Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
2.
Front Cell Infect Microbiol ; 13: 1165756, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37342247

RESUMEN

Introduction: Increasing evidence has shown that coronavirus disease 19 (COVID-19) severity is driven by a dysregulated immunological response. Previous studies have demonstrated that natural killer (NK) cell dysfunction underpins severe illness in COVID-19 patients, but have lacked an in-depth analysis of NK cell markers as a driver of death in the most critically ill patients. Methods: We enrolled 50 non-vaccinated hospitalized patients infected with the initial virus or the alpha variant of SARS-CoV-2 with moderate or severe illness, to evaluate phenotypic and functional features of NK cells. Results: Here, we show that, consistent with previous studies, evolution NK cells from COVID-19 patients are more activated, with the decreased activation of natural cytotoxicity receptors and impaired cytotoxicity and IFN-γ production, in association with disease regardless of the SARS-CoV-2 strain. Fatality was observed in 6 of 17 patients with severe disease; NK cells from all of these patients displayed a peculiar phenotype of an activated memory-like phenotype associated with massive TNF-α production. Discussion: These data suggest that fatal COVID-19 infection is driven by an uncoordinated inflammatory response in part mediated by a specific subset of activated NK cells.


Asunto(s)
COVID-19 , Células Asesinas Naturales , SARS-CoV-2 , COVID-19/inmunología , COVID-19/patología , COVID-19/fisiopatología , Humanos , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Células Asesinas Naturales/patología , SARS-CoV-2/clasificación , SARS-CoV-2/fisiología , Gravedad del Paciente , Resultado Fatal , Vacunas contra la COVID-19 , Masculino , Adulto , Persona de Mediana Edad , Anciano , Anciano de 80 o más Años , Receptores de Células Asesinas Naturales/metabolismo , Factor de Necrosis Tumoral alfa , Activación de Linfocitos
3.
Front Immunol ; 13: 893450, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35911747

RESUMEN

The COVID-19 pandemic has occurred due to infection caused by the SARS-CoV-2 coronavirus, which impacts gestation and pregnancy. In SARS-CoV-2 infection, only very rare cases of vertical transmission have been reported, suggesting that fetal immune imprinting due to a maternal infection is probably a result of changes in maternal immunity. Natural killer (NK) cells are the leading maternal immune cells that act as a natural defense system to fight infections. They also play a pivotal role in the establishment and maintenance of pregnancy. While peripheral NK cells display specific features in patients infected with SARS-CoV-2 in the general population, information remains elusive in pregnant mothers and neonates. In the present study, we analyzed the characteristics of NK cells isolated from both neonatal umbilical cord blood and maternal peripheral blood close to the time of delivery. Phenotype and functions were compared in 18 healthy pregnant women and 34 COVID-19 patients during pregnancy within an ongoing infection (PCR+; N = 15) or after recovery (IgG+PCR-; N = 19). The frequency of NK cells from infected women and their neonates was correlated with the production of inflammatory cytokines in the serum. The expression of NKG2A and NKp30, as well as degranulation of NK cells in pregnant women with ongoing infection, were both negatively correlated to estradiol level. Furthermore, NK cells from the neonates born to infected women were significantly decreased and also correlated to estradiol level. This study highlights the relationship between NK cells, inflammation, and estradiol in patients with ongoing infection, providing new insights into the impact of maternal SARS-CoV-2 infection on the neonate.


Asunto(s)
COVID-19 , Complicaciones Infecciosas del Embarazo , Estradiol , Femenino , Humanos , Células Asesinas Naturales , Pandemias , Parto , Embarazo , SARS-CoV-2
4.
J Immunol ; 207(5): 1333-1343, 2021 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-34408012

RESUMEN

Zika virus (ZIKV) is a mosquito-borne flavivirus that has emerged as a global concern because of its impact on human health. ZIKV infection during pregnancy can cause microcephaly and other severe brain defects in the developing fetus and there have been reports of the occurrence of Guillain-Barré syndrome in areas affected by ZIKV. NK cells are activated during acute viral infections and their activity contributes to a first line of defense because of their ability to rapidly recognize and kill virus-infected cells. To provide insight into NK cell function during ZIKV infection, we have profiled, using mass cytometry, the NK cell receptor-ligand repertoire in a cohort of acute ZIKV-infected female patients. Freshly isolated NK cells from these patients contained distinct, activated, and terminally differentiated, subsets expressing higher levels of CD57, NKG2C, and KIR3DL1 as compared with those from healthy donors. Moreover, KIR3DL1+ NK cells from these patients produced high levels of IFN-γ and TNF-α, in the absence of direct cytotoxicity, in response to in vitro stimulation with autologous, ZIKV-infected, monocyte-derived dendritic cells. In ZIKV-infected patients, overproduction of IFN-γ correlated with STAT-5 activation (r = 0.6643; p = 0.0085) and was mediated following the recognition of MHC class 1-related chain A and chain B molecules expressed by ZIKV-infected monocyte-derived dendritic cells, in synergy with IL-12 production by the latter cells. Together, these findings suggest that NK cells contribute to the generation of an efficacious adaptive anti-ZIKV immune response that could potentially affect the outcome of the disease and/or the development of persistent symptoms.


Asunto(s)
Células Asesinas Naturales/inmunología , Infección por el Virus Zika/inmunología , Virus Zika/fisiología , Enfermedad Aguda , Células Cultivadas , Estudios de Cohortes , Femenino , Humanos , Interferón gamma/metabolismo , Interleucina-12/metabolismo , Activación de Linfocitos , Embarazo , Receptores KIR3DL1/metabolismo , Factor de Transcripción STAT5/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
5.
Am J Transplant ; 21(8): 2846-2863, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33621411

RESUMEN

EBV-positive and EBV-negative posttransplant lymphoproliferative disorders (PTLDs) arise in different immunovirological contexts and might have distinct pathophysiologies. To examine this hypothesis, we conducted a multicentric prospective study with 56 EBV-positive and 39 EBV-negative PTLD patients of the K-VIROGREF cohort, recruited at PTLD diagnosis and before treatment (2013-2019), and compared them to PTLD-free Transplant Controls (TC, n = 21). We measured absolute lymphocyte counts (n = 108), analyzed NK- and T cell phenotypes (n = 49 and 94), and performed EBV-specific functional assays (n = 16 and 42) by multiparameter flow cytometry and ELISpot-IFNγ assays (n = 50). EBV-negative PTLD patients, NK cells overexpressed Tim-3; the 2-year progression-free survival (PFS) was poorer in patients with a CD4 lymphopenia (CD4+ <300 cells/mm3 , p <  .001). EBV-positive PTLD patients presented a profound NK-cell lymphopenia (median = 60 cells/mm3 ) and a high proportion of NK cells expressing PD-1 (vs. TC, p = .029) and apoptosis markers (vs. TC, p < .001). EBV-specific T cells of EBV-positive PTLD patients circulated in low proportions, showed immune exhaustion (p = .013 vs. TC) and poorly recognized the N-terminal portion of EBNA-3A viral protein. Altogether, this broad comparison of EBV-positive and EBV-negative PTLDs highlight distinct patterns of immunopathological mechanisms between these two diseases and provide new clues for immunotherapeutic strategies and PTLD prognosis.


Asunto(s)
Infecciones por Virus de Epstein-Barr , Trastornos Linfoproliferativos , Trasplante de Órganos , Herpesvirus Humano 4 , Humanos , Trastornos Linfoproliferativos/etiología , Trasplante de Órganos/efectos adversos , Estudios Prospectivos
6.
PLoS One ; 14(10): e0224211, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31639143

RESUMEN

Kidney transplant recipients (KTRs) abnormally replicate the Epstein Barr Virus (EBV). To better understand how long-term immunosuppression impacts the immune control of this EBV re-emergence, we systematically compared 10 clinically stable KTRs to 30 healthy controls (HCs). The EBV-specific T cell responses were determined in both groups by multiparameter flow cytometry with intra cellular cytokine staining (KTRs n = 10; HCs n = 15) and ELISpot-IFNγ assays (KTRs n = 7; HCs n = 7). The T/B/NK cell counts (KTRs n = 10; HCs n = 30) and the NK/T cell differentiation and activation phenotypes (KTRs n = 10; HCs n = 15/30) were also measured. We show that in KTRs, the Th1 effector CD4+ T cell responses against latent EBV proteins are weak (2/7 responders). Conversely, the frequencies total EBV-specific CD8+T cells are conserved in KTRs (n = 10) and span a wider range of EBNA-3A peptides (5/7responders) than in HCs (5/7responders). Those modifications of the EBV-specific T cell response were associated with a profound CD4+ T cell lymphopenia in KTRs compared to HCs, involving the naïve CD4+ T cell subset, and a persistent activation of highly-differentiated senescent CD8+ T cells. The proportion of total NK / CD8+ T cells expressing PD-1 was also increased in KTRs. Noteworthy, PD-1 expression on CD8+ T cells normalized with time after transplantation. In conclusion, we show modifications of the EBV-specific cellular immunity in long term transplant recipients. This may be the result of both persistent EBV antigenic stimulation and profound immunosuppression induced by anti-rejection treatments. These findings provide new insights into the immunopathology of EBV infection after renal transplantation.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Infecciones por Virus de Epstein-Barr/inmunología , Herpesvirus Humano 4/inmunología , Trasplante de Riñón/efectos adversos , Linfopenia/etiología , Receptores de Trasplantes/estadística & datos numéricos , Adulto , Anciano , Linfocitos T CD4-Positivos/virología , Linfocitos T CD8-positivos/virología , Infecciones por Virus de Epstein-Barr/epidemiología , Infecciones por Virus de Epstein-Barr/virología , Femenino , Francia/epidemiología , Humanos , Linfopenia/patología , Masculino , Persona de Mediana Edad , Estudios Retrospectivos
7.
EBioMedicine ; 40: 605-613, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30711514

RESUMEN

BACKGROUND: Lassa virus (LASV) is the etiologic agent of an acute hemorrhagic fever endemic in West Africa. Natural killer (NK) cells control viral infections in part through the interaction between killer cell immunoglobulin-like receptors (KIRs) and their ligands. LASV infection is associated with defective immune responses, including inhibition of NK cell activity in the presence of MHC-class 1+-infected target cells. METHODS: We compared individual KIR and HLA-class 1 genotypes of 68 healthy volunteers to 51 patients infected with LASV in Sierra Leone, including 37 survivors and 14 fatalities. Next, potential HLA-C1, HLA-C2, and HLA-Bw4 binding epitopes were in silico screened among LASV nucleoprotein (NP) and envelope glycoprotein (GP). Selected 10-mer peptides were then tested in peptide-HLA stabilization, KIR binding and polyfunction assays. FINDINGS: LASV-infected patients were similar to healthy controls, except for the inhibitory KIR2DL2 gene. We found a specific increase in the HLA-C1:KIR2DL2 interaction in fatalities (10/11) as compared to survivors (12/19) and controls (19/29). We also identified that strong of NP and GP viral epitopes was only observed with HLA-C molecules, and associated with strong inhibition of degranulation in the presence of KIR2DL+ NK cells. This inhibitory effect significantly increased in the presence of the vGP420 variant, detected in 28.1% of LASV sequences. INTERPRETATION: Our finding suggests that presentation of specific LASV epitopes by HLA-C alleles to the inhibitory KIR2DL2 receptor on NK cells could potentially prevent the killing of infected cells and provides insights into the mechanisms by which LASV can escape NK-cell-mediated immune pressure.


Asunto(s)
Epítopos/inmunología , Antígenos HLA-C/inmunología , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Fiebre de Lassa/inmunología , Fiebre de Lassa/metabolismo , Virus Lassa/inmunología , Receptores KIR2DL2/metabolismo , Antígenos Virales/inmunología , Línea Celular , Citotoxicidad Inmunológica , Mapeo Epitopo/métodos , Genotipo , Antígenos HLA-C/genética , Humanos , Tolerancia Inmunológica , Inmunomodulación , Inmunofenotipificación , Fiebre de Lassa/genética , Fiebre de Lassa/virología , Unión Proteica , Receptores KIR2DL2/genética
8.
J Biol Chem ; 290(29): 18009-18017, 2015 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-26060255

RESUMEN

The prevalent human ΔF508 mutation in the cystic fibrosis transmembrane conductance regulator (CFTR) is associated with reduced bone formation and bone loss in mice. The molecular mechanisms by which the ΔF508-CFTR mutation causes alterations in bone formation are poorly known. In this study, we analyzed the osteoblast phenotype in ΔF508-CFTR mice and characterized the signaling mechanisms underlying this phenotype. Ex vivo studies showed that the ΔF508-CFTR mutation negatively impacted the differentiation of bone marrow stromal cells into osteoblasts and the activity of osteoblasts, demonstrating that the ΔF508-CFTR mutation alters both osteoblast differentiation and function. Treatment with a CFTR corrector rescued the abnormal collagen gene expression in ΔF508-CFTR osteoblasts. Mechanistic analysis revealed that NF-κB signaling and transcriptional activity were increased in mutant osteoblasts. Functional studies showed that the activation of NF-κB transcriptional activity in mutant osteoblasts resulted in increased ß-catenin phosphorylation, reduced osteoblast ß-catenin expression, and altered expression of Wnt/ß-catenin target genes. Pharmacological inhibition of NF-κB activity or activation of canonical Wnt signaling rescued Wnt target gene expression and corrected osteoblast differentiation and function in bone marrow stromal cells and osteoblasts from ΔF508-CFTR mice. Overall, the results show that the ΔF508-CFTR mutation impairs osteoblast differentiation and function as a result of overactive NF-κB and reduced Wnt/ß-catenin signaling. Moreover, the data indicate that pharmacological inhibition of NF-κB or activation of Wnt/ß-catenin signaling can rescue the abnormal osteoblast differentiation and function induced by the prevalent ΔF508-CFTR mutation, suggesting novel therapeutic strategies to correct the osteoblast dysfunctions in cystic fibrosis.


Asunto(s)
Regulador de Conductancia de Transmembrana de Fibrosis Quística/inmunología , FN-kappa B/inmunología , Osteoblastos/citología , Vía de Señalización Wnt , Animales , Diferenciación Celular , Células Cultivadas , Masculino , Ratones , Osteoblastos/inmunología , Osteoblastos/patología , beta Catenina/inmunología
9.
J Exp Med ; 212(6): 939-51, 2015 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-26008899

RESUMEN

Inherited, complete deficiency of human HOIL-1, a component of the linear ubiquitination chain assembly complex (LUBAC), underlies autoinflammation, infections, and amylopectinosis. We report the clinical description and molecular analysis of a novel inherited disorder of the human LUBAC complex. A patient with multiorgan autoinflammation, combined immunodeficiency, subclinical amylopectinosis, and systemic lymphangiectasia, is homozygous for a mutation in HOIP, the gene encoding the catalytic component of LUBAC. The missense allele (L72P, in the PUB domain) is at least severely hypomorphic, as it impairs HOIP expression and destabilizes the whole LUBAC complex. Linear ubiquitination and NF-κB activation are impaired in the patient's fibroblasts stimulated by IL-1ß or TNF. In contrast, the patient's monocytes respond to IL-1ß more vigorously than control monocytes. However, the activation and differentiation of the patient's B cells are impaired in response to CD40 engagement. These cellular and clinical phenotypes largely overlap those of HOIL-1-deficient patients. Clinical differences between HOIL-1- and HOIP-mutated patients may result from differences between the mutations, the loci, or other factors. Our findings show that human HOIP is essential for the assembly and function of LUBAC and for various processes governing inflammation and immunity in both hematopoietic and nonhematopoietic cells.


Asunto(s)
Regulación de la Expresión Génica , Ubiquitina-Proteína Ligasas/deficiencia , Alelos , Secuencia de Aminoácidos , Ligando de CD40/metabolismo , Catálisis , Femenino , Fibroblastos/metabolismo , Prueba de Complementación Genética , Mutación de Línea Germinal , Enfermedad del Almacenamiento de Glucógeno Tipo IV/patología , Homocigoto , Humanos , Síndromes de Inmunodeficiencia/patología , Inflamación , Linfangiectasia/patología , Monocitos/metabolismo , Mutación Missense , FN-kappa B/metabolismo , Homología de Secuencia de Aminoácido , Factores de Transcripción , Ubiquitina/química , Ubiquitina-Proteína Ligasas/genética , Adulto Joven
10.
J Cell Biol ; 204(2): 231-45, 2014 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-24446482

RESUMEN

Nuclear factor κB (NF-κB) essential modulator (NEMO), a regulatory component of the IκB kinase (IKK) complex, controls NF-κB activation through its interaction with ubiquitin chains. We show here that stimulation with interleukin-1 (IL-1) and TNF induces a rapid and transient recruitment of NEMO into punctate structures that are anchored at the cell periphery. These structures are enriched in activated IKK kinases and ubiquitinated NEMO molecules, which suggests that they serve as organizing centers for the activation of NF-κB. These NEMO-containing structures colocalize with activated TNF receptors but not with activated IL-1 receptors. We investigated the involvement of nondegradative ubiquitination in the formation of these structures, using cells deficient in K63 ubiquitin chains or linear ubiquitin chain assembly complex (LUBAC)-mediated linear ubiquitination. Our results indicate that, unlike TNF, IL-1 requires K63-linked and linear ubiquitin chains to recruit NEMO into higher-order complexes. Thus, different mechanisms are involved in the recruitment of NEMO into supramolecular complexes, which appear to be essential for NF-κB activation.


Asunto(s)
Quinasa I-kappa B/metabolismo , Interleucina-1/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Línea Celular Tumoral , Células HeLa , Humanos , Quinasa I-kappa B/análisis , Interleucina-1/análisis , Interleucina-1/fisiología , Quinasas Asociadas a Receptores de Interleucina-1/análisis , Quinasas Asociadas a Receptores de Interleucina-1/metabolismo , FN-kappa B/análisis , FN-kappa B/metabolismo , Receptores de Interleucina-1/análisis , Receptores de Interleucina-1/metabolismo , Receptores del Factor de Necrosis Tumoral/análisis , Receptores del Factor de Necrosis Tumoral/metabolismo , Transducción de Señal , Factor de Necrosis Tumoral alfa/análisis , Factor de Necrosis Tumoral alfa/fisiología , Ubiquitina/genética , Ubiquitina/metabolismo , Ubiquitina/fisiología , Ubiquitinación
11.
FASEB J ; 28(2): 603-14, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24145721

RESUMEN

The Notch signaling pathway is involved in liver development and regeneration. Here, we investigate the role of the 4 mammalian Notch paralogs in the regulation of hepatoblast proliferation and hepatocytic differentiation. Our model is based on bipotential mouse embryonic liver (BMEL) progenitors that can differentiate into hepatocytes or cholangiocytes in vitro and in vivo. BMEL cells were subjected to Notch antagonists or agonists. Blocking Notch activation with a γ-secretase inhibitor, at 50 µM for 48 h, reduced cell growth by 50%. S-phase entry was impaired, but no apoptosis was induced. A systematic paralog-specific strategy was set using lentiviral transduction with constitutively active forms of each Notch receptor along with inhibition of endogenous Notch signaling. This assay demonstrates that proliferation of BMEL cells requires Notch2 and Notch4 activity, resulting in significant down-regulation of p27(Kip1) and p57(Kip2) cyclin-dependent kinase inhibitors. Conversely, Notch3-expressing cells proliferate less and express 3-fold higher levels of p57(Kip2). The Notch3 cells present a hepatocyte-like morphology, enhanced multinucleation, and a ploidy shift. Moreover, Notch3 activity is conducive to hepatocytic differentiation in vitro, while its paralogs impede this fate. Our study provides the first evidence of a functional diversity among the mammalian Notch homologues in the proliferation and hepatocytic-lineage commitment of liver progenitors.


Asunto(s)
Hepatocitos/citología , Hepatocitos/metabolismo , Hígado/citología , Hígado/metabolismo , Receptores Notch/metabolismo , Células Madre/citología , Células Madre/metabolismo , Animales , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Línea Celular , Proliferación Celular , Técnica del Anticuerpo Fluorescente , Ratones , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores Notch/genética
12.
J Immunol ; 171(10): 5305-12, 2003 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-14607932

RESUMEN

We identified two novel isoforms of the human chemokine receptor CX3CR1, produced by alternative splicing and with N-terminal regions extended by 7 and 32 aa. Expression of the messengers coding these isoforms, compared with that of previously described V28 messengers, is lower in monocytes and NK cells, but higher in CD4(+) T lymphocytes. CX3CR1 and its extended isoforms were expressed in HEK-293 cells and compared for expression, ligand binding, and cellular responses. In steady state experiments, all three CX3CR1 isoforms bound CX3CL1 with similar affinity. In kinetic binding studies, however, k(on) and k(off) were significantly greater for the extended CX3CR1 isoforms, thereby suggesting that the N-terminal extensions may alter the functions induced by CX3CL1. In signaling studies, all three CX3CR1 isoforms mediated agonist-dependent calcium mobilization, but the EC(50) was lower for the extended than for the standard isoforms. In addition, chemotactic responses for these extended isoforms shifted left, also indicating a more sensitive response. Finally, the longer variants appeared to be more potent HIV coreceptors when tested in fusion and infection assays. In conclusion, we identified and characterized functionally two novel isoforms of CX3CR1 that respond more sensitively to CX3CL1 and HIV viral envelopes. These data reveal new complexity in CX3CR1 cell activation and confirm the critical role of the N-terminal domain of the chemokine receptors in ligand recognition and cellular response.


Asunto(s)
Quimiocinas CX3C/metabolismo , Proteínas de la Membrana/metabolismo , Receptores del VIH/fisiología , Empalme Alternativo/inmunología , Secuencia de Aminoácidos , Secuencia de Bases , Receptor 1 de Quimiocinas CX3C , Línea Celular , Células Cultivadas , Quimiocina CX3CL1 , Quimiocina CXCL1 , Quimiocinas CX3C/biosíntesis , Quimiocinas CX3C/genética , Quimiocinas CX3C/aislamiento & purificación , Quimiocinas CXC/agonistas , Quimiocinas CXC/metabolismo , Quimiotaxis de Leucocito/genética , Quimiotaxis de Leucocito/inmunología , Regulación de la Expresión Génica/inmunología , Humanos , Péptidos y Proteínas de Señalización Intercelular/agonistas , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Proteínas de la Membrana/biosíntesis , Proteínas de la Membrana/genética , Proteínas de la Membrana/aislamiento & purificación , Datos de Secuencia Molecular , Unión Proteica/genética , Unión Proteica/inmunología , Isoformas de Proteínas/biosíntesis , Isoformas de Proteínas/genética , Isoformas de Proteínas/aislamiento & purificación , Isoformas de Proteínas/metabolismo , ARN Mensajero/biosíntesis , Receptores de Quimiocina/agonistas , Receptores de Quimiocina/biosíntesis , Receptores de Quimiocina/genética , Receptores de Quimiocina/metabolismo , Transfección
13.
Biochem Biophys Res Commun ; 308(4): 744-9, 2003 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-12927781

RESUMEN

P100, which is encoded by NF-kappa B2, inhibits Rel dimers. It can also be processed into p52, one of the DNA binding sub-units of NF-kappa B/Rel factors. Several p100 C-terminal truncations that result from gene rearrangements are associated with lymphomagenesis. Here, we characterized a new p100 mutant that we termed p100HB. It originates from a point-mutation that generates a premature stop-codon, and thus the protein lacks the last 125 amino acids. We have detected p100HB in several human tumor cell lines. The truncated protein is mainly unprocessed, and although it still binds Rel dimers, it has reduced inhibitory potency compared to p100 and translocates into the nucleus. Thus, p100HB may be associated with deregulated NF-kappa B/Rel functions.


Asunto(s)
Mutación , FN-kappa B/química , FN-kappa B/genética , Transporte Activo de Núcleo Celular , Proteínas Adaptadoras Transductoras de Señales , Secuencia de Bases , Western Blotting , Proteínas Portadoras/química , Codón , Dimerización , Exones , Regulación de la Expresión Génica , Humanos , Proteínas I-kappa B/metabolismo , Células Jurkat , Linfocitos/metabolismo , Microscopía Confocal , Datos de Secuencia Molecular , Subunidad p52 de NF-kappa B , Plásmidos/metabolismo , Mutación Puntual , Pruebas de Precipitina , Estructura Terciaria de Proteína , Transporte de Proteínas , ARN Mensajero/metabolismo , Factores de Tiempo , Transfección , Células Tumorales Cultivadas
14.
Cytokine ; 21(6): 303-11, 2003 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-12824004

RESUMEN

Expression of membrane-bound CX3CL1, a CX(3)C chemokine, can be strongly induced by inflammatory cytokines in primary endothelial cells, mediating capture and tight adhesion of cells, such as monocytes, that carry the CX(3)CR1 receptor. Here, we measured CX3CL1 mRNA and protein induction by human aortic smooth muscle cells (SMCs), another major component of vessel walls, in response to inflammatory stimuli, and analyzed the effect of membrane-bound CX3CL1 on monocyte adhesion, tissue factor (TF) expression, and tumor necrosis factor-alpha (TNF-alpha) released. In human vascular SMCs, CX3CL1 transcripts were induced after 4h of stimulation with a combination of TNF-alpha and interferon-gamma. Cell-associated and shedded CX3CL1 were measured with a specific ELISA, showing that only 30% of the protein was cleaved from the membrane. Expression of CX3CL1 by SMC increased adhesion of monocytic cells, an effect, which was blocked by soluble CX3CL1. Interestingly, monocyte adhesion to CX3CL1-coated plates partially inhibited lipopolysaccharide-induced TF expression and TNF-alpha release. Thus, CX3CL1, in addition to its adhesive/chemotactic functions, directly promotes monocyte antiinflammatory and antiprocoagulant responses. This could have important implications in clinical settings such as atherosclerosis, in which SMCs and monocytic cells are in close proximity.


Asunto(s)
Factores de Coagulación Sanguínea/metabolismo , Quimiocinas CX3C/metabolismo , Inflamación/metabolismo , Proteínas de la Membrana/metabolismo , Monocitos/metabolismo , Músculo Liso Vascular/metabolismo , Aorta/citología , Adhesión Celular/fisiología , Comunicación Celular/inmunología , Células Cultivadas , Quimiocina CX3CL1 , Quimiocinas CX3C/genética , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo/métodos , Humanos , Interferón gamma/metabolismo , Interferón gamma/farmacología , Lipopolisacáridos , Proteínas de la Membrana/genética , Monocitos/inmunología , Músculo Liso Vascular/citología , ARN Mensajero/metabolismo , Tromboplastina/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Factor de Necrosis Tumoral alfa/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA