilos/inmunología , Neutrófilos/metabolismo , Receptores Inmunológicos/metabolismo , Animales , Biomarcadores , Susceptibilidad a Enfermedades , Regulación de la Expresión Génica , Humanos , Inmunidad Innata , Activación Neutrófila/genética , Activación Neutrófila/inmunología , Neutrófilos/química , Unión Proteica , Receptores Inmunológicos/genética , Transducción de Señal
7.
Immunol Rev ; 273(1): 299-311, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27558342

RESUMEN

Neutrophils play a critical role in the prevention of invasive fungal infections. Whereas mouse studies have demonstrated the role of various neutrophil pathogen recognition receptors (PRRs), signal transduction pathways, and cytotoxicity in the murine antifungal immune response, much less is known about the killing of fungi by human neutrophils. Recently, novel primary immunodeficiencies have been identified in patients with a susceptibility to fungal infections. These human 'knock-out' neutrophils expand our knowledge to understand the role of PRRs and signaling in human fungal killing. From the studies with these patients it is becoming clear that neutrophils employ fundamentally distinct mechanisms to kill Candida albicans or Aspergillus fumigatus.


Asunto(s)
Aspergillus fumigatus/inmunología , Candida albicans/inmunología , Citotoxicidad Inmunológica , Micosis/inmunología , Neutrófilos/inmunología , Animales , Humanos , Inmunidad Innata , Ratones , Neutrófilos/microbiología , Receptores de Reconocimiento de Patrones/metabolismo , Transducción de Señal
8.
Immunol Rev ; 273(1): 312-28, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27558343

RESUMEN

Neutrophils play an important role in cancer. This does not only relate to the well-established prognostic value of the presence of neutrophils, either in the blood or in tumor tissue, in the context of cancer progression or for the monitoring of therapy, but also to their active role in the progression of cancer. In the current review, we describe what is known in general about the role of neutrophils in cancer. What is emerging is a complex, rather heterogeneous picture with both pro- and anti-tumorigenic roles, which apparently differs with cancer type and disease stage. Furthermore, we will discuss the well-known role of neutrophils as myeloid-derived suppressor cells (MDSC), and also on the role of neutrophils as important effector cells during antibody therapy in cancer. It is clear that neutrophils contribute substantially to cancer progression in multiple ways, and this includes both direct effects on the cancer cells and indirect effect on the tumor microenvironment. While in many cases neutrophils have been shown to promote tumor progression, for instance by acting as MDSC, there are also protective effects, particularly when antibody immunotherapy is performed. A better understanding of the role of neutrophils is likely to provide opportunities for immunomodulation and for improving the treatment of cancer patients.


Asunto(s)
Inmunoterapia/métodos , Células Supresoras de Origen Mieloide/inmunología , Neoplasias/inmunología , Neutrófilos/inmunología , Microambiente Tumoral , Animales , Anticuerpos/uso terapéutico , Carcinogénesis , Humanos , Inmunomodulación , Neoplasias/terapia
9.
Br J Haematol ; 186(6): 887-899, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31168801

RESUMEN

Most sickle cell disease (SCD) patients rely on blood transfusion as their main treatment strategy. However, frequent blood transfusion poses the risk of alloimmunization. On average, 30% of SCD patients will alloimmunize while other patient groups form antibodies less frequently. Identification of genetic markers may help to predict which patients are at risk to form alloantibodies. The aim of this study was to evaluate whether genetic variations in the Toll-like receptor pathway or in genes previously associated with antibody-mediated conditions are associated with red blood cell (RBC) alloimmunization in a cohort of SCD patients. In this case-control study, cases had a documented history of alloimmunization while controls had received ≥20 RBC units without alloantibody formation. We used a customized single nucleotide polymorphism (SNP) panel to genotype 690 SNPs in 275 (130 controls, 145 cases) patients. Frequencies were compared using multiple logistic regression analysis. In our primary analysis, no SNPs were found to be significantly associated with alloimmunization after correction for multiple testing. However, in a secondary analysis with a less stringent threshold for significance we found 19 moderately associated SNPs. Among others, SNPs in TLR1/TANK and MALT1 were associated with a higher alloimmunization risk, while SNPs in STAM/IFNAR1 and STAT4 conferred a lower alloimmunization risk.


Asunto(s)
Anemia de Células Falciformes/genética , Genotipo , Polimorfismo de Nucleótido Simple , Reacción a la Transfusión/genética , Adulto , Anemia de Células Falciformes/inmunología , Anemia de Células Falciformes/terapia , Transfusión de Eritrocitos/efectos adversos , Femenino , Estudios de Seguimiento , Marcadores Genéticos , Humanos , Inmunización , Isoanticuerpos/inmunología , Masculino , Estudios Retrospectivos , Factores de Riesgo , Reacción a la Transfusión/inmunología
10.
Eur J Immunol ; 48(2): 344-354, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-28952147

RESUMEN

The efficacy of cancer therapeutic antibodies varies considerably among patients. Anti-cancer antibodies act through different mechanisms, including antibody-dependent cellular cytotoxicity (ADCC) triggered via Fcγ receptors (FcγR). This phagocyte ADCC can be promoted by interference with CD47-SIRPα interactions, but the magnitude of this enhancement also varies among individuals. Both FcγR and SIRPα display considerable genetic variation, and we investigated whether this explains some of the variability in ADCC. Because of linkage disequilibrium between FcγR variants the interpretation of previous reports suggesting a potential link between FcγR polymorphisms and ADCC has been troublesome. We performed an integrated genetic analysis that enables stratification. ADCC by activated human neutrophils towards Trastuzumab-coated breast cancer cells was predominantly dependent on FcγRIIa. Neutrophils from individuals with the FcγRIIa-131H polymorphic variant displayed significantly higher killing capacity relative to those with FcγRIIa-131R. Furthermore, ADCC was consistently enhanced by targeting CD47-SIRPα interactions, and there were no significant functional differences between the two most prevalent SIRPα polymorphic variants. Thus, neutrophil ADCC capacity is directly related to the FcγRIIa polymorphism, and targeting CD47-SIRPα interactions enhances ADCC independently of FcγR and SIRPα genotype, thereby further suggesting that CD47-SIRPα interference might be a generic strategy for potentiating the efficacy of antibody therapy in cancer.


Asunto(s)
Citotoxicidad Celular Dependiente de Anticuerpos/genética , Antígenos de Diferenciación/genética , Neoplasias de la Mama/genética , Genotipo , Inmunoterapia/métodos , Neutrófilos/fisiología , Receptores de IgG/genética , Receptores Inmunológicos/genética , Antígenos de Diferenciación/metabolismo , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Neoplasias de la Mama/inmunología , Antígeno CD47/metabolismo , Línea Celular Tumoral , Femenino , Predisposición Genética a la Enfermedad , Humanos , Desequilibrio de Ligamiento , Polimorfismo Genético , Receptor ErbB-2/inmunología , Receptores de IgG/metabolismo , Receptores Inmunológicos/metabolismo , Trastuzumab/farmacología , Trastuzumab/uso terapéutico , Resultado del Tratamiento
11.
Blood ; 130(19): 2121-2130, 2017 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-28899854

RESUMEN

Red blood cell (RBC) transfusions are of vital importance in patients with sickle cell disease (SCD). However, a major complication of transfusion therapy is alloimmunization. The low-affinity Fcγ receptors, expressed on immune cells, are important regulators of antibody responses. Genetic variation in FCGR genes has been associated with various auto- and alloimmune diseases. The aim of this study was to evaluate the association between genetic variation of FCGR and RBC alloimmunization in SCD. In this case-control study, DNA samples from 2 cohorts of transfused SCD patients were combined (France and The Netherlands). Cases had a positive history of alloimmunization, having received ≥1 RBC unit. Controls had a negative history of alloimmunization, having received ≥20 RBC units. Single nucleotide polymorphisms and copy number variation of the FCGR2/3 gene cluster were studied in a FCGR-specific multiplex ligation-dependent probe amplification assay. Frequencies were compared using logistic regression. Two hundred seventy-two patients were included (130 controls, 142 cases). The nonclassical open reading frame in the FCGR2C gene (FCGR2C.nc-ORF) was strongly associated with a decreased alloimmunization risk (odds ratio [OR] 0.26, 95% confidence [CI] 0.11-0.64). This association persisted when only including controls with exposure to ≥100 units (OR 0.30, CI 0.11-0.85) and appeared even stronger when excluding cases with Rh or K antibodies only (OR 0.19, CI 0.06-0.59). In conclusion, SCD patients with the FCGR2Cnc-ORF polymorphism have over a 3-fold lower risk for RBC alloimmunization in comparison with patients without this mutation. This protective effect was strongest for exposure to antigens other than the immunogenic Rh or K antigens.


Asunto(s)
Anemia de Células Falciformes/genética , Transfusión de Eritrocitos/efectos adversos , Eritrocitos , Haplotipos/genética , Inmunización , Polimorfismo Genético , Receptores de IgG/genética , Adulto , Anemia de Células Falciformes/inmunología , Anemia de Células Falciformes/terapia , Femenino , Estudios de Seguimiento , Humanos , Masculino , Familia de Multigenes , Receptores de IgG/inmunología , Factores de Riesgo
12.
J Immunol ; 199(1): 204-211, 2017 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-28566370

RESUMEN

Abs of the IgG isotype are glycosylated in their Fc domain at a conserved asparagine at position 297. Removal of the core fucose of this glycan greatly increases the affinity for FcγRIII, resulting in enhanced FcγRIII-mediated effector functions. Normal plasma IgG contains ∼94% fucosylated Abs, but alloantibodies against, for example, Rhesus D (RhD) and platelet Ags frequently have reduced fucosylation that enhances their pathogenicity. The increased FcγRIII-mediated effector functions have been put to use in various afucosylated therapeutic Abs in anticancer treatment. To test the functional consequences of Ab fucosylation, we produced V-gene-matched recombinant anti-RhD IgG Abs of the four different subclasses (IgG1-4) with and without core fucose (i.e., 20% fucose remaining). Binding to all human FcγR types and their functional isoforms was assessed with surface plasmon resonance. All hypofucosylated anti-RhD IgGs of all IgG subclasses indeed showed enhanced binding affinity for isolated FcγRIII isoforms, without affecting binding affinity to other FcγRs. In contrast, when testing hypofucosylated anti-RhD Abs with FcγRIIIa-expressing NK cells, a 12- and 7-fold increased erythrocyte lysis was observed with the IgG1 and IgG3, respectively, but no increase with IgG2 and IgG4 anti-RhD Abs. Notably, none of the hypofucosylated IgGs enhanced effector function of macrophages, which, in contrast to NK cells, express a complex set of FcγRs, including FcγRIIIa. Our data suggest that the beneficial effects of afucosylated biologicals for clinical use can be particularly anticipated when there is a substantial involvement of FcγRIIIa-expressing cells, such as NK cells.


Asunto(s)
Fucosa/química , Inmunoglobulina G/química , Inmunoglobulina G/inmunología , Receptores de IgG/inmunología , Citotoxicidad Celular Dependiente de Anticuerpos , Fucosa/inmunología , Fucosa/metabolismo , Proteínas Ligadas a GPI/química , Proteínas Ligadas a GPI/genética , Proteínas Ligadas a GPI/inmunología , Glicosilación , Humanos , Fragmentos Fc de Inmunoglobulinas/química , Fragmentos Fc de Inmunoglobulinas/inmunología , Inmunoglobulina G/aislamiento & purificación , Inmunoglobulina G/metabolismo , Células Asesinas Naturales/inmunología , Macrófagos/inmunología , Unión Proteica , Receptores de IgG/química , Receptores de IgG/genética , Sistema del Grupo Sanguíneo Rh-Hr/inmunología , Resonancia por Plasmón de Superficie
13.
Platelets ; 30(3): 368-379, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-29553857

RESUMEN

Platelet concentrates (PCs) represent a blood transfusion product with a major concern for safety as their storage temperature (20-24°C) allows bacterial growth, and their maximum storage time period (less than a week) precludes complete microbiological testing. Pathogen inactivation technologies (PITs) provide an additional layer of safety to the blood transfusion products from known and unknown pathogens such as bacteria, viruses, and parasites. In this context, PITs, such as Mirasol Pathogen Reduction Technology (PRT), have been developed and are implemented in many countries. However, several studies have shown in vitro that Mirasol PRT induces a certain level of platelet shape change, hyperactivation, basal degranulation, and increased oxidative damage during storage. It has been suggested that Mirasol PRT might accelerate what has been described as the platelet storage lesion (PSL), but supportive molecular signatures have not been obtained. We aimed at dissecting the influence of both variables, that is, Mirasol PRT and storage time, at the proteome level. We present comprehensive proteomics data analysis of Control PCs and PCs treated with Mirasol PRT at storage days 1, 2, 6, and 8. Our workflow was set to perform proteomics analysis using a gel-free and label-free quantification (LFQ) approach. Semi-quantification was based on LFQ signal intensities of identified proteins using MaxQuant/Perseus software platform. Data are available via ProteomeXchange with identifier PXD008119. We identified marginal differences between Mirasol PRT and Control PCs during storage. However, those significant changes at the proteome level were specifically related to the functional aspects previously described to affect platelets upon Mirasol PRT. In addition, the effect of Mirasol PRT on the platelet proteome appeared not to be exclusively due to an accelerated or enhanced PSL. In summary, semi-quantitative proteomics allows to discern between proteome changes due to Mirasol PRT or PSL, and proves to be a methodology suitable to phenotype platelets in an unbiased manner, in various physiological contexts.


Asunto(s)
Plaquetas/metabolismo , Pruebas de Función Plaquetaria/métodos , Proteoma/metabolismo , Proteómica/métodos , Humanos
14.
J Med Genet ; 55(3): 166-172, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29331982

RESUMEN

BACKGROUND: Mutations in the NCF1 gene that encodes p47phox, a subunit of the NADPH oxidase complex, cause chronic granulomatous disease (CGD). In Kavkazi Jews, a c.579G>A (p.Trp193Ter) mutation in NCF1 is frequently found, leading to CGD. The same mutation is found in about 1% of Ashkenazi Jews, although Ashkenazi CGD patients with this mutation have never been described. METHODS: We used Sanger sequencing, multiplex ligation-dependent probe amplification (MLPA), gene scan analysis and Ion Torrent Next Generation Sequencing for genetic analysis, and measured NADPH oxidase activity and p47phox expression. RESULTS: In an Ashkenazi couple expecting a baby, both parents were found to be heterozygotes for this mutation, as was the fetus. However, segregation analysis in the extended family was consistent with the fetus inheriting both carrier alleles from the parents. MLPA indicated four complete NCF1 genes in the fetus and three in each parent. Gene sequencing confirmed these results. Analysis of fetal leucocytes obtained by cordocentesis revealed substantial oxidase activity with three different assays, which was confirmed after birth. In six additional Ashkenazi carriers of the NCF1 c.579G>A mutation, we found five individuals with three complete NCF1 genes of which one was mutated (like the parents), and one individual with in addition a fusion gene of NCF1 with a pseudogene. CONCLUSION: These results point to the existence of a 'false-carrier' state in Ashkenazi Jews and have wide implications regarding pre-pregnancy screening in this and other population groups.


Asunto(s)
Enfermedad Granulomatosa Crónica/genética , Heterocigoto , Judíos/genética , NADPH Oxidasas/genética , Alelos , Exones/genética , Femenino , Tamización de Portadores Genéticos , Pruebas Genéticas , Enfermedad Granulomatosa Crónica/patología , Humanos , Masculino , Mutación , Embarazo
15.
J Immunol ; 196(3): 1272-83, 2016 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-26718340

RESUMEN

Neutrophils are known to play a pivotal role in the host defense against Aspergillus infections. This is illustrated by the prevalence of Aspergillus infections in patients with neutropenia or phagocyte functional defects, such as chronic granulomatous disease. However, the mechanisms by which human neutrophils recognize and kill Aspergillus are poorly understood. In this work, we have studied in detail which neutrophil functions, including neutrophil extracellular trap (NET) formation, are involved in the killing of Aspergillus fumigatus conidia and hyphae, using neutrophils from patients with well-defined genetic immunodeficiencies. Recognition of conidia involves integrin CD11b/CD18 (and not dectin-1), which triggers a PI3K-dependent nonoxidative intracellular mechanism of killing. When the conidia escape from early killing and germinate, the extracellular destruction of the Aspergillus hyphae needs opsonization by Abs and involves predominantly recognition via Fcγ receptors, signaling via Syk, PI3K, and protein kinase C to trigger the production of toxic reactive oxygen metabolites by the NADPH oxidase and myeloperoxidase. A. fumigatus induces NET formation; however, NETs did not contribute to A. fumigatus killing. Thus, our findings reveal distinct killing mechanisms of Aspergillus conidia and hyphae by human neutrophils, leading to a comprehensive insight in the innate antifungal response.


Asunto(s)
Aspergilosis/inmunología , Aspergillus fumigatus/inmunología , Hifa/inmunología , Neutrófilos/inmunología , Esporas Fúngicas/inmunología , Citotoxicidad Inmunológica/inmunología , Trampas Extracelulares/inmunología , Humanos , Inmunidad Innata , Síndromes de Inmunodeficiencia/inmunología , Microscopía Confocal , Fagocitos/inmunología
16.
J Immunol ; 197(11): 4312-4324, 2016 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-27815426

RESUMEN

Dendritic cells (DCs) play a pivotal role in the regulation of the immune response. DC development and activation is finely orchestrated through transcriptional programs. GATA1 transcription factor is required for murine DC development, and data suggest that it might be involved in the fine-tuning of the life span and function of activated DCs. We generated DC-specific Gata1 knockout mice (Gata1-KODC), which presented a 20% reduction of splenic DCs, partially explained by enhanced apoptosis. RNA sequencing analysis revealed a number of deregulated genes involved in cell survival, migration, and function. DC migration toward peripheral lymph nodes was impaired in Gata1-KODC mice. Migration assays performed in vitro showed that this defect was selective for CCL21, but not CCL19. Interestingly, we show that Gata1-KODC DCs have reduced polysialic acid levels on their surface, which is a known determinant for the proper migration of DCs toward CCL21.


Asunto(s)
Movimiento Celular/inmunología , Quimiocina CCL21/inmunología , Células Dendríticas/inmunología , Factor de Transcripción GATA1/inmunología , Ganglios Linfáticos/inmunología , Ácidos Siálicos/inmunología , Animales , Movimiento Celular/genética , Quimiocina CCL19/genética , Quimiocina CCL19/inmunología , Quimiocina CCL21/genética , Células Dendríticas/citología , Factor de Transcripción GATA1/deficiencia , Ganglios Linfáticos/citología , Ratones , Ratones Noqueados , Ácidos Siálicos/genética
17.
Hum Mutat ; 38(10): 1402-1411, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28585318

RESUMEN

Hermansky-Pudlak syndrome type 2 (HPS2) is a syndrome caused by mutations in the beta-3A subunit of the adaptor protein (AP)-3 complex (AP3B1 gene). We describe five unreported cases with four novel mutations, one of which caused aberrant pre-mRNA splicing. A point mutation c.2702C>G in exon 23 of the AP3B1 gene caused deletion of 112 bp in the mRNA in two siblings. This mutation activates a cryptic donor splice site that overrules the wild-type donor splice site of this exon. Three other novel mutations in AP3B1 were identified, that is, a nonsense mutation c.716G>A (p.Trp239Ter), a 1-bp and a 4-bp deletion c.177delA and c.1839_1842delTAGA, respectively, both causing frameshift and premature termination of translation. Mass spectrometry in four of these HPS2 patients demonstrated the (near) absence of all AP-3 complex subunits. Immunoelectron microscopy on the neutrophils of two of these patients showed abnormal granule formation. We found clear mislocalization of myeloperoxidase in the neutrophils even though the content of this protein but not the activity seemed to be present at normal levels. In sum, HPS2 is the result of the absence of the entire AP-3 complex, which results in severe neutropenia with a defect in granule formation as the major hematological finding.


Asunto(s)
Complejo 3 de Proteína Adaptadora/genética , Subunidades beta de Complejo de Proteína Adaptadora/genética , Síndrome de Hermanski-Pudlak/genética , Precursores del ARN/genética , Empalme del ARN/genética , Adolescente , Adulto , Niño , Preescolar , Codón sin Sentido/genética , Exones/genética , Femenino , Síndrome de Hermanski-Pudlak/fisiopatología , Humanos , Lactante , Masculino , Persona de Mediana Edad , Neutrófilos/metabolismo , Neutrófilos/patología , Fenotipo , Mutación Puntual , Sitios de Empalme de ARN/genética , Eliminación de Secuencia/genética
18.
Blood ; 125(12): 1957-67, 2015 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-25538045

RESUMEN

Sp1 and Sp3 belong to the specificity proteins (Sp)/Krüppel-like transcription factor family. They are closely related, ubiquitously expressed, and recognize G-rich DNA motifs. They are thought to regulate generic processes such as cell-cycle and growth control, metabolic pathways, and apoptosis. Ablation of Sp1 or Sp3 in mice is lethal, and combined haploinsufficiency results in hematopoietic defects during the fetal stages. Here, we show that in adult mice, conditional pan-hematopoietic (Mx1-Cre) ablation of either Sp1 or Sp3 has minimal impact on hematopoiesis, whereas the simultaneous loss of Sp1 and Sp3 results in severe macrothrombocytopenia. This occurs in a cell-autonomous manner as shown by megakaryocyte-specific (Pf4-Cre) double-knockout mice. We employed flow cytometry, cell culture, and electron microscopy and show that although megakaryocyte numbers are normal in bone marrow and spleen, they display a less compact demarcation membrane system and a striking inability to form proplatelets. Through megakaryocyte transcriptomics and platelet proteomics, we identified several cytoskeleton-related proteins and downstream effector kinases, including Mylk, that were downregulated upon Sp1/Sp3 depletion, providing an explanation for the observed defects in megakaryopoiesis. Supporting this notion, selective Mylk inhibition by ML7 affected proplatelet formation and stabilization and resulted in defective ITAM receptor-mediated platelet aggregation.


Asunto(s)
Plaquetas/citología , Megacariocitos/citología , Factor de Transcripción Sp1/genética , Factor de Transcripción Sp3/genética , Animales , Azepinas/química , Plaquetas/metabolismo , Médula Ósea/metabolismo , Citometría de Flujo , Lectinas Tipo C/metabolismo , Ratones , Ratones Noqueados , Naftalenos/química , Agregación Plaquetaria , Glicoproteínas de Membrana Plaquetaria/metabolismo , Proteoma , Transducción de Señal , Factor de Transcripción Sp1/metabolismo , Factor de Transcripción Sp3/metabolismo , Bazo/metabolismo , Trombocitopenia/metabolismo , Factores de Transcripción/metabolismo
19.
Transfusion ; 57(3): 674-684, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28032635

RESUMEN

BACKGROUND: Granulocyte transfusion (GTX) is a potential approach to correcting neutropenia and relieving the increased risk of infection in patients who are refractory to antibiotics. To mobilize enough granulocytes for transfusion, healthy donors are premedicated with granulocyte-colony-stimulating factor (G-CSF) and dexamethasone. Granulocytes have a short circulatory half-life. Consequently, patients need to receive GTX every other day to keep circulating granulocyte counts at an acceptable level. We investigated whether plasma from premedicated donors was capable of prolonging neutrophil survival and, if so, which factor could be held responsible. STUDY DESIGN AND METHODS: The effects of plasma from G-CSF/dexamethasone-treated donors on neutrophil survival were assessed by annexin-V, CD16. and CXCR4 staining and nuclear morphology. We isolated an albumin-bound protein using α-chymotrypsin and albumin-depletion and further characterized it using protein analysis. The effects of dexamethasone and G-CSF were assessed using mifepristone and G-CSF-neutralizing antibody. G-CSF plasma concentrations were determined by Western blot and Luminex analyses. RESULTS: G-CSF/dexamethasone plasma contained a survival-promoting factor for at least 2 days. This factor was recognized as an albumin-associated protein and was identified as G-CSF itself, which was surprising considering its reported half-life of only 4.5 hours. Compared with coadministration of dexamethasone, administration of G-CSF alone to the same GTX donors led to a faster decline in circulating G-CSF levels, whereas dexamethasone itself did not induce any G-CSF, demonstrating a role for dexamethasone in increasing G-CSF half-life. CONCLUSION: Dexamethasone increases granulocyte yield upon coadministration with G-CSF by extending G-CSF half-life. This observation might also be exploited in the coadministration of dexamethasone with other recombinant proteins to modulate their half-life.


Asunto(s)
Dexametasona , Transfusión de Leucocitos , Neutrófilos/metabolismo , Adulto , Anexina A5/metabolismo , Supervivencia Celular/efectos de los fármacos , Dexametasona/administración & dosificación , Dexametasona/farmacocinética , Filgrastim/administración & dosificación , Filgrastim/farmacocinética , Proteínas Ligadas a GPI/metabolismo , Semivida , Humanos , Masculino , Persona de Mediana Edad , Mifepristona/farmacología , Neutrófilos/citología , Plasma/metabolismo , Receptores CXCR4/metabolismo , Receptores de IgG/metabolismo
20.
J Immunol ; 194(8): 3909-16, 2015 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-25750432

RESUMEN

Macrophages form a heterogeneous population of immune cells, which is critical for both the initiation and resolution of inflammation. They can be skewed to a proinflammatory subtype by the Th1 cytokine IFN-γ and further activated with TLR triggers, such as LPS. In this work, we investigated the effects of IFN-γ priming on LPS-induced gene expression in primary mouse macrophages. Surprisingly, we found that IFN-γ priming represses a subset of LPS-induced genes, particularly genes involved in cellular movement and leukocyte recruitment. We found STAT1-binding motifs enriched in the promoters of these repressed genes. Furthermore, in the absence of STAT1, affected genes are derepressed. We also observed epigenetic remodeling by IFN-γ priming on enhancer or promoter sites of repressed genes, which resulted in less NF-κB p65 recruitment to these sites without effects on global NF-κB activation. Finally, the epigenetic and transcriptional changes induced by IFN-γ priming reduce neutrophil recruitment in vitro and in vivo. Our data show that IFN-γ priming changes the inflammatory repertoire of macrophages, leading to a change in neutrophil recruitment to inflammatory sites.


Asunto(s)
Movimiento Celular/inmunología , Epigénesis Genética/inmunología , Interferón gamma/inmunología , Macrófagos/inmunología , Neutrófilos/inmunología , Animales , Movimiento Celular/efectos de los fármacos , Epigénesis Genética/efectos de los fármacos , Femenino , Inflamación/inmunología , Lipopolisacáridos/farmacología , Ratones , Ratones Noqueados , Elementos de Respuesta/inmunología , Factor de Transcripción STAT1/inmunología , Receptores Toll-Like/agonistas , Receptores Toll-Like/inmunología , Factor de Transcripción ReIA/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA