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

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 110(17): 6847-52, 2013 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-23569252

RESUMEN

DEK is a biochemically distinct, conserved nonhistone protein that is vital to global heterochromatin integrity. In addition, DEK can be secreted and function as a chemotactic, proinflammatory factor. Here we show that exogenous DEK can penetrate cells, translocate to the nucleus, and there carry out its endogenous nuclear functions. Strikingly, adjacent cells can take up DEK secreted from synovial macrophages. DEK internalization is a heparan sulfate-dependent process, and cellular uptake of DEK into DEK knockdown cells corrects global heterochromatin depletion and DNA repair deficits, the phenotypic aberrations characteristic of these cells. These findings thus unify the extracellular and intracellular activities of DEK, and suggest that this paracrine loop involving DEK plays a role in chromatin biology.


Asunto(s)
Proteínas Cromosómicas no Histona/metabolismo , Reparación del ADN/fisiología , Heterocromatina/metabolismo , Histonas/metabolismo , Proteínas Oncogénicas/metabolismo , Comunicación Paracrina/fisiología , Fraccionamiento Celular , Células HeLa , Humanos , Procesamiento de Imagen Asistido por Computador , Immunoblotting , Inmunoprecipitación , Microscopía Fluorescente , Proteínas de Unión a Poli-ADP-Ribosa , Transporte de Proteínas/fisiología , ARN Interferente Pequeño/genética
2.
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
4.
Stem Cells ; 31(8): 1447-53, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23733396

RESUMEN

Understanding the factors that regulate hematopoiesis opens up the possibility of modifying these factors and their actions for clinical benefit. DEK, a non-histone nuclear phosphoprotein initially identified as a putative proto-oncogene, has recently been linked to regulate hematopoiesis. DEK has myelosuppressive activity in vitro on proliferation of human and mouse hematopoietic progenitor cells and enhancing activity on engraftment of long-term marrow repopulating mouse stem cells, has been linked in coordinate regulation with the transcription factor C/EBPα, for differentiation of myeloid cells, and apparently targets a long-term repopulating hematopoietic stem cell for leukemic transformation. This review covers the uniqueness of DEK, what is known about how it now functions as a nuclear protein and also as a secreted molecule that can act in paracrine fashion, and how it may be regulated in part by dipeptidylpeptidase 4, an enzyme known to truncate and modify a number of proteins involved in activities on hematopoietic cells. Examples are provided of possible future areas of investigation needed to better understand how DEK may be regulated and function as a regulator of hematopoiesis, information possibly translatable to other normal and diseased immature cell systems.


Asunto(s)
Proteínas Cromosómicas no Histona/fisiología , Proteínas de Unión al ADN/fisiología , Hematopoyesis/fisiología , Células Madre Hematopoyéticas/fisiología , Proteínas Oncogénicas/fisiología , Animales , Humanos , Proteínas de Unión a Poli-ADP-Ribosa , Proto-Oncogenes Mas
5.
Arthritis Rheum ; 63(2): 556-67, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21280010

RESUMEN

OBJECTIVE: DEK is a nuclear phosphoprotein and autoantigen in a subset of children with juvenile idiopathic arthritis (JIA). Autoantibodies to DEK are also found in a broad spectrum of disorders associated with abnormal immune activation. We previously demonstrated that DEK is secreted by macrophages, is released by apoptotic T cells, and attracts leukocytes. Since DEK has been identified in the synovial fluid (SF) of patients with JIA, this study was undertaken to investigate how DEK protein and/or autoantibodies may contribute to the pathogenesis of JIA. METHODS: DEK autoantibodies, immune complexes (ICs), and synovial macrophages were purified from the SF of patients with JIA. DEK autoantibodies and ICs were purified by affinity-column chromatography and analyzed by 2-dimensional gel electrophoresis, immunoblotting, and enzyme-linked immunosorbent assay. DEK in supernatants and exosomes was purified by serial centrifugation and immunoprecipitation with magnetic beads, and posttranslational modifications of DEK were identified by nano-liquid chromatography tandem mass spectrometry (nano-LC-MS/MS). RESULTS: DEK autoantibodies and protein were found in the SF of patients with JIA. Secretion of DEK by synovial macrophages was observed both in a free form and via exosomes. DEK autoantibodies (IgG2) may activate the complement cascade, primarily recognize the C-terminal portion of DEK protein, and exhibit higher affinity for acetylated DEK. Consistent with these observations, DEK underwent acetylation on an unprecedented number of lysine residues, as demonstrated by nano-LC-MS/MS. CONCLUSION: These results indicate that DEK can contribute directly to joint inflammation in JIA by generating ICs through high-affinity interaction between DEK and DEK autoantibodies, a process enhanced by acetylation of DEK in the inflamed joint.


Asunto(s)
Artritis Juvenil/metabolismo , Autoantígenos/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Proteínas Oncogénicas/metabolismo , Procesamiento Proteico-Postraduccional , Membrana Sinovial/metabolismo , Acetilación , Complejo Antígeno-Anticuerpo/inmunología , Complejo Antígeno-Anticuerpo/metabolismo , Artritis Juvenil/inmunología , Artritis Juvenil/patología , Autoanticuerpos/sangre , Autoantígenos/inmunología , Niño , Proteínas Cromosómicas no Histona/inmunología , Humanos , Articulaciones/metabolismo , Articulaciones/patología , Macrófagos/metabolismo , Macrófagos/patología , Proteínas Oncogénicas/inmunología , Proteínas de Unión a Poli-ADP-Ribosa , Líquido Sinovial/química , Líquido Sinovial/metabolismo , Membrana Sinovial/patología
6.
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
7.
Nat Cell Biol ; 5(1): 59-63, 2003 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-12483219

RESUMEN

Vimentin is a widely expressed intermediate filament protein thought to be involved mainly in structural processes, such as wound healing. We now demonstrate that activated human macrophages secrete vimentin into the extracellular space. The maturation of blood-derived monocytes into macrophages involves several signalling pathways. We show that secretion of vimentin, which is phosphorylated at serine and threonine residues, is enhanced by the phosphatase inhibitor okadaic acid and blocked by the specific protein kinase C inhibitor GO6983. These findings are consistent with previous observations that phosphorylation of vimentin affects its intracellular localization and that vimentin is a substrate for protein kinase C (PKC). We also show that the anti-inflammatory cytokine interleukin-10 (IL-10), which inhibits PKC activity, blocks secretion of vimentin. In contrast, the pro-inflammatory cytokine tumour necrosis factor alpha (TNF-alpha) can trigger secretion of vimentin. Finally, we found that extracellular vimentin is involved in bacterial killing and the generation of oxidative metabolites, two important functions of activated macrophages. These data establish that vimentin is secreted by macrophages in response to pro-inflammatory signalling pathways and is probably involved in immune function.


Asunto(s)
Activación de Macrófagos/fisiología , Macrófagos/metabolismo , Vimentina/metabolismo , Células Cultivadas , Inhibidores Enzimáticos/farmacología , Humanos , Inmunohistoquímica , Interleucina-10/farmacología , Cinética , Macrófagos/citología , Macrófagos/efectos de los fármacos , Monocitos/citología , Ácido Ocadaico/farmacología , Fosforilación , Proteína Quinasa C/antagonistas & inhibidores , Transducción de Señal , Vimentina/antagonistas & inhibidores
8.
Prostate ; 70(4): 377-89, 2010 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-19866464

RESUMEN

BACKGROUND: Histological evidence of pervasive inflammatory infiltrate has been noted in both benign prostatic hyperplasia/hypertrophy (BPH) and prostate cancer (PCa). Cytokines known to attract particular leukocyte subsets are secreted from prostatic stroma consequent to aging and also from malignant prostate epithelium. Therefore, we hypothesized that leukocytes associated with either acute or chronic inflammation attracted to the prostate consequent to aging or tumorigenesis may promote the abnormal cellular proliferation associated with BPH and PCa. METHODS: An in vitro system designed to mimic the human prostatic microenvironment incorporating prostatic stroma (primary and immortalized prostate stromal fibroblasts), epithelium (N15C6, BPH-1, LNCaP, and PC3 cells), and inflammatory infiltrate (HL-60 cells, HH, and Molt-3 T-lymphocytes) was developed. Modified Boyden chamber assays were used to test the ability of prostate stromal and epithelial cells to attract leukocytes and to test the effect of leukocytes on prostate cellular proliferation. Antibody arrays were used to identify leukocyte-secreted cytokines mediating prostate cellular proliferation. RESULTS: Leukocytic cells migrated towards both prostate stromal and epithelial cells. CD4+ T-lymphocytes promoted the proliferation of both transformed and non-transformed prostate epithelial cell lines tested, whereas CD8+ T-lymphocytes as well as dHL-60M macrophagic and dHL-60N neutrophilic cells selectively promoted the proliferation of PCa cells. CONCLUSIONS: The results of these studies show that inflammatory cells can be attracted to the prostate tissue microenvironment and can selectively promote the proliferation of non-transformed or transformed prostate epithelial cells, and are consistent with differential role(s) for inflammatory infiltrate in the etiologies of benign and malignant proliferative disease in the prostate.


Asunto(s)
Linfocitos B/fisiología , Proliferación Celular , Transformación Celular Neoplásica/patología , Próstata/citología , Linfocitos T/fisiología , Línea Celular Transformada , Movimiento Celular/fisiología , Citocinas/metabolismo , Células Epiteliales/citología , Fibroblastos/citología , Humanos , Masculino , Próstata/metabolismo , Hiperplasia Prostática/patología , Células del Estroma/citología
9.
Mol Cell Biol ; 26(24): 9484-96, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17030615

RESUMEN

The nuclear DNA-binding protein DEK is an autoantigen that has been implicated in the regulation of transcription, chromatin architecture, and mRNA processing. We demonstrate here that DEK is actively secreted by macrophages and is also found in synovial fluid samples from patients with juvenile arthritis. Secretion of DEK is modulated by casein kinase 2, stimulated by interleukin-8, and inhibited by dexamethasone and cyclosporine A, consistent with a role as a proinflammatory molecule. DEK is secreted in both a free form and in exosomes, vesicular structures in which transcription-modulating factors such as DEK have not previously been found. Furthermore, DEK functions as a chemotactic factor, attracting neutrophils, CD8+ T lymphocytes, and natural killer cells. Therefore, the DEK autoantigen, previously described as a strictly nuclear protein, is secreted and can act as an extracellular chemoattractant, suggesting a direct role for DEK in inflammation.


Asunto(s)
Artritis Juvenil/metabolismo , Artritis Juvenil/patología , Autoantígenos/fisiología , Factores Quimiotácticos/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Proteínas Oncogénicas/metabolismo , Artritis Juvenil/inmunología , Autoantígenos/metabolismo , Células Cultivadas , Factores Quimiotácticos/fisiología , Quimiotaxis de Leucocito , Niño , Proteínas Cromosómicas no Histona/fisiología , Líquido Extracelular/inmunología , Líquido Extracelular/metabolismo , Humanos , Macrófagos/inmunología , Macrófagos/metabolismo , Monocitos/citología , Monocitos/inmunología , Monocitos/metabolismo , Proteínas Oncogénicas/fisiología , Proteínas de Unión a Poli-ADP-Ribosa
10.
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
11.
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
12.
Chem Biol ; 14(6): 623-34, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17584610

RESUMEN

The natural product withaferin A (WFA) exhibits antitumor and antiangiogenesis activity in vivo, which results from this drug's potent growth inhibitory activities. Here, we show that WFA binds to the intermediate filament (IF) protein, vimentin, by covalently modifying its cysteine residue, which is present in the highly conserved alpha-helical coiled coil 2B domain. WFA induces vimentin filaments to aggregate in vitro, an activity manifested in vivo as punctate cytoplasmic aggregates that colocalize vimentin and F-actin. WFA's potent dominant-negative effect on F-actin requires vimentin expression and induces apoptosis. Finally, we show that WFA-induced inhibition of capillary growth in a mouse model of corneal neovascularization is compromised in vimentin-deficient mice. These findings identify WFA as a chemical genetic probe of IF functions, and illuminate a potential molecular target for withanolide-based therapeutics for treating angioproliferative and malignant diseases.


Asunto(s)
Antineoplásicos/farmacología , Ergosterol/análogos & derivados , Vimentina/metabolismo , Inhibidores de la Angiogénesis/química , Inhibidores de la Angiogénesis/farmacología , Inhibidores de la Angiogénesis/uso terapéutico , Animales , Antineoplásicos/química , Antineoplásicos/uso terapéutico , Apoptosis/efectos de los fármacos , Sitios de Unión , Western Blotting , Línea Celular , Neovascularización de la Córnea/tratamiento farmacológico , Electroforesis en Gel Bidimensional , Ergosterol/química , Ergosterol/farmacología , Ergosterol/uso terapéutico , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Citometría de Flujo , Humanos , Ratones , Ratones Noqueados , Modelos Moleculares , Estructura Molecular , Unión Proteica , Vimentina/genética , Witanólidos
13.
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
14.
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
15.
Mol Cell Biol ; 24(20): 9198-206, 2004 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15456890

RESUMEN

During mammalian vascular development, endothelial cells form a complex array of vessels that differ markedly in structure and function, but the molecular basis for this vascular complexity is poorly understood. Recent insights into endothelial diversity have come from the identification of molecular markers expressed on distinct endothelial cell populations. One such marker, the PAL-E antibody, has been used for almost 20 years to distinguish blood and lymphatic vessels, but the identity of the protein recognized by PAL-E has been unknown. In the present study we have used protein purification and tandem mass spectrometry analysis of tryptic peptides to identify the PAL-E antigen as a secreted form of vimentin. Vimentin has been well characterized as an intracellular intermediate filament protein expressed broadly in mesenchymal cells. In contrast, PAL-E-reactive vimentin is secreted extracellularly, its synthesis is restricted to a distinct population of blood endothelial cells and activated macrophages, and PAL-E-reactive vimentin is found in circulating human blood. PAL-E-reactive vimentin does not arise from an endothelial cell-specific mRNA transcript but is the product of cell-specific posttranslational modification. The PAL-E antibody therefore defines secretion of vimentin as a molecular distinction among endothelial cells and exposes a novel, extracellular role for vimentin in the blood vasculature.


Asunto(s)
Anticuerpos Monoclonales/metabolismo , Vasos Sanguíneos/metabolismo , Células Endoteliales/metabolismo , Isoformas de Proteínas/sangre , Vimentina/sangre , Secuencia de Aminoácidos , Vasos Sanguíneos/citología , Línea Celular , Células Endoteliales/ultraestructura , Endotelio Vascular/citología , Humanos , Activación de Macrófagos , Macrófagos/citología , Macrófagos/metabolismo , Espectrometría de Masas , Datos de Secuencia Molecular , Fragmentos de Péptidos/química , Fragmentos de Péptidos/aislamiento & purificación , Fragmentos de Péptidos/metabolismo , Biosíntesis de Proteínas , Isoformas de Proteínas/genética , Isoformas de Proteínas/aislamiento & purificación , Isoformas de Proteínas/metabolismo , Procesamiento Proteico-Postraduccional , ARN Mensajero/metabolismo , Vimentina/genética , Vimentina/aislamiento & purificación , Vimentina/metabolismo
16.
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
17.
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.

18.
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
20.
Sci Rep ; 3: 1045, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23304436

RESUMEN

Vimentin, an abundant intermediate filament protein, presumably has an important role in stabilizing intracellular architecture, but its function is otherwise poorly understood. In a vimentin knockout (Vim KO) mouse model, we note that Vim KO mice challenged with intraperitoneal Escherichia coli control bacterial infection better than do wild-type (WT) mice. In vitro, Vim KO phagocytes show significantly increased capacity to mediate bacterial killing by abundant production of reactive oxygen species (ROS) and nitric oxides, likely due to interactions with the p47phox active subunit of NADPH oxidase. In acute colitis induced by dextran sodium sulfate (DSS), Vim KO mice develop significantly less gut inflammation than do WT mice. Further, Vim KO mice have markedly decreased bacterial extravasation in the setting of DSS-induced acute colitis, consistent with decreased intestinal disease. Our results suggest that vimentin impedes bacterial killing and production of ROS, thereby contributing to the pathogenesis of acute colitis.


Asunto(s)
Colitis/metabolismo , Vimentina/metabolismo , Animales , Colitis/inducido químicamente , Colitis/patología , Sulfato de Dextran/toxicidad , Escherichia coli/patogenicidad , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , Ratones Noqueados , NADPH Oxidasas/metabolismo , Óxido Nítrico/metabolismo , Fagocitosis , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Vimentina/antagonistas & inhibidores , Vimentina/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA