Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
J Leukoc Biol ; 112(3): 449-456, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35137444

RESUMEN

Few cytokines/growth modulating proteins are known to be chemoattractants for hematopoietic stem (HSC) and progenitor cells (HPC); stromal cell-derived factor 1α (SDF1α/CXCL12) being the most potent known such protein. DEK, a nuclear DNA-binding chromatin protein with hematopoietic cytokine-like activity, is a chemotactic factor attracting mature immune cells. Transwell migration assays were performed to test whether DEK serves as a chemotactic agent for HSC/HPC. DEK induced dose- and time-dependent directed migration of lineage negative (Lin- ) Sca-1+ c-Kit+ (LSK) bone marrow (BM) cells, HSCs and HPCs. Checkerboard assays demonstrated that DEK's activity was chemotactic (directed), not chemokinetic (random migration), in nature. DEK and SDF1α compete for HSC/HPC chemotaxis. Blocking CXCR2 with neutralizing antibodies or inhibiting Gαi protein signaling with Pertussis toxin pretreatment inhibited migration of LSK cells toward DEK. Thus, DEK is a novel and rare chemotactic agent for HSC/HPC acting in a direct or indirect CXCR2 and Gαi protein-coupled signaling-dependent manner.


Asunto(s)
Células Madre Hematopoyéticas , Proteínas Nucleares , Células de la Médula Ósea/metabolismo , Quimiotaxis , Proteínas de Unión al ADN/metabolismo , Células Madre Hematopoyéticas/metabolismo , Proteínas Nucleares/metabolismo , Receptores de Interleucina-8B/metabolismo
3.
J Clin Invest ; 129(6): 2555-2570, 2019 05 20.
Artículo en Inglés | MEDLINE | ID: mdl-31107242

RESUMEN

The nuclear protein DEK is an endogenous DNA-binding chromatin factor regulating hematopoiesis. DEK is one of only 2 known secreted nuclear chromatin factors, but whether and how extracellular DEK regulates hematopoiesis is not known. We demonstrated that extracellular DEK greatly enhanced ex vivo expansion of cytokine-stimulated human and mouse hematopoietic stem cells (HSCs) and regulated HSC and hematopoietic progenitor cell (HPC) numbers in vivo and in vitro as determined both phenotypically (by flow cytometry) and functionally (through transplantation and colony formation assays). Recombinant DEK increased long-term HSC numbers and decreased HPC numbers through a mechanism mediated by the CXC chemokine receptor CXCR2 and heparan sulfate proteoglycans (HSPGs) (as determined utilizing Cxcr2-/- mice, blocking CXCR2 antibodies, and 3 different HSPG inhibitors) that was associated with enhanced phosphorylation of ERK1/2, AKT, and p38 MAPK. To determine whether extracellular DEK required nuclear function to regulate hematopoiesis, we utilized 2 mutant forms of DEK: one that lacked its nuclear translocation signal and one that lacked DNA-binding ability. Both altered HSC and HPC numbers in vivo or in vitro, suggesting the nuclear function of DEK is not required. Thus, DEK acts as a hematopoietic cytokine, with the potential for clinical applicability.


Asunto(s)
Proteínas Cromosómicas no Histona/metabolismo , Citocinas/metabolismo , Proteínas de Unión al ADN/metabolismo , Hematopoyesis , Células Madre Hematopoyéticas/metabolismo , Sistema de Señalización de MAP Quinasas , Proteínas Oncogénicas/metabolismo , Proteínas de Unión a Poli-ADP-Ribosa/metabolismo , Animales , Proteínas Cromosómicas no Histona/genética , Citocinas/genética , Proteínas de Unión al ADN/genética , Quinasas MAP Reguladas por Señal Extracelular/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Ratones , Ratones Noqueados , Mutación , Proteínas Oncogénicas/genética , Proteínas de Unión a Poli-ADP-Ribosa/genética , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas Sprague-Dawley , Receptores de Interleucina-8B
4.
Front Immunol ; 10: 2957, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31998291

RESUMEN

Sepsis results in millions of deaths every year, with acute lung injury (ALI) being one of the leading causes of mortality in septic patients. As neutrophil extracellular traps (NETs) are abundant in sepsis, neutralizing components of NETs may be a useful strategy to improve outcomes of sepsis. Citrullinated histone H3 (CitH3) has been recently shown to be involved in the NET formation. In this study, we demonstrate that CitH3 damages human umbilical vein endothelial cells (HUVECs) and potentiates NET formation through a positive feedback mechanism. We developed a novel CitH3 monoclonal antibody to target peptidylarginine deiminase (PAD) 2 and PAD 4 generated CitH3. In a mouse model of lethal lipopolysaccharide (LPS) induced shock, neutralizing CitH3 with the newly developed anti-CitH3 monoclonal antibody attenuates inflammatory responses, ameliorates ALI, and improves survival. Our study suggests that effectively blocking circulating CitH3 might be a potential therapeutic method for the treatment of endotoxemia.


Asunto(s)
Histonas/inmunología , Choque Séptico/inmunología , Animales , Anticuerpos Monoclonales/administración & dosificación , Citrulinación , Modelos Animales de Enfermedad , Trampas Extracelulares/inmunología , Histonas/genética , Humanos , Masculino , Ratones , Neutrófilos/inmunología , Arginina Deiminasa Proteína-Tipo 2/inmunología , Arginina Deiminasa Proteína-Tipo 4/inmunología , Choque Séptico/tratamiento farmacológico , Choque Séptico/genética
5.
mBio ; 9(1)2018 02 27.
Artículo en Inglés | MEDLINE | ID: mdl-29487235

RESUMEN

Listeria monocytogenes is a facultative intracellular bacterial pathogen that is frequently associated with food-borne infection. Of particular concern is the ability of L. monocytogenes to breach the blood-brain barrier, leading to life-threatening meningitis and encephalitis. The mechanisms used by bacterial pathogens to infect the brain are not fully understood. Here we show that L. monocytogenes is able to utilize vimentin for invasion of host cells. Vimentin is a type III intermediate filament protein within the cytosol but is also expressed on the host cell surface. We found that L. monocytogenes interaction with surface-localized vimentin promoted bacterial uptake. Furthermore, in the absence of vimentin, L. monocytogenes colonization of the brain was severely compromised in mice. The L. monocytogenes virulence factor InlF was found to bind vimentin and was necessary for optimal bacterial colonization of the brain. These studies reveal a novel receptor-ligand interaction that enhances infection of the brain by L. monocytogenes and highlights the importance of surface vimentin in host-pathogen interactions.IMPORTANCEListeria monocytogenes is an intracellular bacterial pathogen that is capable of invading numerous host cells during infection. L. monocytogenes can cross the blood-brain barrier, leading to life-threatening meningitis. Here we show that an L. monocytogenes surface protein, InlF, is necessary for optimal colonization of the brain in mice. Furthermore, in the absence of vimentin, a cytosolic intermediate filament protein that is also present on the surface of brain endothelial cells, colonization of the brain was significantly impaired. We further show that InlF binds vimentin to mediate invasion of host cells. This work identifies InlF as a bacterial surface protein with specific relevance for infection of the brain and underscores the significance of host cell surface vimentin interactions in microbial pathogenesis.


Asunto(s)
Encéfalo/parasitología , Endocitosis , Interacciones Huésped-Patógeno , Listeria monocytogenes/fisiología , Listeriosis/parasitología , Vimentina/metabolismo , Animales , Encéfalo/patología , Línea Celular , Modelos Animales de Enfermedad , Listeriosis/patología , Ratones , Ratas
6.
Arthritis Rheumatol ; 70(4): 594-605, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29287303

RESUMEN

OBJECTIVE: The nuclear oncoprotein DEK is an autoantigen associated with juvenile idiopathic arthritis (JIA), especially the oligoarticular subtype. DEK is a secreted chemotactic factor. Abundant levels of DEK and DEK autoantibodies are found in inflamed synovium in JIA. We undertook this study to further characterize the nature of DEK autoantibodies in screening serum samples from 2 different cohorts that consisted mostly of patients with JIA. METHODS: DEK autoantibody levels were analyzed in sera from 33 JIA patients, 13 patients with other inflammatory conditions, and 11 healthy controls, as well as in 89 serum samples from JIA patients receiving anti-tumor necrosis factor (anti-TNF) therapy. Recombinant His-tagged full-length DEK protein (1-375 amino acids [aa]) and the 187-375-aa and 1-350-aa His-tagged DEK fragments made in a baculovirus system were used for enzyme-linked immunosorbent assay (ELISA) and immunoblotting. The C-terminal 25-aa fragment of DEK was expressed in a glutathione S-transferase-tagged vector. ELISA results were calculated as area under the curve by the trapezoidal rule. RESULTS: DEK autoantibody levels were significantly higher in patients with polyarticular JIA than in those with oligoarticular JIA, and were higher in patients with polyarticular JIA who had more active disease after cessation of anti-TNF therapy. Immunoblotting against the C-terminal 25-aa fragment of DEK confirmed that this section of the DEK molecule is the most immunogenic domain. CONCLUSION: DEK autoantibody levels are higher in patients with polyarticular JIA than in those with oligoarticular JIA, and higher in patients who have disease flares after cessation of anti-TNF therapy. The C-terminal 25-aa fragment is the most immunogenic portion of DEK. These findings are significant with respect to the nature of DEK autoantibodies, their contribution to JIA pathogenesis, and their implications for JIA management.


Asunto(s)
Antirreumáticos/inmunología , Artritis Juvenil/sangre , Autoanticuerpos/sangre , Proteínas Cromosómicas no Histona/inmunología , Proteínas Oncogénicas/inmunología , Proteínas de Unión a Poli-ADP-Ribosa/inmunología , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Adolescente , Artritis Juvenil/tratamiento farmacológico , Artritis Juvenil/inmunología , Autoanticuerpos/inmunología , Estudios de Casos y Controles , Niño , Femenino , Humanos , Masculino , Brote de los Síntomas , Privación de Tratamiento
7.
Nat Commun ; 8: 14252, 2017 02 06.
Artículo en Inglés | MEDLINE | ID: mdl-28165452

RESUMEN

Novel therapeutics are required for improving the management of chronic inflammatory diseases. Aptamers are single-stranded RNA or DNA molecules that have recently shown utility in a clinical setting, as they can specifically neutralize biomedically relevant proteins, particularly cell surface and extracellular proteins. The nuclear chromatin protein DEK is a secreted chemoattractant that is abundant in the synovia of patients with juvenile idiopathic arthritis (JIA). Here, we show that DEK is crucial to the development of arthritis in mouse models, thus making it an appropriate target for aptamer-based therapy. Genetic depletion of DEK or treatment with DEK-targeted aptamers significantly reduces joint inflammation in vivo and greatly impairs the ability of neutrophils to form neutrophil extracellular traps (NETs). DEK is detected in spontaneously forming NETs from JIA patient synovial neutrophils, and DEK-targeted aptamers reduce NET formation. DEK is thus key to joint inflammation, and anti-DEK aptamers hold promise for the treatment of JIA and other types of arthritis.


Asunto(s)
Aptámeros de Nucleótidos/uso terapéutico , Artritis Juvenil/terapia , Factores Quimiotácticos/antagonistas & inhibidores , Proteínas Cromosómicas no Histona/antagonistas & inhibidores , Proteínas de Unión al ADN/genética , Trampas Extracelulares/inmunología , Proteínas Oncogénicas/antagonistas & inhibidores , Proteínas Oncogénicas/genética , Proteínas de Unión a Poli-ADP-Ribosa/antagonistas & inhibidores , Proteínas de Unión a Poli-ADP-Ribosa/genética , Adulto , Animales , Artritis Juvenil/inmunología , Factores Quimiotácticos/genética , Factores Quimiotácticos/inmunología , Factores Quimiotácticos/metabolismo , Proteínas Cromosómicas no Histona/genética , Proteínas Cromosómicas no Histona/inmunología , Proteínas Cromosómicas no Histona/metabolismo , Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/metabolismo , Modelos Animales de Enfermedad , Trampas Extracelulares/metabolismo , Femenino , Voluntarios Sanos , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neutrófilos/inmunología , Proteínas Oncogénicas/inmunología , Proteínas Oncogénicas/metabolismo , Proteínas de Unión a Poli-ADP-Ribosa/inmunología , Proteínas de Unión a Poli-ADP-Ribosa/metabolismo , Cultivo Primario de Células , Líquido Sinovial/química , Líquido Sinovial/citología , Líquido Sinovial/inmunología , Zimosan/inmunología
8.
PLoS One ; 11(9): e0162641, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27657497

RESUMEN

BACKGROUND: NF-κB activation, pathogen invasion, polymorphonuclear leukocytes (PMN) transmigration (PMNT) across the blood-brain barrier (BBB) are the pathogenic triad hallmark features of bacterial meningitis, but the mechanisms underlying these events remain largely unknown. Vimentin, which is a novel NF-κB regulator, is the primary receptor for the major Escherichia coli K1 virulence factor IbeA that contributes to the pathogenesis of neonatal bacterial sepsis and meningitis (NSM). We have previously shown that IbeA-induced NF-κB signaling through its primary receptor vimentin as well as its co-receptor PTB-associated splicing factor (PSF) is required for pathogen penetration and leukocyte transmigration across the BBB. This is the first in vivo study to demonstrate how vimentin and related factors contributed to the pathogenic triad of bacterial meningitis. METHODOLOGY/PRINCIPAL FINDINGS: The role of vimentin in IbeA+ E. coli K1-induced NF-κB activation, pathogen invasion, leukocyte transmigration across the BBB has now been demonstrated by using vimentin knockout (KO) mice. In the in vivo studies presented here, IbeA-induced NF-κB activation, E. coli K1 invasion and polymorphonuclear neutrophil (PMN) transmigration across the BBB were significantly reduced in Vim-/- mice. Decreased neuronal injury in the hippocampal dentate gyrus was observed in Vim-/- mice with meningitis. The major inflammatory regulator α7 nAChR and several signaling molecules contributing to NF-κB activation (p65 and p-CamKII) were significantly reduced in the brain tissues of the Vim-/- mice with E. coli meningitis. Furthermore, Vim KO resulted in significant reduction in neuronal injury and in α7 nAChR-mediated calcium signaling. CONCLUSION/SIGNIFICANCE: Vimentin, a novel NF-κB regulator, plays a detrimental role in the host defense against meningitic infection by modulating the NF-κB signaling pathway to increase pathogen invasion, PMN recruitment, BBB permeability and neuronal inflammation. Our findings provide the first evidence for Vim-dependent mechanisms underlying the pathogenic triad of bacterial meningitis.

9.
Nat Microbiol ; 1: 16025, 2016 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-27572444

RESUMEN

Type 3 secretion systems (T3SSs) of bacterial pathogens translocate bacterial effector proteins that mediate disease into the eukaryotic cytosol. Effectors traverse the plasma membrane through a translocon pore formed by T3SS proteins. In a genome-wide selection, we identified the intermediate filament vimentin as required for infection by the T3SS-dependent pathogen S. flexneri. We found that vimentin is required for efficient T3SS translocation of effectors by S. flexneri and other pathogens that use T3SS, Salmonella enterica serovar Typhimurium and Yersinia pseudotuberculosis. Vimentin and the intestinal epithelial intermediate filament keratin 18 interact with the C-terminus of the Shigella translocon pore protein IpaC. Vimentin and its interaction with IpaC are dispensable for pore formation, but are required for stable docking of S. flexneri to cells; moreover, stable docking triggers effector secretion. These findings establish that stable docking of the bacterium specifically requires intermediate filaments, is a process distinct from pore formation, and is a prerequisite for effector secretion.


Asunto(s)
Adhesión Bacteriana , Salmonella typhimurium/fisiología , Shigella flexneri/fisiología , Sistemas de Secreción Tipo III/metabolismo , Vimentina/metabolismo , Factores de Virulencia/metabolismo , Yersinia pseudotuberculosis/fisiología , Animales , Antígenos Bacterianos/metabolismo , Línea Celular , Interacciones Huésped-Patógeno , Humanos , Queratina-18/metabolismo , Ratones , Unión Proteica , Transporte de Proteínas
10.
Blood Cells Mol Dis ; 54(1): 123-31, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25128083

RESUMEN

DEK is important in regulating cellular processes including proliferation, differentiation and maintenance of stem cell phenotype. The translocation t(6;9) in Acute Myeloid Leukemia (AML), which fuses DEK with NUP214, confers a poor prognosis and a higher risk of relapse. The over-expression of DEK in AML has been reported, but different studies have shown diminished levels in pediatric and promyelocytic leukemias. This study has characterized DEK expression, in silico, using a large multi-center cohort of leukemic and normal control cases. Overall, DEK was under-expressed in AML compared to normal bone marrow (NBM). Studying specific subtypes of AML confirmed either no significant change or a significant reduction in DEK expression compared to NBM. Importantly, the similarity of DEK expression between AML and NBM was confirmed using immunohistochemistry analysis of tissue mircorarrays. In addition, stratification of AML patients based on median DEK expression levels indicated that DEK showed no effect on the overall survival of patients. DEK expression during normal hematopoiesis did reveal a relationship with specific cell types implicating a distinct function during myeloid differentiation. Whilst DEK may play a potential role in hematopoiesis, it remains to be established whether it is important for leukemagenesis, except when involved in the t(6;9) translocation.


Asunto(s)
Proteínas Cromosómicas no Histona/biosíntesis , Proteínas de Unión al ADN/biosíntesis , Bases de Datos Genéticas , Regulación Leucémica de la Expresión Génica , Hematopoyesis , Leucemia Mieloide Aguda/metabolismo , Proteínas Oncogénicas/biosíntesis , Animales , Proteínas Cromosómicas no Histona/genética , Cromosomas Humanos Par 6/genética , Cromosomas Humanos Par 9/genética , Estudios de Cohortes , Proteínas de Unión al ADN/genética , Supervivencia sin Enfermedad , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patología , Estudios Multicéntricos como Asunto , Proteínas Oncogénicas/genética , Proteínas de Unión a Poli-ADP-Ribosa , Tasa de Supervivencia , Translocación Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...