Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
1.
J Immunol ; 198(8): 3296-3306, 2017 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-28298524

RESUMEN

The p38 MAPK family is composed of four kinases of which p38α/MAPK14 is the major proinflammatory member. These kinases contribute to many inflammatory diseases, but the currently available p38 catalytic inhibitors (e.g., SB203580) are poorly effective and cause toxicity. We reasoned that the failure of catalytic p38 inhibitors may derive from their activity against noninflammatory p38 isoforms (e.g., p38ß/MAPK11) and loss of all p38α-dependent responses, including anti-inflammatory, counterregulatory responses via mitogen- and stress-activated kinase (MSK) 1/2 and Smad3. We used computer-aided drug design to target small molecules to a pocket near the p38α glutamate-aspartate (ED) substrate-docking site rather than the catalytic site, the sequence of which had only modest homology among p38 isoforms. We identified a lead compound, UM101, that was at least as effective as SB203580 in stabilizing endothelial barrier function, reducing inflammation, and mitigating LPS-induced mouse lung injury. Differential scanning fluorimetry and saturation transfer difference-nuclear magnetic resonance demonstrated specific binding of UM101 to the computer-aided drug design-targeted pockets in p38α but not p38ß. RNA sequencing analysis of TNF-α-stimulated gene expression revealed that UM101 inhibited only 28 of 61 SB203580-inhibited genes and 7 of 15 SB203580-inhibited transcription factors, but spared the anti-inflammatory MSK1/2 pathway. We provide proof of principle that small molecules that target the ED substrate-docking site may exert anti-inflammatory effects similar to the catalytic p38 inhibitors, but their isoform specificity and substrate selectivity may confer inherent advantages over catalytic inhibitors for treating inflammatory diseases.


Asunto(s)
Antiinflamatorios/farmacología , Diseño Asistido por Computadora , Células Endoteliales/efectos de los fármacos , Proteína Quinasa 14 Activada por Mitógenos/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Lesión Pulmonar Aguda/patología , Animales , Modelos Animales de Enfermedad , Diseño de Fármacos , Humanos , Ratones , Modelos Moleculares
2.
Int J Hyperthermia ; 34(1): 1-10, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-28540808

RESUMEN

BACKGROUND: As environmental and body temperatures vary, lung epithelial cells experience temperatures significantly different from normal core temperature. Our previous studies in human lung epithelium showed that: (i) heat shock accelerates wound healing and activates profibrotic gene expression through heat shock factor-1 (HSF1); (ii) HSF1 is activated at febrile temperatures (38-41 °C) and (iii) hypothermia (32 °C) activates and hyperthermia (39.5 °C) reduces expression of a subset of miRNAs that target protein kinase-Cα (PKCα) and enhance proliferation. METHODS: We analysed the effect of hypo- and hyperthermia exposure on Wnt signalling by exposing human small airway epithelial cells (SAECs) and HEK293T cells to 32, 37 or 39.5 °C for 24 h, then analysing Wnt-3a-induced epithelial-mesenchymal transition (EMT) gene expression by qRT-PCR and TOPFlash reporter plasmid activity. Effects of miRNA mimics and inhibitors and the HSF1 inhibitor, KNK437, were evaluated. RESULTS: Exposure to 39.5 °C for 24 h increased subsequent Wnt-3a-induced EMT gene expression in SAECs and Wnt-3a-induced TOPFlash activity in HEK293T cells. Increased Wnt responsiveness was associated with HSF1 activation and blocked by KNK437. Overexpressing temperature-responsive miRNA mimics reduced Wnt responsiveness in 39.5 °C-exposed HEK293T cells, but inhibitors of the same miRNAs failed to restore Wnt responsiveness in 32 °C-exposed HEK293T cells. CONCLUSIONS: Wnt responsiveness, including expression of genes associated with EMT, increases after exposure to febrile-range temperature through an HSF1-dependent mechanism that is independent of previously identified temperature-dependent miRNAs. This process may be relevant to febrile fibrosing lung diseases, including the fibroproliferative phase of acute respiratory distress syndrome (ARDS) and exacerbations of idiopathic pulmonary fibrosis (IPF).


Asunto(s)
Transición Epitelial-Mesenquimal/fisiología , Epitelio/metabolismo , Fiebre/genética , Fiebre/fisiopatología , Expresión Génica/genética , Pulmón/metabolismo , Adulto , Humanos , Masculino , Transducción de Señal
3.
RNA ; 21(7): 1261-73, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26018549

RESUMEN

Previous studies have revealed that clinically relevant changes in temperature modify clinically relevant gene expression profiles through transcriptional regulation. Temperature dependence of post-transcriptional regulation, specifically, through expression of miRNAs has been less studied. We comprehensively analyzed the effect of 24 h exposure to 32°C or 39.5°C on miRNA expression profile in primary cultured human small airway epithelial cells (hSAECs) and its impact on expression of a targeted protein, protein kinase C α (PKCα). Using microarray, and solution hybridization-based nCounter assays, with confirmation by quantitative RT-PCR, we found significant temperature-dependent changes in expression level of only five mature human miRNAs, representing only 1% of detected miRNAs. Four of these five miRNAs are the less abundant passenger (star) strands. They exhibited a similar pattern of increased expression at 32°C and reduced expression at 39.5°C relative to 37°C. As PKCα mRNA has multiple potential binding sites for three of these miRNAs, we analyzed PKCα protein expression in HEK 293T cells and hSAECs. PKCα protein levels were lowest at 32°C and highest at 39.5°C and specific miRNA inhibitors reduced these effects. Finally, we analyzed cell-cycle progression in hSAECs and found 32°C cells exhibited the greatest G1 to S transition, a process known to be inhibited by PKCα, and the effect was mitigated by specific miRNA inhibitors. These results demonstrate that exposure to clinically relevant hypothermia or hyperthermia modifies expression of a narrow subset of miRNAs and impacts expression of at least one signaling protein involved in multiple important cellular processes.


Asunto(s)
Calor , MicroARNs/metabolismo , Humanos , MicroARNs/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena de la Polimerasa
4.
Am J Physiol Lung Cell Mol Physiol ; 311(5): L941-L955, 2016 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-27638903

RESUMEN

We previously showed that coincident exposure to heat shock (HS; 42°C for 2 h) and TNF-α synergistically induces apoptosis in mouse lung epithelium. We extended this work by analyzing HS effects on human lung epithelial responses to clinically relevant injury. Cotreatment with TNF-α and HS induced little caspase-3 and poly(ADP-ribose) polymerase cleavage in human small airway epithelial cells, A549 cells, and BEAS2B cells. Scratch wound closure rates almost doubled when A549 and BEAS2B cells and air-liquid interface cultures of human bronchial epithelial cells were heat shocked immediately after wounding. Microarray, qRT-PCR, and immunoblotting showed fibroblast growth factor 1 (FGF1) to be synergistically induced by HS and wounding. Enhanced FGF1 expression in HS/wounded A549 was blocked by inhibitors of p38 MAPK (SB203580) or HS factor (HSF)-1 (KNK-437) and in HSF1 knockout BEAS2B cells. PCR demonstrated FGF1 to be expressed from the two most distal promoters in wounded/HS cells. Wound closure in HS A549 and BEAS2B cells was reduced by FGF receptor-1/3 inhibition (SU-5402) or FGF1 depletion. Exogenous FGF1 accelerated A549 wound closure in the absence but not presence of HS. In the presence of exogenous FGF1, HS slowed wound closure, suggesting that it increases FGF1 expression but impairs FGF1-stimulated wound closure. Frozen sections from normal and idiopathic pulmonary fibrosis (IPF) lung were analyzed for FGF1 and HSP70 by immunofluorescence confocal microscopy and qRT-PCR. FGF1 and HSP70 mRNA levels were 7.5- and 5.9-fold higher in IPF than normal lung, and the proteins colocalized to fibroblastic foci in IPF lung. We conclude that HS signaling may have an important impact on gene expression contributing to lung injury, healing, and fibrosis.


Asunto(s)
Epitelio/metabolismo , Epitelio/patología , Factor 1 de Crecimiento de Fibroblastos/metabolismo , Respuesta al Choque Térmico , Lesión Pulmonar/patología , Animales , Apoptosis/genética , Sitios de Unión , Línea Celular , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Células Epiteliales/metabolismo , Células Epiteliales/patología , Factor 1 de Crecimiento de Fibroblastos/genética , Regulación de la Expresión Génica , Proteínas HSP70 de Choque Térmico/metabolismo , Factores de Transcripción del Choque Térmico , Respuesta al Choque Térmico/genética , Humanos , Fibrosis Pulmonar Idiopática/genética , Pulmón/metabolismo , Pulmón/patología , Lesión Pulmonar/genética , Ratones , Reacción en Cadena de la Polimerasa , Regiones Promotoras Genéticas/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Cicatrización de Heridas/genética
5.
J Biol Chem ; 288(4): 2756-66, 2013 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-23212905

RESUMEN

Heat shock protein (Hsp) 70 expression can be stimulated by febrile range temperature (FRT). Hsp70 has been shown to be elevated in serum of patients with sepsis, and when released from cells, extracellular Hsp70 exerts endotoxin-like effects through Toll-like receptor 4 (TLR4) receptors. Circulating TLR agonists and fever both persist for the first several days of sepsis, and each can activate Hsp70 expression; however, the effect of combined exposure to FRT and TLR agonists on Hsp70 expression is unknown. We found that concurrent exposure to FRT (39.5 °C) and agonists for TLR4 (LPS), TLR2 (Pam3Cys), or TLR3 (poly(IC)) synergized to increase Hsp70 expression and extracellular release in RAW264.7 macrophages. The increase in Hsp70 expression was associated with activation of p38 and ERK MAP kinases, phosphorylation of histone H3, and increased recruitment of HSF1 to the Hsp70 promoter. Pretreatment with the p38 MAPK inhibitor SB283580 but not the ERK pathway inhibitor UO126 significantly reduced Hsp70 gene modification and Hsp70 expression in RAW cells co-exposed to LPS and FRT. In mice challenged with intratracheal LPS and then exposed to febrile range hyperthermia (core temperature, ∼39.5 °C), Hsp70 levels in lung tissue and in cell-free lung lavage were increased compared with mice exposed to either hyperthermia or LPS alone. We propose a model of how enhanced Hsp70 expression and extracellular release in patients concurrently exposed to fever and TLR agonists may contribute to the pathogenesis of sepsis.


Asunto(s)
Fiebre/metabolismo , Proteínas HSP70 de Choque Térmico/metabolismo , Sepsis/metabolismo , Receptores Toll-Like/agonistas , Animales , Línea Celular Tumoral , Humanos , Inflamación , Lipopolisacáridos/metabolismo , Sistema de Señalización de MAP Quinasas , Macrófagos/metabolismo , Masculino , Ratones , Modelos Biológicos , ARN Interferente Pequeño/metabolismo , Choque/metabolismo , Transducción de Señal , Receptor Toll-Like 4/metabolismo
6.
Int J Hyperthermia ; 29(5): 423-35, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23863046

RESUMEN

The heat shock response is a highly conserved primitive response that is essential for survival against a wide range of stresses, including extremes of temperature. Fever is a more recently evolved response, during which organisms raise their core body temperature and temporarily subject themselves to thermal stress in the face of infections. The present review documents studies showing the potential overlap between the febrile response and the heat shock response and how both activate the same common transcriptional programme (although with different magnitudes) including the stress-activated transcription factor, heat shock factor-1, to modify host defences in the context of infection, inflammation and injury. The review focuses primarily on how hyperthermia within the febrile range that often accompanies infections and inflammation acts as a biological response modifier and modifies innate immune responses. The characteristic 2-3 °C increase in core body temperature during fever activates and utilises elements of the heat shock response pathway to modify cytokine and chemokine gene expression, cellular signalling and immune cell mobilisation to sites of inflammation, infection and injury. Interestingly, typical proinflammatory agonists such as Toll-like receptor agonists modify the heat shock-induced transcriptional programme and expression of HSP genes following co-exposure to febrile range hyperthermia or heat shock, suggesting a complex reciprocal regulation between the inflammatory pathway and the heat shock response pathway.


Asunto(s)
Fiebre/fisiopatología , Respuesta al Choque Térmico/fisiología , Animales , Proteínas de Choque Térmico/fisiología , Humanos , Infecciones/fisiopatología , Inflamación/fisiopatología , Receptores Toll-Like/agonistas , Receptores Toll-Like/fisiología
7.
Am J Respir Cell Mol Biol ; 46(6): 807-14, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22281986

RESUMEN

Acute respiratory distress syndrome (ARDS) is a neutrophil (polymorphonuclear leukocyte; PMN)-driven lung injury that is associated with fever and heat-stroke, and involves approximately 40% mortality. In murine models of acute lung injury (ALI), febrile-range hyperthermia (FRH) enhanced PMN accumulation, vascular permeability, and epithelial injury, in part by augmenting pulmonary cysteine-x-cysteine (CXC) chemokine expression. To determine whether FRH increases chemokine responsiveness within the lung, we used in vivo and in vitro models that bypass the endogenous generation of chemokines. We measured PMN transalveolar migration (TAM) in mice after intratracheal instillations of the human CXC chemokine IL-8 in vivo, and of IL-8-directed PMN transendothelial migration (TEM) through human lung microvascular endothelial cell (HMVEC-L) monolayers in vitro. Pre-exposure to FRH increased in vivo IL-8-directed PMN TAM by 23.5-fold and in vitro TEM by 7-fold. Adoptive PMN transfer demonstrated that enhanced PMN TAM required both PMN donors and recipients to be exposed to FRH, suggesting interdependent effects on PMNs and endothelium. FRH exposure caused the activation of extracellular signal-regulated kinase (ERK) and p38 mitogen-activated protein kinase in lung homogenates and circulating PMNs, with an associated increase in HSP27 phosphorylation and stress-fiber formation. The inhibition of these signaling pathways with U0126 and SB203580 blocked the effects of FRH on PMN extravasation in vivo and in vitro. Collectively, these results (1) demonstrate that FRH augments chemokine-directed PMN extravasation through direct effects on endothelium and PMNs, (2) identify ERK and p38 signaling pathways in the effect, and (3) underscore the complex effects of physiologic temperature change on innate immune function and its potential consequences for lung injury.


Asunto(s)
Endotelio/patología , Fiebre/patología , Fiebre/fisiopatología , Neutrófilos/patología , Animales , Ratones
8.
Am J Respir Cell Mol Biol ; 47(6): 824-33, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22962066

RESUMEN

Hyperthermia has been shown to confer cytoprotection and to augment apoptosis in different experimental models. We analyzed the mechanisms of both effects in the same mouse lung epithelial (MLE) cell line (MLE15). Exposing MLE15 cells to heat shock (HS; 42°C, 2 h) or febrile-range hyperthermia (39.5°C) concurrent with activation of the death receptors, TNF receptor 1 or Fas, greatly accelerated apoptosis, which was detectable within 30 minutes and was associated with accelerated activation of caspase-2, -8, and -10, and the proapoptotic protein, Bcl2-interacting domain (Bid). Caspase-3 activation and cell death were partially blocked by inhibitors targeting all three initiator caspases. Cells expressing the IκB superrepessor were more susceptible than wild-type cells to TNF-α-induced apoptosis at 37°C, but HS and febrile-range hyperthermia still increased apoptosis in these cells. Delaying HS for 3 hours after TNF-α treatment abrogated its proapoptotic effect in wild-type cells, but not in IκB superrepressor-expression cells, suggesting that TNF-α stimulates delayed resistance to the proapoptotic effects of HS through an NF-κB-dependent mechanism. Pre-exposure to 2-hour HS beginning 6 to16 hours before TNF-α treatment or Fas activation reduced apoptosis in MLE15 cells. The antiapoptotic effects of HS pretreatment were reduced in TNF-α-treated embryonic fibroblasts from heat shock factor-1 (HSF1)-deficient mice, but the proapoptotic effects of concurrent HS were preserved. Thus, depending on the temperature and timing relative to death receptor activation, hyperthermia can exert pro- and antiapoptotic effects through distinct mechanisms.


Asunto(s)
Apoptosis , Células Epiteliales/fisiología , Respuesta al Choque Térmico , Sistema Respiratorio/citología , Análisis de Varianza , Animales , Proteína Proapoptótica que Interacciona Mediante Dominios BH3/metabolismo , Inhibidores de Caspasas/farmacología , Caspasas/metabolismo , Línea Celular , Supervivencia Celular , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Activación Enzimática , Factores de Transcripción del Choque Térmico , Ratones , Ratones Noqueados , FN-kappa B/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Factor de Necrosis Tumoral alfa/farmacología , Factor de Necrosis Tumoral alfa/fisiología
9.
Int J Hyperthermia ; 28(8): 747-55, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23153218

RESUMEN

PURPOSE: Chronic heat exposure in mice has cellular and physiological effects that improve thermal tolerance [1], but also modifies innate immune responses with potential adverse consequences [2]. While male and female mice are known to respond differently to acute exposure to severe hyperthermia, sex-based differences in responses to chronic moderate heat exposure have not been reported. The major objective of this study was to compare the tolerance of male and female mice for chronic heat exposure. MATERIALS AND METHODS: We used a mouse model of 5-day moderate heat exposure (ambient temperature ∼37°C) to compare the physiological and cellular heat shock response in male and female mice. Core temperature, heart rate, and activity were monitored telemetrically and heat shock protein levels were measured in brain and lung by western blotting. RESULTS: Adult CD-1 female mice maintained a 1.2°C lower core temperature (38.31 ± 0.64 versus 39.51 ± 0.72°C; p = 0.002), experienced less weight loss (1.54 ± 0.45 versus 4.54 ± 1.97 g; p = 0.0007), and had improved survival (16/16 survived versus 13/21, p < 0.006) than male mice of the same age. After 5 days of moderate heat exposure Hsp72 levels in brain and lung increased 2.1-fold (p = 0.007) and 5-fold (p = 0.048) in male mice compared with 1.3- (p = 0.054) and 1.5-fold (p = 0.134) in female mice. CONCLUSIONS: This study reveals previously unknown and potentially important differences between male and female mice in physiological and cellular responses to chronic heat exposure, which had consequences for survival. Future studies may identify biomarkers of differential heat tolerance and treatments to improve heat tolerance in humans.


Asunto(s)
Adaptación Fisiológica/fisiología , Encéfalo , Respuesta al Choque Térmico/fisiología , Calor/efectos adversos , Animales , Temperatura Corporal , Encéfalo/metabolismo , Femenino , Proteínas de Choque Térmico/metabolismo , Humanos , Pulmón/metabolismo , Masculino , Ratones , Caracteres Sexuales
10.
Int J Hyperthermia ; 28(7): 627-35, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22834633

RESUMEN

Fever commonly occurs in acute lung injury (ALI) and ALI occurs in 25% of victims of heat stroke. We have shown in mouse models of ALI that exposure to febrile-range hyperthermia (FRH), 39.5°C, increases non-cardiogenic pulmonary oedema. In this study we studied the direct effects of FRH on endothelial barrier integrity using human microvascular endothelial cells (HMVEC-Ls). We analysed the effect of exposure to culture temperatures between 38.5° and 41°C with and without tumour necrosis factor-α (TNF-α) up to 250 U/mL for 6-24 h. We found that exposure to 2.5-250 U/mL TNF-α increased HMVEC-L permeability by 4.1-15.8-fold at 37°C. Exposure to 39.5°C alone caused variable, modest, lot-specific increases in HMVEC-L permeability, however raising culture temperature to 39.5°C in the presence of TNF-α increased permeability an additional 1.6-4.5-fold compared with cells incubated with the same TNF-α concentration at 37°C. Permeability occurred without measurable cytotoxicity and was reversible upon removal of TNF-α and reduction in temperature to 37°C. Exposure to 39.5°C or TNF-α each stimulated rapid activation of p38 and ERK but the effects were not additive. Treatment with inhibitors of ERK (U0126) or p38 (SB203580) each reduced TNF-α-induced permeability in 39.5°C monolayers to levels in 37°C cells, but did not alter TNF-α-induced permeability in the 37°C cells. These results demonstrate that FRH directly increases paracellular pathway opening through a process that requires ERK and p38 MAPKs. A better understanding of this mechanism may provide new understanding about how fever may contribute to the pathogenesis of ALI and provide new therapeutic targets to improve clinical outcomes.


Asunto(s)
Células Endoteliales/metabolismo , Fiebre/metabolismo , Línea Celular , Endotelio Vascular/citología , Humanos , Pulmón/citología , Sistema de Señalización de MAP Quinasas/fisiología , Permeabilidad , Factor de Necrosis Tumoral alfa , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
11.
Cytokine ; 54(1): 61-7, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21266308

RESUMEN

The heat shock (HS) response, a phylogenetically conserved ubiquitous response to stress, is generally characterized by the induced expression of heat shock protein (HSP) genes. Our earlier studies showed that the stress-activated transcription factor, heat shock factor-1 (HSF1), activated at febrile range or HS temperatures also modified expression of non-HSP genes including cytokine and chemokine genes. We also showed by in silico analysis that 28 among 29 human and mouse CXC chemokine genes had multiple putative heat shock response elements (HSEs) present in their gene promoters. To further determine whether these potential HSEs were functional and bound HSF1, we analyzed the recruitment of HSF1 to promoters of 5 human CXC chemokine genes (CXCL-1, 2, 3, 5 and 8) by chromatin immunoprecipitation (ChIP) assay and analyzed the effect of HS exposure on tumor necrosis factor-α (TNFα)-induced expression of these genes in human lung epithelial-like A549 cells. HSF1 ChIP analysis showed that HSF1 was recruited to all but one of these CXC chemokine genes (CXCL-3) and HS caused a significant increase in recruitment of HSF1 to one or multiple HSEs present in the promoters of CXCL-1, 2, 5 and 8 genes. However, the effect of HS exposure on expression of these genes showed a variable gene-specific effect. For example, CXCL8 expression was markedly enhanced (p<0.05) whereas CXCL5 expression was significantly repressed (p<0.05) in cells exposed to HS coincident with TNFα stimulation. In contrast, expression of CXCL1 and CXCL2, despite HSF1 recruitment to their promoters, was not affected by HS exposure. Our results indicate that some, if not all, putative HSEs present in the CXC chemokine gene promoters are functional and recruit HSF1 in vivo but the effects on gene expression are variable and gene specific. We speculate, the physical proximity and interactions of other transcription factors and co-regulators with HSF1 could be critical to determining the effects of HS on the expression of these genes.


Asunto(s)
Quimiocinas/metabolismo , Proteínas de Unión al ADN/biosíntesis , Regulación de la Expresión Génica , Receptores CXCR/biosíntesis , Factores de Transcripción/biosíntesis , Animales , Línea Celular Tumoral , Quimiocina CXCL1/metabolismo , Quimiocina CXCL2/metabolismo , Quimiocina CXCL5/metabolismo , Inmunoprecipitación de Cromatina , Proteínas de Unión al ADN/metabolismo , Factores de Transcripción del Choque Térmico , Humanos , Interleucina-8/metabolismo , Ratones , Modelos Biológicos , Regiones Promotoras Genéticas , Receptores CXCR/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Transcripción/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
12.
J Immunol ; 183(3): 1657-66, 2009 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-19592659

RESUMEN

CD1d is a nonclassical Ag-presenting molecule that presents glycolipid Ags to NKT cells that are involved in immune defense and tumor rejection. It also plays a role in immunoregulatory functions in the epidermis. The mechanisms controlling the expression of CD1d are not well understood. Therefore, we cloned the CD1d gene promoter and characterized its activities in primary human keratinocytes and other cell lines of epithelial origin. We found that a CCAAT box in the CD1d promoter is required for its expression in keratinocytes. We show here that transcription factor C/EBP-beta binds to the CCAAT box in the CD1d promoter in vitro and in vivo. Consistent with these observations, deletion of the gene encoding for C/EBP-beta caused a loss of CD1d expression. The in vivo regulation of CD1d has significant implications for the pathologic mechanisms of certain immunologic skin diseases in which NKT cells play a role, such as allergic contact dermatitis and psoriasis. Together, these data show a central role for C/EBP-beta in regulating CD1d transcription.


Asunto(s)
Antígenos CD1d/genética , Proteína beta Potenciadora de Unión a CCAAT/fisiología , Regulación de la Expresión Génica , Queratinocitos/metabolismo , Transcripción Genética , Sitios de Unión , Línea Celular , Clonación Molecular , Células Epiteliales , Humanos , Células T Asesinas Naturales , Regiones Promotoras Genéticas , Enfermedades de la Piel/etiología , Enfermedades de la Piel/inmunología
13.
Int J Hyperthermia ; 27(7): 717-25, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21992563

RESUMEN

The effects of heat, especially long-term heat exposure, are complex and incompletely understood and few studies have analysed the immunological consequences of such exposures. In the present study we analysed how long-term hyperthermia modified the pulmonary immune responses, especially recruitment of neutrophils to sites of inflammation, infection and injury. Using our mouse model of long-term whole body hyperthermia (continuous 5-day passive febrile range hyperthermia (5d-FRH)) we found that bacterial lipopolysaccharide (LPS) challenge greatly increased neutrophil accumulation in bronchoalveolar lavage and lung parenchyma in 5d-FRH exposed mice in comparison to LPS-treated controls. Moreover, the effect was sustained, and persisted during the post-exposure recovery period, and LPS challenge on days 5-7 post-recovery also exhibited similarly augmented neutrophil response. Lung lavage from 5d-FRH mice, either immediately or up to 7 days post-exposure, showed significantly increased levels of ELR + CXC chemokines, KC or LIX in response to LPS challenge, indicating that enhanced chemokines could contribute to the increased recruitment of neutrophils to the lung. However, an in vivo neutrophil migration assay following 5d-FRH and during the post-exposure recovery period also showed persistently enhanced neutrophil influx in response to a fixed chemotactic gradient generated by recombinant human IL-8, suggesting that additional mechanisms besides increased ELR + CXC chemokines contributed to the augmented neutrophil response caused by 5d-FRH exposure. These previously unappreciated profound and lasting effects of long-term hyperthermia may have important consequences and may help explain the increased risk of respiratory illnesses in active duty personnel and returning veterans.


Asunto(s)
Fiebre/inmunología , Trastornos de Estrés por Calor/inmunología , Pulmón/inmunología , Infiltración Neutrófila/inmunología , Animales , Humanos , Hipertermia Inducida , Interleucina-8/farmacología , Lipopolisacáridos/farmacología , Pulmón/efectos de los fármacos , Masculino , Ratones , Infiltración Neutrófila/efectos de los fármacos
14.
Am J Physiol Cell Physiol ; 298(1): C171-81, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19846753

RESUMEN

We previously showed that exposure to febrile-range temperatures (FRT, 39.5-40 degrees C) reduces LPS-induced TNF-alpha expression, in part through the direct interaction of heat shock factor-1 (HSF1) with the TNF-alpha gene promoter. However, it is not known whether exposure to FRT also modifies more proximal LPS-induced signaling events. Using HSF1-null mice, we confirmed that HSF1 is required for FRT-induced repression of TNF-alpha in vitro by LPS-stimulated bone marrow-derived macrophages and in vivo in mice challenged intratracheally with LPS. Exposing LPS-stimulated RAW 264.7 mouse macrophages to FRT reduced TNF-alpha expression while increasing IL-1beta expression despite the two genes sharing a common myeloid differentiation protein-88 (MyD88)-dependent pathway. Global activation of the three LPS-induced signaling intermediates that lead to cytokine gene expression, ERK and p38 MAPKs and NF-kappaB, was not affected by exposing RAW 264.7 cells to FRT as assessed by ERK and p38 phosphorylation and NF-kappaB in vitro DNA-binding activity and activation of a NF-kappaB-dependent synthetic promoter. However, chromatin immunoprecipitation (ChIP) analysis demonstrated that exposure to FRT reduced LPS-induced recruitment of NF-kappaB p65 to the TNF-alpha promoter while simultaneously increasing its recruitment to the IL-1beta promoter. These data suggest that FRT exerts its effects on cytokine gene expression in a gene-specific manner through distal effects on promoter activation rather than proximal receptor activation and signal transduction.


Asunto(s)
Citocinas/genética , Fiebre/fisiopatología , Regulación de la Expresión Génica/fisiología , Lipopolisacáridos/farmacología , Macrófagos/fisiología , FN-kappa B/fisiología , Regiones Promotoras Genéticas , Factor de Necrosis Tumoral alfa/genética , Animales , Cromatina/fisiología , Cruzamientos Genéticos , ADN/genética , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/fisiología , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Factores de Transcripción del Choque Térmico , Interleucina-1beta/genética , Macrófagos/efectos de los fármacos , Masculino , Ratones , Ratones Noqueados , Reacción en Cadena de la Polimerasa/métodos , ARN/genética , ARN/aislamiento & purificación , Temperatura , Factores de Transcripción/deficiencia , Factores de Transcripción/fisiología
15.
Prostaglandins Other Lipid Mediat ; 93(1-2): 1-7, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20382255

RESUMEN

The heat shock (HS) response is an important cytoprotective response comprising the expression of heat shock proteins (HSPs) and orchestrated by the heat/stress-induced transcription factor, heat shock factor-1 (HSF-1). Previous studies suggest that the activation threshold and magnitude of the HS response may be modified by treatment with arachidonic acid (AA). We analyzed the effect of exogenous AA and its metabolites, PGE(2), LTD(4), and 15-HETE on HSF-1-dependent gene expression in A549 human respiratory epithelial-like cells. When added at 1microM, PGE(2) much more than LTD(4), but not 15-HETE increased activity of a synthetic HSF-1-dependent reporter after HS exposure (42 degrees C for 2h), but had no effect in the absence of HS. Exposing A549 cells to HS stimulated the release of PGE(2) and treatment with the cyclooxygenase inhibitor, ibuprofen, reduced HS-induced HSF-1-dependent transcription. PGE(2) increased HS-induced HSP72 mRNA and protein expression but EMSA and Western blot analysis failed to show an effect on HSF-1 DNA binding activity or post-translational modification. In summary, we showed that HS stimulates the generation of PGE(2), which augments the generation of HSPs. The clinical consequences of this pathway have yet to be determined.


Asunto(s)
Dinoprostona/farmacología , Proteínas del Choque Térmico HSP72/genética , Respuesta al Choque Térmico/genética , Línea Celular Tumoral , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Dinoprostona/metabolismo , Proteínas del Choque Térmico HSP72/metabolismo , Factores de Transcripción del Choque Térmico , Proteínas de Choque Térmico/genética , Proteínas de Choque Térmico/metabolismo , Humanos , Proteínas de Plantas/genética , Proteínas de Plantas/metabolismo , ARN Mensajero/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
16.
J Immunol ; 181(4): 2636-43, 2008 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-18684954

RESUMEN

Human neutrophilic polymorphonuclear leukocytes (PMNs) are central to innate immunity and are responsible for clearance of pathogens. PMNs undergo a tightly regulated apoptosis program that allows for timely clearance of PMNs without extravasation of toxic intracellular contents. We investigated the rate of spontaneous apoptosis of human peripheral blood PMNs cultured at basal (37 degrees C) and febrile-range (39.5 degrees C) temperatures (FRT). We found that PMN apoptosis is accelerated at FRT, reaching approximately 90% completion by 8 h at 39.5 degrees C vs 18 h at 37 degrees C based on morphologic criteria. Caspase-8 activation peaked within 15 min of PMN exposure to FRT, and subsequent activation of caspase-3 and -9, cleavage of the BH3 (Bcl-2 homology domain 3) only protein Bid, and mitochondrial release of cytochrome c were also greater in FRT-exposed PMNs. Inhibition of caspase-3, -8, and -9 conferred comparable protection from apoptosis in FRT-exposed PMNs. These results demonstrate that exposure to FRT enhances caspase-8 activation and subsequent mitochondrial-dependent and mitochondrial-independent apoptosis pathways. The PMN survival factors G-CSF, GM-CSF, and IL-8 each prolonged PMN survival at 37 degrees C and 39.5 degrees C, but did not reduce the difference in survival at the two temperatures. In a mouse model of intratracheal endotoxin-induced alveolitis, coexposure to FRT (core temperature approximately 39.5 degrees C) doubled the proportion of bronchoalveolar PMNs undergoing apoptosis compared with euthermic mice. This process may play an important role in limiting inflammation and tissue injury during febrile illnesses.


Asunto(s)
Caspasas/fisiología , Fiebre/enzimología , Fiebre/patología , Neutrófilos/enzimología , Neutrófilos/patología , Animales , Caspasa 3/metabolismo , Caspasa 8/metabolismo , Caspasa 9/metabolismo , Caspasas/metabolismo , Movimiento Celular/inmunología , Supervivencia Celular/inmunología , Células Cultivadas , Activación Enzimática/inmunología , Fiebre/inmunología , Humanos , Mediadores de Inflamación/metabolismo , Mediadores de Inflamación/fisiología , Ratones , Temperatura
17.
Am J Respir Cell Mol Biol ; 39(2): 235-42, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18367728

RESUMEN

The heat shock (HS) response is a phylogenetically ancient cellular response to stress, including heat, that shifts gene expression to a set of conserved HS protein (HSP) genes. In our earlier studies, febrile-range hyperthermia (FRH) not only activated HSP gene expression, but also increased expression of CXC chemokines in mice, leading us to hypothesize that the CXC chemokine family of genes might be HS-responsive. To address this hypothesis we analyzed the effect of HS on the expression of IL-8/CXCL-8, a member of the human CXC family of ELR(+) chemokines. HS markedly enhanced TNF-alpha-induced IL-8 secretion in human A549 respiratory epithelial-like cells and in primary human small airway epithelial cells. IL-8 mRNA was also up-regulated by HS, but the stability of IL-8 mRNA was not affected. TNF-alpha-induced reporter activity of an IL-8 promoter construct IL8(-1471/+44)-luc stably transfected in A549 cells was also enhanced by HS. Electrophoretic mobility and chromatin immunoprecipitation assays showed that the stress-activated transcription factor heat shock factor-1 (HSF-1) binds to at least two putative heat shock response elements (HSE) present in the IL-8 promoter. Deletional reporter constructs lacking either one or both of these sites showed reduced HS responsiveness. Furthermore, depletion of HSF-1 using siRNA also reduced the effects HS on TNF-alpha-induced IL-8 expression, demonstrating that HSF-1 could also act to regulate IL-8 gene transcription. We speculate that during evolution the CXC chemokine genes may have co-opted elements of the HS response to amplify their expression and enhance neutrophil delivery during febrile illnesses.


Asunto(s)
Proteínas de Choque Térmico/metabolismo , Respuesta al Choque Térmico/fisiología , Interleucina-8/metabolismo , Activación Transcripcional/fisiología , Animales , Línea Celular , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Fiebre/metabolismo , Factores de Transcripción del Choque Térmico , Humanos , Interleucina-8/genética , Ratones , Ratones Noqueados , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
18.
J Neuroimmunol ; 196(1-2): 67-81, 2008 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-18471898

RESUMEN

Interferon-beta (IFN-beta), an approved treatment of multiple sclerosis (MS), produces only partial clinical responses. IFN-beta therapy has been limited by its short serum half-life and limited ability to cross the blood brain barrier. We have developed a means of delivering the IFN-beta gene both systemically and into the central nervous system (CNS) using bone marrow stem cells (BMSCs) as a vehicle and examined the therapeutic efficacy of this approach in experimental autoimmune encephalomyelitis (EAE), an animal model of MS. A retroviral expression vector (pLXSN-IFNbeta) was used to stably transfect virus producer PA317 cells to generate retrovirus containing the IFN-beta gene which then was used to transduce BMSCs. IFN-beta engineered BMSCs were transplanted (i.v.) into mice that then were immunized with proteolipoprotein (PLP) to initiate EAE. IFN-beta-engineered BMSCs transplanted mice showed a significant inhibition of EAE onset, and the overall clinical severity was less compared to control groups. IFN-beta delivery strongly reduced infiltration of mononuclear cells possibly by inhibiting cell adhesion molecules. Reduced demyelination and increased remyelination were also observed in the IFN-beta treated group. Furthermore, inhibition of the pro-inflammatory cytokines TNF-alpha, IFN-gamma and IL-12 and enhanced expression of the anti-inflammatory cytokines IL-10, IL-4 and TGF-beta was observed in CNS tissue. In addition, mice receiving IFN-beta had reduced apoptosis and increases in growth promoting factors including BDNF, CNTF, PDGF and VEGF. These results suggest that BMSCs can be used as vehicles to deliver the IFN-beta into the CNS. This is a potentially novel therapeutic approach which might be used in MS and other diseases of the CNS in which drug access is limited.


Asunto(s)
Células de la Médula Ósea/fisiología , Trasplante de Médula Ósea/métodos , Encefalomielitis Autoinmune Experimental/prevención & control , Interferón beta/uso terapéutico , Animales , Antígenos CD34/metabolismo , Células de la Médula Ósea/metabolismo , Caspasa 3/genética , Caspasa 3/metabolismo , Células Cultivadas , Citocinas/genética , Citocinas/metabolismo , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/inducido químicamente , Ensayo de Inmunoadsorción Enzimática/métodos , Femenino , Técnicas de Transferencia de Gen , Etiquetado Corte-Fin in Situ , Molécula 1 de Adhesión Intercelular/genética , Molécula 1 de Adhesión Intercelular/metabolismo , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Interferón beta/biosíntesis , Interferón beta/genética , Ratones , Proteína Básica de Mielina/genética , Proteína Básica de Mielina/metabolismo , Proteína Proteolipídica de la Mielina , Fragmentos de Péptidos , Prevención Secundaria , Molécula 1 de Adhesión Celular Vascular/genética , Molécula 1 de Adhesión Celular Vascular/metabolismo
19.
J Neuroimmunol ; 292: 9-20, 2016 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-26943953

RESUMEN

7,8-Dihydroxyflavone (DHF), is a recently described TrkB agonist that readily crosses the blood brain barrier. We treated C57Bl/6 mice with MOG--induced EAE daily with DHF starting on the day of disease induction. Clinical severity of impairment was reduced throughout the course of disease. Pathological examination of brains and spinal cords on day 28 showed that DHF treatment increased the phosphorylation of TrkB and activated downstream signaling pathways including AKT and STAT3 and reduced inflammation, demyelination and axonal loss compared to EAE controls. DHF treatment duplicated the central nervous system effects of brain derived neurotrophic factor in the EAE.


Asunto(s)
Encéfalo/patología , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Encefalomielitis Autoinmune Experimental/patología , Flavonas/uso terapéutico , Esclerosis Múltiple/tratamiento farmacológico , Médula Espinal/patología , 2',3'-Nucleótido Cíclico Fosfodiesterasas/metabolismo , Animales , Apoptosis/efectos de los fármacos , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Citocinas/metabolismo , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/inmunología , Femenino , Humanos , Ratones , Ratones Endogámicos C57BL , Proteína Básica de Mielina/metabolismo , Glicoproteína Mielina-Oligodendrócito/toxicidad , Fragmentos de Péptidos/toxicidad , Índice de Severidad de la Enfermedad , Transducción de Señal/efectos de los fármacos , Médula Espinal/efectos de los fármacos , Médula Espinal/metabolismo , Factores de Tiempo , Proteína X Asociada a bcl-2/metabolismo
20.
J Appl Physiol (1985) ; 98(5): 1799-804, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-15829718

RESUMEN

We previously showed that sustained exposure to febrile-range hyperthermia (FRH) for 24 h caused an increase in circulating granulocyte colony-stimulating factor (G-CSF) levels and a peripheral neutrophilia in mice (Hasday J, Garrison A, Singh I, Standiford T, Ellis G, Rao S, He JR, Rice P, Frank M, Goldblum S, and Viscardi R. Am J Pathol 162: 2005-2017, 2003). In this study, we utilized a conscious temperature-clamped mouse model to analyze the kinetics of G-CSF expression and peripheral neutrophil expansion and the contributions of FRH-induced G-CSF expression, glucocorticoid generation, and catecholamine-induced neutrophil demargination. In conscious mice housed at an ambient temperature of 34.5 degrees C, core temperature rapidly equilibrated at 39.5-40 degrees C. Peripheral neutrophil counts increased 2-fold after 24-h exposure to hyperthermia, peaked at 3.6-fold baseline levels after 36-h exposure to FRH, and returned to baseline levels after 42 h of sustained hyperthermia. Plasma G-CSF levels were increased by 6.8-fold after 24 h and peaked at 40-fold baseline levels after 36 h in the hyperthermic mice. Plasma corticosterone levels peaked at 3.3-fold baseline levels after 30-h sustained hyperthermia and returned to baseline by 42 h. Immunoneutralization of G-CSF blocked FRH-induced peripheral neutrophilia, but blockade of the glucocorticoid receptor with mifepristone failed to modify FRH-induced neutrophilia. Epinephrine induced similar increases in peripheral blood absolute neutrophil counts in euthermic mice (2.2-fold increase) and mice exposed to FRH for 36 h (1.8-fold increase). Collectively, these data suggest that FRH-induced expression of G-CSF drives the sustained peripheral neutrophilia that occurs during sustained (36 h) hyperthermia, whereas glucocorticoid generation and catecholamine-induced demargination play little role in this response.


Asunto(s)
Corticosterona/sangre , Fiebre/sangre , Factor Estimulante de Colonias de Granulocitos/sangre , Neutrófilos/metabolismo , Animales , Factor Estimulante de Colonias de Granulocitos/fisiología , Masculino , Ratones , Neutrófilos/citología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA