Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Harm Reduct J ; 20(1): 110, 2023 08 16.
Artículo en Inglés | MEDLINE | ID: mdl-37587466

RESUMEN

BACKGROUND: The opioid crisis continues in full force, as physicians and caregivers are desperate for resources to help patients with opioid use and chronic pain disorders find safer and more accessible non-opioid tools. MAIN BODY: The purpose of this article is to review the current state of the opioid epidemic; the shifting picture of cannabinoids; and the research, policy, and current events that make opioid risk reduction an urgent public health challenge. The provided table contains an evidence-based clinical framework for the utilization of cannabinoids to treat patients with chronic pain who are dependent on opioids, seeking alternatives to opioids, and tapering opioids. CONCLUSION: Based on a comprehensive review of the literature and epidemiological evidence to date, cannabinoids stand to be one of the most interesting, safe, and accessible tools available to attenuate the devastation resulting from the misuse and abuse of opioid narcotics. Considering the urgency of the opioid epidemic and broadening of cannabinoid accessibility amidst absent prescribing guidelines, the authors recommend use of this clinical framework in the contexts of both clinical research continuity and patient care.


Asunto(s)
Dolor Crónico , Epidemias , Humanos , Analgésicos Opioides/uso terapéutico , Epidemia de Opioides , Dolor Crónico/tratamiento farmacológico , Narcóticos
2.
Am J Physiol Lung Cell Mol Physiol ; 321(5): L837-L843, 2021 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-34494468

RESUMEN

Bronchoalveolar lavage (BAL) samples from Severe Asthma Research Program (SARP) patients display suppression of a module of genes involved in cAMP-signaling pathways (BALcAMP) correlating with severity, therapy, and macrophage constituency. We sought to establish if gene expression changes were specific to macrophages and compared gene expression trends from multiple sources. Datasets included single-cell RNA sequencing (scRNA-seq) from lung specimens including a fatal exacerbation of severe Asthma COPD Overlap Syndrome (ACOS) after intense therapy and controls without lung disease, bulk RNA sequencing from cultured macrophage (THP-1) cells after acute or prolonged ß-agonist exposure, SARP datasets, and data from the Immune Modulators of Severe Asthma (IMSA) cohort. THP monocytes suppressed BALcAMP network gene expression after prolonged relative to acute ß-agonist exposure, corroborating SARP observations. scRNA-seq from healthy and diseased lung tissue revealed 13 cell populations enriched for macrophages. In severe ACOS, BALcAMP gene network expression scores were decreased in many cell populations, most significantly for macrophage populations (P < 3.9e-111). Natural killer (NK) cells and type II alveolar epithelial cells displayed less robust network suppression (P < 9.2e-8). Alveolar macrophages displayed the most numerous individual genes affected and the highest amplitude of modulation. Key BALcAMP genes demonstrate significantly decreased expression in severe asthmatics in the IMSA cohort. We conclude that suppression of the BALcAMP gene module identified from SARP BAL samples is validated in the IMSA patient cohort with physiological parallels observed in a monocytic cell line and in a severe ACOS patient sample with effects preferentially localizing to macrophages.


Asunto(s)
Agonistas de Receptores Adrenérgicos beta 2/farmacología , Síndrome de Superposición de la Enfermedad Pulmonar Obstructiva Crónica-Asmática/tratamiento farmacológico , Síndrome de Superposición de la Enfermedad Pulmonar Obstructiva Crónica-Asmática/patología , Broncodilatadores/farmacología , AMP Cíclico/biosíntesis , Macrófagos Alveolares/inmunología , Líquido del Lavado Bronquioalveolar/química , Línea Celular , AMP Cíclico/genética , Expresión Génica/genética , Regulación de la Expresión Génica/genética , Humanos , Células Asesinas Naturales/inmunología , Pulmón/patología , Macrófagos Alveolares/metabolismo , Análisis de la Célula Individual , Células THP-1
3.
Am J Respir Crit Care Med ; 200(7): 837-856, 2019 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-31161938

RESUMEN

Rationale: Gene expression of BAL cells, which samples the cellular milieu within the lower respiratory tract, has not been well studied in severe asthma.Objectives: To identify new biomolecular mechanisms underlying severe asthma by an unbiased, detailed interrogation of global gene expression.Methods: BAL cell expression was profiled in 154 asthma and control subjects. Of these participants, 100 had accompanying airway epithelial cell gene expression. BAL cell expression profiles were related to participant (age, sex, race, and medication) and sample traits (cell proportions), and then severity-related gene expression determined by correlating transcripts and coexpression networks to lung function, emergency department visits or hospitalizations in the last year, medication use, and quality-of-life scores.Measurements and Main Results: Age, sex, race, cell proportions, and medications strongly influenced BAL cell gene expression, but leading severity-related genes could be determined by carefully identifying and accounting for these influences. A BAL cell expression network enriched for cAMP signaling components most differentiated subjects with severe asthma from other subjects. Subsequently, an in vitro cellular model showed this phenomenon was likely caused by a robust upregulation in cAMP-related expression in nonsevere and ß-agonist-naive subjects given a ß-agonist before cell collection. Interestingly, ELISAs performed on BAL lysates showed protein levels may partly disagree with expression changes.Conclusions: Gene expression in BAL cells is influenced by factors seldomly considered. Notably, ß-agonist exposure likely had a strong and immediate impact on cellular gene expression, which may not translate to important disease mechanisms or necessarily match protein levels. Leading severity-related genes were discovered in an unbiased, system-wide analysis, revealing new targets that map to asthma susceptibility loci.


Asunto(s)
Asma/genética , Líquido del Lavado Bronquioalveolar/citología , Expresión Génica/genética , Agonistas Adrenérgicos beta/farmacología , Adulto , Asma/metabolismo , Estudios de Casos y Controles , AMP Cíclico/metabolismo , Eosinófilos/metabolismo , Células Epiteliales/metabolismo , Femenino , Expresión Génica/efectos de los fármacos , Humanos , Técnicas In Vitro , Linfocitos/metabolismo , Macrófagos Alveolares/metabolismo , Masculino , Neutrófilos/metabolismo , Análisis de Secuencia de ARN , Índice de Severidad de la Enfermedad , Transducción de Señal/genética , Células THP-1/metabolismo
4.
J Biol Chem ; 293(41): 15790-15800, 2018 10 12.
Artículo en Inglés | MEDLINE | ID: mdl-30093408

RESUMEN

Interleukin (IL)-1ß plays a critical role in IL-6ß- and transforming growth factor ß (TGFß)-initiated Th17 differentiation and induction of Th17-mediated autoimmunity. However, the means by which IL-1 regulates various aspects of Th17 development remain poorly understood. We recently reported that IL-1ß enhances STAT3 phosphorylation via NF-κB-mediated repression of SOCS3 to facilitate Il17 transcription and Th17 differentiation, identifying an effect of IL-1 signaling on proximal events of STAT3 signaling. Here, we show that IL-1ß promotes STAT3 binding to key cis-elements that control IL-17 expression. Additionally, we demonstrate that the IL-1-induced NF-κB factor RelA directly regulates the Il17a/f loci in cooperation with STAT3. Our findings reveal that IL-1 impacts both proximal signaling events and downstream interactions between transcription factors and cis-regulatory elements to promote Il17a/f transcription and Th17 differentiation.


Asunto(s)
Interleucina-17/metabolismo , Receptores Tipo II de Interleucina-1/metabolismo , Factor de Transcripción STAT3/metabolismo , Factor de Transcripción ReIA/metabolismo , Animales , ADN/química , ADN/genética , Interleucina-17/genética , Ratones Endogámicos C57BL , Secuencias Reguladoras de Ácidos Nucleicos/genética , Factor de Transcripción STAT3/genética , Transducción de Señal/fisiología , Células Th17 , Factor de Transcripción ReIA/genética , Activación Transcripcional
5.
Crit Care Med ; 47(12): 1724-1734, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31634231

RESUMEN

OBJECTIVES: Classification of patients with acute respiratory distress syndrome into hyper- and hypoinflammatory subphenotypes using plasma biomarkers may facilitate more effective targeted therapy. We examined whether established subphenotypes are present not only in patients with acute respiratory distress syndrome but also in patients at risk for acute respiratory distress syndrome (ARFA) and then assessed the prognostic information of baseline subphenotyping on the evolution of host-response biomarkers and clinical outcomes. DESIGN: Prospective, observational cohort study. SETTING: Medical ICU at a tertiary academic medical center. PATIENTS: Mechanically ventilated patients with acute respiratory distress syndrome or ARFA. INTERVENTIONS: None. MEASUREMENTS AND MAIN RESULTS: We performed longitudinal measurements of 10 plasma biomarkers of host injury and inflammation. We applied unsupervised latent class analysis methods utilizing baseline clinical and biomarker variables and demonstrated that two-class models (hyper- vs hypoinflammatory subphenotypes) offered improved fit compared with one-class models in both patients with acute respiratory distress syndrome and ARFA. Baseline assignment to the hyperinflammatory subphenotype (39/104 [38%] acute respiratory distress syndrome and 30/108 [28%] ARFA patients) was associated with higher severity of illness by Sequential Organ Failure Assessment scores and incidence of acute kidney injury in patients with acute respiratory distress syndrome, as well as higher 30-day mortality and longer duration of mechanical ventilation in ARFA patients (p < 0.0001). Hyperinflammatory patients exhibited persistent elevation of biomarkers of innate immunity for up to 2 weeks postintubation. CONCLUSIONS: Our results suggest that two distinct subphenotypes are present not only in patients with established acute respiratory distress syndrome but also in patients at risk for its development. Hyperinflammatory classification at baseline is associated with higher severity of illness, worse clinical outcomes, and trajectories of persistently elevated biomarkers of host injury and inflammation during acute critical illness compared with hypoinflammatory patients. Our findings provide strong rationale for examining treatment effect modifications by subphenotypes in randomized clinical trials to inform precision therapeutic approaches in critical care.


Asunto(s)
Síndrome de Dificultad Respiratoria/sangre , Síndrome de Dificultad Respiratoria/complicaciones , Adulto , Anciano , Biomarcadores/sangre , Femenino , Humanos , Inflamación/sangre , Inflamación/complicaciones , Masculino , Persona de Mediana Edad , Fenotipo , Pronóstico , Estudios Prospectivos , Síndrome de Dificultad Respiratoria/clasificación , Síndrome de Dificultad Respiratoria/genética , Medición de Riesgo
6.
FASEB J ; 32(1): 230-242, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28874458

RESUMEN

The tumor necrosis factor receptor-associated factor 2 (TRAF2) is a second messenger adaptor protein that plays an essential role in propagating TNF-α-mediated signaling pathways. Modulation of TRAF2 activity by ubiquitination is well studied; however, the deubiquitinating enzyme (DUB), which regulates TRAF2 stability, has not been identified. Here we reveal USP48 as the first identified DUB to deubiquitinate and stabilize TRAF2 in epithelial cells. Down-regulation of USP48 increases K48-linked polyubiquitination of TRAF2 and reduces TRAF2 protein levels. Interestingly, USP48 only targets the TRAF2 related to JNK pathway, not the TRAF2 related to NF-κB and p38 pathways. USP48 is serine phosphorylated in response to TNF-α. The phosphorylation is catalyzed by glycogen synthase kinase 3ß (GSK3ß), ultimately resulting in increases in USP48 DUB activity. Furthermore, we reveal a new biologic function of TRAF2 that contributes to epithelial barrier dysfunction, which is attenuated by knockdown of USP48. Inhibition of TRAF2/JNK pathway increases E (epithelial)-cadherin expression and enhances epithelial barrier integrity, while knockdown of USP48 attenuates TNF-α/JNK pathway and increases E-cadherin expression and cell-cell junction in epithelial cells. These data, taken together, indicate that USP48 stabilizes TRAF2, which is promoted by GSK3ß-mediated phosphorylation. Further, down-regulation of USP48 increases E-cadherin expression and epithelial barrier integrity through reducing TRAF2 stability.-Li, S., Wang, D., Zhao, J., Weathington, N. M., Shang, D., Zhao, Y. The deubiquitinating enzyme USP48 stabilizes TRAF2 and reduces E-cadherin-mediated adherens junctions.


Asunto(s)
Uniones Adherentes/metabolismo , Cadherinas/metabolismo , Factor 2 Asociado a Receptor de TNF/metabolismo , Proteasas Ubiquitina-Específicas/metabolismo , Células A549 , Animales , Antígenos CD , Línea Celular , Células Epiteliales/metabolismo , Técnicas de Silenciamiento del Gen , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Células HEK293 , Humanos , Sistema de Señalización de MAP Quinasas , Ratones , Fosforilación , Estabilidad Proteica , Factor 2 Asociado a Receptor de TNF/química , Factor de Necrosis Tumoral alfa/metabolismo , Proteasas Ubiquitina-Específicas/antagonistas & inhibidores , Proteasas Ubiquitina-Específicas/genética , Ubiquitinación
7.
Biochem J ; 474(20): 3543-3557, 2017 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-28883123

RESUMEN

The IFN gamma receptor 1 (IFNGR1) binds IFN-γ and activates gene transcription pathways crucial for controlling bacterial and viral infections. Although decreases in IFNGR1 surface levels have been demonstrated to inhibit IFN-γ signaling, little is known regarding the molecular mechanisms controlling receptor stability. Here, we show in epithelial and monocytic cell lines that IFNGR1 displays K48 polyubiquitination, is proteasomally degraded, and harbors three ubiquitin acceptor sites at K277, K279, and K285. Inhibition of glycogen synthase kinase 3 beta (GSK3ß) destabilized IFNGR1 while overexpression of GSK3ß increased receptor stability. We identified critical serine and threonine residues juxtaposed to ubiquitin acceptor sites that impacted IFNGR1 stability. In CRISPR-Cas9 IFNGR1 generated knockout cell lines, cellular expression of IFNGR1 plasmids encoding ubiquitin acceptor site mutations demonstrated significantly impaired STAT1 phosphorylation and decreased STAT1-dependent gene induction. Thus, IFNGR1 undergoes rapid site-specific polyubiquitination, a process modulated by GSK3ß. Ubiquitination appears to be necessary for efficient IFNGR1-dependent gamma gene induction and represents a relatively uncharacterized regulatory mechanism for this receptor.


Asunto(s)
Procesamiento Proteico-Postraduccional/fisiología , Receptores de Interferón/genética , Receptores de Interferón/metabolismo , Transducción de Señal/fisiología , Sistemas CRISPR-Cas/genética , Células HEK293 , Humanos , Interferón gamma/farmacología , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Estabilidad Proteica/efectos de los fármacos , Receptores de Interferón/química , Transducción de Señal/efectos de los fármacos , Receptor de Interferón gamma
8.
Am J Respir Cell Mol Biol ; 57(3): 346-354, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28421819

RESUMEN

IL-25 and IL-4 signaling in the setting of infection or allergic responses can drive Type 2 inflammation. IL-25 requires the IL-17 receptor B (IL-17Rb) to mediate signaling through nuclear factor κ B (NF-κB) transcriptional activation. Despite the known coexistence of these two cytokines in the Type 2 inflammatory environment, collaborative signaling between the IL-4 and IL-25 axes is poorly explored. Here we demonstrate IL-4 induction of both IL-25 and IL-17Rb protein in human lung tissue culture, primary alveolar macrophages, and the THP-1 monocytic cell line. IL-4 treatment triggers gene transcription for both IL-25 and IL-17Rb but does not alter the receptor mRNA stability. Genetic antagonism of the IL-4 second messenger, signal transducer and activator of transcription 6 (STAT6), with small interfering RNA (siRNA) blunts IL-17Rb mRNA induction by IL-4. IL-25 induces signaling through the canonical NF-κB pathway, and STAT6 or NF-κB signaling inhibitors prevent IL-17Rb expression. Blockade of IL-25 with monoclonal antibody suppresses NF-κB activation after IL-4 treatment, and IL-4-mediated induction of IL-17Rb is suppressed by IL-25 siRNA. IL-25 and IL-17Rb promoter regions harbor putative NF-κB and STAT6 consensus sites, and chromatin immunoprecipitation identified these transcription factors in complex with the IL-17Rb 5' untranslated region. In bronchoalveolar lavage RNA preparations, IL-25 and IL-17Rb mRNA transcripts are increased in asthmatics compared with healthy control subjects, and IL-25 transcript abundance correlates strongly with IL-4 mRNA levels. Thus, these results indicate that IL-4 signaling up-regulates the IL-25 axis in human monocytic cells, and that IL-25 may provide autocrine signals in monocytes and macrophages to sustain IL-17Rb expression and predispose to alternative activation.


Asunto(s)
Comunicación Autocrina/genética , Interleucina-17/metabolismo , Interleucina-4/metabolismo , Monocitos/metabolismo , Receptores de Interleucina-17/genética , Transcripción Genética , Asma/genética , Asma/patología , Secuencia de Bases , Línea Celular , Humanos , Macrófagos Alveolares/metabolismo , FN-kappa B/metabolismo , Regiones Promotoras Genéticas/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de Interleucina-17/metabolismo , Factor de Transcripción STAT6/metabolismo
9.
Respir Res ; 18(1): 131, 2017 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-28659178

RESUMEN

BACKGROUND: Azithromycin, an antibiotic used for multiple infectious disorders, exhibits anti-inflammatory effects, but the molecular basis for this activity is not well characterized. Azithromycin inhibits IL-1ß-mediated inflammation that is dependent, in part, on inflammasome activity. Here, we investigated the effects of azithromycin on the NACHT, LRR, and PYD domains-containing protein 3 (NALP3) protein, which is the sensing component of the NALP3 inflammasome, in human monocytes. METHODS: THP-1 cells were treated with azithromycin alone, LPS alone, or both. NALP3 and IL-1ß protein levels were determined by immunoblotting. NLRP3 gene (encoding NALP3) transcript levels were determined by quantitative qPCR. In order to measure NLRP3 transcript decay, actinomycin D was used to impair gene transcription. THP-1 Lucia cells which contain an NF-κB responsive luciferase element were used to assess NF-κB activity in response to azithromycin, LPS, and azithromycin/LPS by measuring luminescence. To confirm azithromycin's effects on NLRP3 mRNA and promoter activity conclusively, HEK cells were lipofected with luciferase reporter constructs harboring either the 5' untranslated region (UTR) of the NLRP3 gene which included the promoter, the 3' UTR of the gene, or an empty plasmid prior to treatment with azithromycin and/or LPS, and luminescence was measured. RESULTS: Azithromycin decreased IL-1ß levels and reduced NALP3 protein levels in LPS-stimulated THP-1 monocytes through a mechanism involving decreased mRNA stability of the NALP3 - coding NLRP3 gene transcript as well as by decreasing NF-κB activity. Azithromycin accelerated NLRP3 transcript decay confirmed by mRNA stability and 3'UTR luciferase reporter assays, and yet the antibiotic had no effect on NLRP3 promoter activity in cells containing a 5' UTR reporter. CONCLUSIONS: These studies provide a unique mechanism whereby azithromycin exerts immunomodulatory actions in monocytes by destabilizing mRNA levels for a key inflammasome component, NALP3, leading to decreased IL-1ß-mediated inflammation.


Asunto(s)
Azitromicina/farmacología , Inflamasomas/metabolismo , Monocitos/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , ARN Mensajero/metabolismo , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Azitromicina/uso terapéutico , Línea Celular , Células HEK293 , Humanos , Inflamasomas/antagonistas & inhibidores , Inflamasomas/genética , Inflamación/tratamiento farmacológico , Inflamación/genética , Inflamación/metabolismo , Monocitos/efectos de los fármacos , FN-kappa B/antagonistas & inhibidores , FN-kappa B/genética , FN-kappa B/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/antagonistas & inhibidores , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Estabilidad Proteica/efectos de los fármacos , ARN Mensajero/antagonistas & inhibidores , ARN Mensajero/genética
10.
Mar Drugs ; 14(7)2016 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-27455285

RESUMEN

Long-term cigarette smoking increases the risk for chronic obstructive pulmonary disease (COPD), characterized by irreversible expiratory airflow limitation. The pathogenesis of COPD involves oxidative stress and chronic inflammation. Various natural marine compounds possess both anti-oxidant and anti-inflammatory properties, but few have been tested for their efficacy in COPD models. In this study, we conducted an in vitro screening test to identify natural compounds isolated from various brown algae species that might provide protection against cigarette smoke extract (CSE)-induced cytotoxicity. Among nine selected natural compounds, apo-9'-fucoxanthinone (Apo9F) exhibited the highest protection against CSE-induced cytotoxicity in immortalized human bronchial epithelial cells (HBEC2). Furthermore, the protective effects of Apo9F were observed to be associated with a significant reduction in apoptotic cell death, DNA damage, and the levels of mitochondrial reactive oxygen species (ROS) released from CSE-exposed HBEC2 cells. These results suggest that Apo9F protects against CSE-induced DNA damage and apoptosis by regulating mitochondrial ROS production.


Asunto(s)
Apoptosis/efectos de los fármacos , Células Epiteliales/efectos de los fármacos , Nicotiana/efectos adversos , Estrés Oxidativo/efectos de los fármacos , Rhodophyta/química , Humo/efectos adversos , Terpenos/farmacología , Línea Celular , Daño del ADN/efectos de los fármacos , Células Epiteliales/metabolismo , Humanos , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Sustancias Protectoras/química , Sustancias Protectoras/farmacología , Enfermedad Pulmonar Obstructiva Crónica/etiología , Enfermedad Pulmonar Obstructiva Crónica/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Fumar/efectos adversos , Terpenos/química
11.
Am J Respir Cell Mol Biol ; 52(5): 622-33, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25286244

RESUMEN

Acute cellular rejection is a known risk factor for the development of obliterative bronchiolitis, which limits the long-term survival of lung transplant recipients. However, the T cell effector mechanisms in both of these processes remain incompletely understood. Using the mouse orthotopic lung transplant model, we investigated whether C57BL/6 T-bet(-/-) recipients of major histocompatibility complex (MHC)-mismatched BALB/c lung grafts develop rejection pathology and allospecific cytokine responses that differ from wild-type mice. T-bet(-/-) recipients demonstrated vigorous allograft rejection at 10 days, characterized by neutrophilic inflammation and predominantly CD8(+) T cells producing allospecific IL-17 and/or IFN-γ, in contrast to IFN-γ-dominant responses in WT mice. CD4(+) T cells produced IL-17 but not IFN-γ responses in T-bet(-/-) recipients, in contrast to WT controls. Costimulation blockade using anti-CD154 Ab significantly reduced allospecific CD8(+)IFN-γ(+) responses in both T-bet(-/-) and WT mice but had no attenuating effect on lung rejection pathology in T-bet(-/-) recipients or on the development of obliterative airway inflammation that occurred only in T-bet(-/-) recipients. However, neutralization of IL-17A significantly attenuated costimulation blockade-resistant rejection pathology and airway inflammation in T-bet(-/-) recipients. In addition, CXCL1 (neutrophil chemokine) was increased in T-bet(-/-) allografts, and IL-17 induced CXCL1 from mouse lung epithelial cells in vitro. Taken together, our data show that T-bet-deficient recipients of complete MHC-mismatched lung allografts develop costimulation blockade-resistant rejection characterized by neutrophilia and obliterative airway inflammation that is predominantly mediated by CD8(+)IL-17(+) T cells. Our data support T-bet-deficient mouse recipients of lung allografts as a viable animal model to study the immunopathogenesis of small airway injury in lung transplantation.


Asunto(s)
Linfocitos T CD8-positivos/metabolismo , Rechazo de Injerto/etiología , Mediadores de Inflamación/metabolismo , Interleucina-17/metabolismo , Trasplante de Pulmón/efectos adversos , Pulmón/metabolismo , Neutrófilos/metabolismo , Neumonía/etiología , Proteínas de Dominio T Box/metabolismo , Enfermedad Aguda , Aloinjertos , Animales , Anticuerpos/farmacología , Ligando de CD40/inmunología , Ligando de CD40/metabolismo , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología , Quimiotaxis de Leucocito , Modelos Animales de Enfermedad , Rechazo de Injerto/inmunología , Rechazo de Injerto/metabolismo , Rechazo de Injerto/patología , Rechazo de Injerto/prevención & control , Histocompatibilidad , Mediadores de Inflamación/inmunología , Interferón gamma/inmunología , Interferón gamma/metabolismo , Interleucina-17/inmunología , Pulmón/efectos de los fármacos , Pulmón/inmunología , Pulmón/patología , Ratones de la Cepa 129 , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Neutrófilos/inmunología , Neumonía/inmunología , Neumonía/metabolismo , Neumonía/patología , Neumonía/prevención & control , Proteínas de Dominio T Box/deficiencia , Proteínas de Dominio T Box/genética
12.
J Biol Chem ; 289(25): 17610-9, 2014 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-24742671

RESUMEN

Signaling through the interleukin (IL)-22 cytokine axis provides essential immune protection in the setting of extracellular infection as part of type 17 immunity. Molecular regulation of IL-22 receptor (IL-22R) protein levels is unknown. In murine lung epithelia, IL-22R is a relatively short-lived protein (t½ ∼1.5 h) degraded by the ubiquitin proteasome under normal unstimulated conditions, but its degradation is accelerated by IL-22 treatment. Lys(449) within the intracellular C-terminal domain of the IL-22R serves as a ubiquitin acceptor site as disruption of this site by deletion or site-directed mutagenesis creates an IL-22R variant that, when expressed in cells, is degradation-resistant and not ubiquitinated. Glycogen synthase kinase (GSK)-3ß phosphorylates the IL-22R within a consensus phosphorylation signature at Ser(410) and Ser(414), and IL-22 treatment of cells triggers GSK-3ß inactivation. GSK-3ß overexpression results in accumulation of IL-22R protein, whereas GSK-3ß depletion in cells reduces levels of the receptor. Mutagenesis of IL-22R at Ser(410) and Ser(414) results in receptor variants that display reduced phosphorylation levels and are more labile as compared with wild-type IL-22R when expressed in cells. Further, the cytoskeletal protein cortactin, which is important for epithelial spreading and barrier formation, is phosphorylated and activated at the epithelial cell leading edge after treatment with IL-22, but this effect is reduced after GSK-3ß knockdown. These findings reveal the ability of GSK-3ß to modulate IL-22R protein stability that might have significant implications for cytoprotective functions and therapeutic targeting of the IL-22 signaling axis.


Asunto(s)
Glucógeno Sintasa Quinasa 3/metabolismo , Pulmón/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteolisis , Receptores de Interleucina/metabolismo , Mucosa Respiratoria/metabolismo , Animales , Línea Celular , Células Epiteliales/citología , Células Epiteliales/metabolismo , Técnicas de Silenciamiento del Gen , Glucógeno Sintasa Quinasa 3/genética , Glucógeno Sintasa Quinasa 3 beta , Pulmón/citología , Ratones , Mutagénesis , Fosforilación/fisiología , Complejo de la Endopetidasa Proteasomal/genética , Estabilidad Proteica , Receptores de Interleucina/genética , Mucosa Respiratoria/citología , Transducción de Señal/fisiología , Ubiquitina/genética , Ubiquitina/metabolismo
13.
J Immunol ; 191(10): 5247-55, 2013 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-24123678

RESUMEN

Cytokine-driven inflammation underlies the pathobiology of a wide array of infectious and immune-related disorders. The TNFR-associated factor (TRAF) proteins have a vital role in innate immunity by conveying signals from cell surface receptors to elicit transcriptional activation of genes encoding proinflammatory cytokines. We discovered that a ubiquitin E3 ligase F box component, termed Fbxo3, potently stimulates cytokine secretion from human inflammatory cells by mediating the degradation of the TRAF inhibitory protein, Fbxl2. Analysis of the Fbxo3 C-terminal structure revealed that the bacterial-like ApaG molecular signature was indispensible for mediating Fbxl2 disposal and stimulating cytokine secretion. By targeting this ApaG motif, we developed a highly unique, selective genus of small-molecule Fbxo3 inhibitors that by reducing TRAF protein levels, potently inhibited cytokine release from human blood mononuclear cells. The Fbxo3 inhibitors effectively lessened the severity of viral pneumonia, septic shock, colitis, and cytokine-driven inflammation systemically in murine models. Thus, pharmacological targeting of Fbxo3 might be a promising strategy for immune-related disorders characterized by a heightened host inflammatory response.


Asunto(s)
Bencilaminas/uso terapéutico , Proteínas F-Box/metabolismo , Inflamación/tratamiento farmacológico , Piridinas/uso terapéutico , Péptidos y Proteínas Asociados a Receptores de Factores de Necrosis Tumoral/metabolismo , Animales , Bencilaminas/farmacología , Línea Celular , Colitis/tratamiento farmacológico , Citocinas/biosíntesis , Citocinas/metabolismo , Proteínas F-Box/antagonistas & inhibidores , Inflamación/inmunología , Subtipo H1N1 del Virus de la Influenza A/efectos de los fármacos , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/inmunología , Lesión Pulmonar/tratamiento farmacológico , Lesión Pulmonar/virología , Masculino , Ratones , Ratones Endogámicos C57BL , Infecciones por Orthomyxoviridae/tratamiento farmacológico , Piridinas/farmacología , Péptidos y Proteínas Asociados a Receptores de Factores de Necrosis Tumoral/antagonistas & inhibidores , Ubiquitina-Proteína Ligasas/metabolismo
14.
Am J Physiol Cell Physiol ; 307(9): C893-9, 2014 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-25163517

RESUMEN

E-cadherin is essential for the integrity of adherens junctions between lung epithelial cells, and the loss of E-cadherin allows cell motility and is thought to promote lung cancer metastasis. While the downregulation of E-cadherin expression has been well characterized and is seen with transforming growth factor-ß1 (TGF-ß1) exposure, few studies have focused on E-cadherin upregulation. Here, we show that serum starvation causes increased E-cadherin expression via the activation of c-Src kinase in non-small-cell lung cancer A549 cells. Serum starvation increased E-cadherin protein levels in a time- and dose-dependent manner. E-cadherin mRNA transcripts were unchanged with starvation, while protein translation inhibition with cycloheximide attenuated E-cadherin protein induction by starvation, suggesting that E-cadherin is regulated at the translational level by serum starvation. c-Src is a nonreceptor tyrosine kinase known to regulate protein translation machinery; serum starvation caused early and sustained activation of c-Src in A549 cells followed by E-cadherin upregulation. Furthermore, overexpression of a dominant negative c-Src attenuated the induction of E-cadherin by serum deprivation. Finally, we observed that TGF-ß1 treatment attenuated the serum activation of c-Src as well as E-cadherin expression when cells were deprived of serum. In conclusion, our data demonstrate that the c-Src kinase is activated by serum starvation to increase E-cadherin expression in A549 cells, and these phenomena are antagonized by TGF-ß1. These novel observations implicate the c-Src kinase as an upstream inducer of E-cadherin protein translation with serum starvation and TGF-ß1 diametrically regulating c-Src kinase activity and thus E-cadherin abundance in A549 cells.


Asunto(s)
Cadherinas/biosíntesis , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Neoplasias Pulmonares/metabolismo , Proteínas Proto-Oncogénicas pp60(c-src)/metabolismo , Cadherinas/genética , Carcinoma de Pulmón de Células no Pequeñas/enzimología , Carcinoma de Pulmón de Células no Pequeñas/genética , Línea Celular Tumoral , Células Cultivadas , Medio de Cultivo Libre de Suero , Activación Enzimática , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Pulmonares/enzimología , Neoplasias Pulmonares/genética , Biosíntesis de Proteínas , Factor de Crecimiento Transformador beta1/farmacología , Regulación hacia Arriba/efectos de los fármacos
15.
Am J Respir Crit Care Med ; 188(5): 530-7, 2013 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-23713962

RESUMEN

Derangements in normal cellular homeostasis at the protein level can cause or be the consequence of initiation and progression of pulmonary diseases related to genotype, infection, injury, smoking, toxin exposure, or neoplasm. We discuss one of the fundamental mechanisms of protein homeostasis, the ubiquitin proteasome system (UPS), as it relates to lung disease. The UPS effects selective degradation of ubiquitinated target proteins via ubiquitin ligase activity. Important pathobiological mechanisms relating to the UPS and lung disease have been the focus of research, with inappropriate cellular proteolysis now a validated therapeutic target. We review the contributions of this system in various lung diseases, and discuss the exciting area of UPS-targeting drug development for pulmonary disease.


Asunto(s)
Enfermedades Pulmonares/fisiopatología , Pulmón/fisiología , Complejo de la Endopetidasa Proteasomal/fisiología , Ubiquitina/fisiología , Proteínas F-Box/fisiología , Humanos , Pulmón/metabolismo , Enfermedades Pulmonares/tratamiento farmacológico , Enfermedades Pulmonares/metabolismo , Complejo de la Endopetidasa Proteasomal/efectos de los fármacos , Complejo de la Endopetidasa Proteasomal/metabolismo , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitina-Proteína Ligasas/fisiología
16.
Nat Med ; 12(3): 317-23, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16474398

RESUMEN

We describe the tripeptide neutrophil chemoattractant N-acetyl Pro-Gly-Pro (PGP), derived from the breakdown of extracellular matrix (ECM), which shares sequence and structural homology with an important domain on alpha chemokines. PGP caused chemotaxis and production of superoxide through CXC receptors, and administration of peptide caused recruitment of neutrophils (PMNs) into lungs of control, but not CXCR2-deficient mice. PGP was generated in mouse lung after exposure to lipopolysaccharide, and in vivo and in vitro blockade of PGP with monoclonal antibody suppressed PMN responses as much as chemokine-specific monoclonal antibody. Extended PGP treatment caused alveolar enlargement and right ventricular hypertrophy in mice. PGP was detectable in substantial concentrations in a majority of bronchoalveolar lavage samples from individuals with chronic obstructive pulmonary disease, but not control individuals. Thus, PGP's activity links degradation of ECM with neutrophil recruitment in airway inflammation, and PGP may be a biomarker and therapeutic target for neutrophilic inflammatory diseases.


Asunto(s)
Matriz Extracelular/metabolismo , Pulmón/metabolismo , Pulmón/patología , Oligopéptidos/metabolismo , Receptores de Quimiocina/metabolismo , Animales , Lavado Broncoalveolar , Quimiotaxis de Leucocito/inmunología , Femenino , Células HL-60 , Humanos , Inflamación/metabolismo , Inflamación/patología , Ligandos , Lipopolisacáridos/inmunología , Lipopolisacáridos/farmacología , Pulmón/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Neutrófilos/efectos de los fármacos , Neutrófilos/metabolismo , Enfermedad Pulmonar Obstructiva Crónica/patología , Receptores de Interleucina-8A/metabolismo , Receptores de Interleucina-8B/metabolismo , Homología Estructural de Proteína
17.
Mol Med ; 16(5-6): 159-66, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20111696

RESUMEN

Cystic fibrosis (CF) is a lethal genetic disorder characterized by airway remodeling and inflammation, leading to premature death. Recent evidence suggests the importance of protease activity in CF pathogenesis. One prominent protease, matrix metalloprotease (MMP)-9, demonstrates increased activity in CF individuals undergoing acute pulmonary exacerbation. This is thought to be mediated by both direct MMP-9 activation and the degradation of its natural inhibitor, tissue inhibitor of metalloprotease-1 (TIMP-1). To examine if this relationship exists in nonexacerbating CF individuals, we examined protease activity in sputum from these individuals compared with nondisease controls. We demonstrated increased gelatinolytic activity in CF sputum. These samples had elevated human neutrophil elastase (HNE) levels which correlated with an increased MMP-9/TIMP-1 ratio. To determine if HNE could discretely cleave and activate MMP-9, these enzymes were coincubated and two specific cleavage sites, between Valine(38) and Alanine(39), and between Alanine (39) and glutamic acid(40) were observed. These sites corresponded with appropriate molecular weight for the activated MMP-9 isoform in CF sputum. Using N-terminal sequencing of cleavage fragments obtained with TIMP-1 incubation with HNE, we confirmed the TIMP-1 cleavage site for HNE is at Valine(69)-Cysteine(70). We also show for the first time that human neutrophils were capable of degrading TIMP-1 ex vivo and that a 16 kDa TIMP-1 fragment was identified in CF sputum, consistent with the expected cleavage of TIMP-1 by HNE. These results demonstrate increased MMP-9 activity in stable CF lung disease, and the presence of specific protease products in CF sputum highlights that HNE-mediated activity plays a role in this dysregulation.


Asunto(s)
Fibrosis Quística/metabolismo , Elastasa de Leucocito/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Inhibidor Tisular de Metaloproteinasa-1/metabolismo , Adulto , Secuencia de Aminoácidos , Western Blotting , Estudios de Casos y Controles , Fibrosis Quística/enzimología , Electroforesis en Gel de Poliacrilamida , Femenino , Humanos , Masculino , Modelos Moleculares , Datos de Secuencia Molecular , Colorantes de Rosanilina , Transducción de Señal , Esputo/enzimología , Esputo/metabolismo , Estadísticas no Paramétricas
18.
Pharmacol Ther ; 121(2): 132-46, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19026684

RESUMEN

Protease activity in inflammation is complex. Proteases released by cells in response to infection, cytokines, or environmental triggers like cigarette smoking cause breakdown of the extracellular matrix (ECM). In chronic inflammatory diseases like chronic obstructive pulmonary disease (COPD), current findings indicate that pathology and morbidity are driven by dysregulation of protease activity, either through hyperactivity of proteases or deficiency or dysfunction their antiprotease regulators. Animal studies demonstrate the accuracy of this hypothesis through genetic and pharmacologic tools. New work shows that ECM destruction generates peptide fragments active on leukocytes via neutrophil or macrophage chemotaxis towards collagen and elastin derived peptides respectively. Such fragments now have been isolated and characterized in vivo in each case. Collectively, this describes a biochemical circuit in which protease activity leads to activation of local immunocytes, which in turn release cytokines and more proteases, leading to further leukocyte infiltration and cyclical disease progression that is chronic. This circuit concept is well known, and is intrinsic to the protease-antiprotease hypothesis; recently analytic techniques have become sensitive enough to establish fundamental mechanisms of this hypothesis, and basic and clinical data now implicate protease activity and peptide signaling as pathologically significant pharmacologic targets. This review discusses targeting protease activity for chronic inflammatory disease with special attention to COPD, covering important basic and clinical findings in the field; novel therapeutic strategies in animal or human studies; and a perspective on the successes and failures of agents with a focus on clinical potential in human disease.


Asunto(s)
Péptido Hidrolasas/efectos de los fármacos , Inhibidores de Proteasas/farmacología , Enfermedad Pulmonar Obstructiva Crónica/tratamiento farmacológico , Animales , Quimiotaxis/efectos de los fármacos , Ensayos Clínicos como Asunto , Modelos Animales de Enfermedad , Sistemas de Liberación de Medicamentos , Matriz Extracelular/efectos de los fármacos , Matriz Extracelular/metabolismo , Humanos , Péptido Hidrolasas/metabolismo , Enfermedad Pulmonar Obstructiva Crónica/fisiopatología
19.
Mol Cell Biol ; 40(12)2020 05 28.
Artículo en Inglés | MEDLINE | ID: mdl-32205409

RESUMEN

FBXL2 is an important ubiquitin E3 ligase component that modulates inflammatory signaling and cell cycle progression, but its molecular regulation is largely unknown. Here, we show that tumor necrosis factor alpha (TNF-α), a critical cytokine linked to the inflammatory response during skeletal muscle regeneration, suppressed Fbxl2 mRNA expression in C2C12 myoblasts and triggered significant alterations in cell cycle, metabolic, and protein translation processes. Gene silencing of Fbxl2 in skeletal myoblasts resulted in increased proliferative responses characterized by activation of mitogen-activated protein (MAP) kinases and nuclear factor kappa B and decreased myogenic differentiation, as reflected by reduced expression of myogenin and impaired myotube formation. TNF-α did not destabilize the Fbxl2 transcript (half-life [t1/2], ∼10 h) but inhibited SP1 transactivation of its core promoter, localized to bp -160 to +42 within the proximal 5' flanking region of the Fbxl2 gene. Chromatin immunoprecipitation and gel shift studies indicated that SP1 interacted with the Fbxl2 promoter during cellular differentiation, an effect that was less pronounced during proliferation or after TNF-α exposure. TNF-α, via activation of JNK, mediated phosphorylation of SP1 that impaired its binding to the Fbxl2 promoter, resulting in reduced transcriptional activity. The results suggest that SP1 transcriptional activation of Fbxl2 is required for skeletal muscle differentiation, a process that is interrupted by a key proinflammatory myopathic cytokine.IMPORTANCE Skeletal muscle regeneration and repair involve the recruitment and proliferation of resident satellite cells that exit the cell cycle during the process of myogenic differentiation to form myofibers. We demonstrate that the ubiquitin E3 ligase subunit FBXL2 is essential for skeletal myogenesis through its important effects on cell cycle progression and cell proliferative signaling. Further, we characterize a new mechanism whereby sustained stimulation by a major proinflammatory cytokine, TNF-α, regulates skeletal myogenesis by inhibiting the interaction of SP1 with the Fbxl2 core promoter in proliferating myoblasts. Our findings contribute to the understanding of skeletal muscle regeneration through the identification of Fbxl2 as both a critical regulator of myogenic proliferative processes and a susceptible gene target during inflammatory stimulation by TNF-α in skeletal muscle. Modulation of Fbxl2 activity may have relevance to disorders of muscle wasting associated with sustained proinflammatory signaling.


Asunto(s)
Proteínas F-Box/genética , Desarrollo de Músculos , Músculo Esquelético/fisiología , Factor de Transcripción Sp1/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Células Cultivadas , Humanos , Mioblastos Esqueléticos/citología , Mioblastos Esqueléticos/metabolismo , Regiones Promotoras Genéticas , Secuencias Reguladoras de Ácidos Nucleicos
20.
JCI Insight ; 5(11)2020 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-32493843

RESUMEN

Mitochondrial quality control is mediated by the PTEN-induced kinase 1 (PINK1), a cytoprotective protein that is dysregulated in inflammatory lung injury and neurodegenerative diseases. Here, we show that a ubiquitin E3 ligase receptor component, FBXO7, targets PINK1 for its cellular disposal. FBXO7, by mediating PINK1 ubiquitylation and degradation, was sufficient to induce mitochondrial injury and inflammation in experimental pneumonia. A computational simulation-based screen led to the identification of a small molecule, BC1464, which abrogated FBXO7 and PINK1 association, leading to increased cellular PINK1 concentrations and activities, and limiting mitochondrial damage. BC1464 exerted antiinflammatory activity in human tissue explants and murine lung inflammation models. Furthermore, BC1464 conferred neuroprotection in primary cortical neurons, human neuroblastoma cells, and patient-derived cells in several culture models of Parkinson's disease. The data highlight a unique opportunity to use small molecule antagonists that disrupt PINK1 interaction with the ubiquitin apparatus to enhance mitochondrial quality, limit inflammatory injury, and maintain neuronal viability.


Asunto(s)
Proteínas F-Box/antagonistas & inhibidores , Proteínas Mitocondriales/metabolismo , Fármacos Neuroprotectores/farmacología , Proteínas Quinasas/metabolismo , Proteolisis/efectos de los fármacos , Ubiquitinación/efectos de los fármacos , Animales , Línea Celular Tumoral , Estabilidad de Enzimas , Proteínas F-Box/metabolismo , Humanos , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Inflamación/patología , Ratones , Fármacos Neuroprotectores/química , Enfermedad de Parkinson/tratamiento farmacológico , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/patología , Neumonía/tratamiento farmacológico , Neumonía/metabolismo , Neumonía/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA