Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 210
Filtrar
1.
Toxicol Appl Pharmacol ; 450: 116160, 2022 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-35817128

RESUMEN

Epidemiological studies associate biomass smoke with an increased risk for respiratory infections in children and adults in the developing world, with 500,000 premature deaths each year attributed to biomass smoke-related acute respiratory infections including infections caused by respiratory viruses. Animal dung is a biomass fuel of particular concern because it generates more toxic compounds per amount burned than wood, and is a fuel of last resort for the poorest households. Currently, there is little biological evidence on the effects of dung biomass smoke exposure on immune responses to respiratory viral infections. Here, we investigated the impact of dung biomass exposure on respiratory infection using a mouse model of dung biomass smoke and cultured primary human small airway epithelial cells (SAECs). Mice infected with influenza A virus (IAV) after dung biomass smoke exposure had increased mortality, lung inflammation and virus mRNA levels, and suppressed expression of innate anti-viral mediators compared to air exposed mice. Importantly, there was still significant tissue inflammation 14 days after infection in dung biomass smoke-exposed mice even after inflammation had resolved in air-exposed mice. Dung biomass smoke exposure also suppressed the production of anti-viral cytokines and interferons in cultured SAECs treated with poly(I:C) or IAV. This study shows that dung biomass smoke exposure impairs the immune response to respiratory viruses and contributes to biomass smoke-related susceptibility to respiratory viral infections, likely due to a failure to resolve the inflammatory effects of biomass smoke exposure.


Asunto(s)
Gripe Humana , Neumonía , Infecciones del Sistema Respiratorio , Animales , Biomasa , Niño , Humanos , Inflamación/inducido químicamente , Inflamación/metabolismo
3.
J Pain ; 22(10): 1195-1209, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-33813057

RESUMEN

Localized provoked vulvodynia (LPV) is the most common cause of chronic dyspareunia in premenopausal women, characterized by pain with light touch to the vulvar vestibule surrounding the vaginal opening. The devastating impact of LPV includes sexual dysfunction, infertility, depression, and even suicide. Yet, its etiology is unclear. No effective medical therapy exists; surgical removal of the painful vestibule is the last resort. In LPV, the vestibule expresses a unique inflammatory profile with elevated levels of pro-nociceptive proinflammatory mediators prostaglandin E2 (PGE2) and interleukin-6 (IL-6), which are linked to lower mechanical sensitivity thresholds. Specialized pro-resolving mediators (SPMs), lipids produced endogenously within the body, hold promise as an LPV treatment by resolving inflammation without impairing host defense. Ten of 13 commercially available SPMs reduced IL-6 and PGE2 production by vulvar fibroblasts, administered either before or after inflammatory stimulation. Using a murine vulvar pain model, coupling proinflammatory mediator quantification with mechanical sensitivity threshold determination, topical treatment with the SPM, maresin 1, decreased sensitivity and suppressed PGE2 levels. Docosahexaenoic acid, a precursor of maresin 1, was also effective in reducing PGE2 in vulvar fibroblasts and rapidly restored mouse sensitivity thresholds. Overall, SPMs and their precursors may be a safe and efficacious for LPV. Perspective: Vulvodynia, like many pain conditions, is difficult to treat because disease origins are incompletely understood. Here, we applied our knowledge of more recently discovered vulvodynia disease mechanisms to screen novel therapeutics. We identified several specialized pro-resolving mediators as likely potent and safe for treating LPV with potential for broader application.


Asunto(s)
Dinoprostona , Ácidos Docosahexaenoicos/farmacología , Fibroblastos/efectos de los fármacos , Inflamación/tratamiento farmacológico , Interleucina-6 , Nocicepción/efectos de los fármacos , Vulvodinia/tratamiento farmacológico , Animales , Modelos Animales de Enfermedad , Femenino , Ratones
5.
FASEB J ; 35(3): e21376, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33605487

RESUMEN

Emphysema, a component of chronic obstructive pulmonary disease (COPD), is characterized by irreversible alveolar destruction that results in a progressive decline in lung function. This alveolar destruction is caused by cigarette smoke, the most important risk factor for COPD. Only 15%-20% of smokers develop COPD, suggesting that unknown factors contribute to disease pathogenesis. We postulate that the aryl hydrocarbon receptor (AHR), a receptor/transcription factor highly expressed in the lungs, may be a new susceptibility factor whose expression protects against COPD. Here, we report that Ahr-deficient mice chronically exposed to cigarette smoke develop airspace enlargement concomitant with a decline in lung function. Chronic cigarette smoke exposure also increased cleaved caspase-3, lowered SOD2 expression, and altered MMP9 and TIMP-1 levels in Ahr-deficient mice. We also show that people with COPD have reduced expression of pulmonary and systemic AHR, with systemic AHR mRNA levels positively correlating with lung function. Systemic AHR was also lower in never-smokers with COPD. Thus, AHR expression protects against the development of COPD by controlling interrelated mechanisms involved in the pathogenesis of this disease. This study identifies the AHR as a new, central player in the homeostatic maintenance of lung health, providing a foundation for the AHR as a novel therapeutic target and/or predictive biomarker in chronic lung disease.


Asunto(s)
Enfermedad Pulmonar Obstructiva Crónica/etiología , Receptores de Hidrocarburo de Aril/deficiencia , Anciano , Anciano de 80 o más Años , Animales , Translocador Nuclear del Receptor de Aril Hidrocarburo/fisiología , Enfisema/etiología , Volumen Espiratorio Forzado , Humanos , Pulmón/fisiopatología , Masculino , Ratones , Persona de Mediana Edad , Enfermedad Pulmonar Obstructiva Crónica/fisiopatología , Receptores de Hidrocarburo de Aril/genética , Receptores de Hidrocarburo de Aril/fisiología , Fumar/efectos adversos
6.
Am J Pathol ; 191(1): 18-25, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33031756

RESUMEN

Idiopathic pulmonary fibrosis is a progressive scarring disease characterized by extracellular matrix accumulation and altered mechanical properties of lung tissue. Recent studies support the hypothesis that these compositional and mechanical changes create a progressive feed-forward loop in which enhanced matrix deposition and tissue stiffening contribute to fibroblast and myofibroblast differentiation and activation, which further perpetuates matrix production and stiffening. The biomechanical properties of tissues are sensed and responded to by mechanotransduction pathways that facilitate sensing of changes in mechanical cues by tissue resident cells and convert the mechanical signals into downstream biochemical signals. Although our understanding of mechanotransduction pathways associated with pulmonary fibrosis remains incomplete, recent progress has allowed us to begin to elucidate the specific mechanisms supporting fibrotic feed-forward loops. The mechanosensors discussed here include integrins, Piezo channels, transient receptor potential channels, and nonselective ion channels. Also discussed are downstream transcription factors, including myocardin-related transcription factor and Yes-associated protein/transcriptional coactivator with PDZ-binding motif. This review describes mechanosensors and mechanotransduction pathways associated with fibrosis progression and highlights promising therapeutic insights.


Asunto(s)
Retroalimentación Fisiológica/fisiología , Fibrosis Pulmonar Idiopática/metabolismo , Mecanotransducción Celular/fisiología , Animales , Fibroblastos/metabolismo , Humanos
7.
PLoS One ; 15(5): e0232102, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32437367

RESUMEN

Cigarette smoke exposure is a risk factor for many pulmonary diseases, including Chronic Obstructive Pulmonary Disease (COPD). Cigarette smokers are more prone to respiratory infections with more severe symptoms. In those with COPD, viral infections can lead to acute exacerbations resulting in lung function decline and death. Epithelial cells in the lung are the first line of defense against inhaled insults such as tobacco smoke and are the target for many respiratory pathogens. Endocytosis is an essential cell function involved in nutrient uptake, cell signaling, and sensing of the extracellular environment, yet, the effect of cigarette smoke on epithelial cell endocytosis is not known. Here, we report for the first time that cigarette smoke alters the function of several important endocytic pathways in primary human small airway epithelial cells. Cigarette smoke exposure impairs clathrin-mediated endocytosis and fluid phase macropinocytosis while increasing caveolin mediated endocytosis. We also show that influenza virus uptake is enhanced by cigarette smoke exposure. These results support the concept that cigarette smoke-induced dysregulation of endocytosis contributes to lung infection in smokers. Targeting endocytosis pathways to restore normal epithelial cell function may be a new therapeutic approach to reduce respiratory infections in current and former smokers.


Asunto(s)
Caveolinas/metabolismo , Células Epiteliales/efectos de los fármacos , Infecciones/patología , Pulmón/citología , Nicotiana/química , Humo/efectos adversos , Regulación hacia Arriba/efectos de los fármacos , Susceptibilidad a Enfermedades , Células Epiteliales/metabolismo , Células Epiteliales/virología , Humanos , Infecciones/inducido químicamente , Infecciones/virología , Nicotiana/efectos adversos
8.
Cancer Metastasis Rev ; 39(2): 337-340, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32385712

RESUMEN

Severe coronavirus disease (COVID-19) is characterized by pulmonary hyper-inflammation and potentially life-threatening "cytokine storms". Controlling the local and systemic inflammatory response in COVID-19 may be as important as anti-viral therapies. Endogenous lipid autacoid mediators, referred to as eicosanoids, play a critical role in the induction of inflammation and pro-inflammatory cytokine production. SARS-CoV-2 may trigger a cell death ("debris")-induced "eicosanoid storm", including prostaglandins and leukotrienes, which in turn initiates a robust inflammatory response. A paradigm shift is emerging in our understanding of the resolution of inflammation as an active biochemical process with the discovery of novel endogenous specialized pro-resolving lipid autacoid mediators (SPMs), such as resolvins. Resolvins and other SPMs stimulate macrophage-mediated clearance of debris and counter pro-inflammatory cytokine production, a process called inflammation resolution. SPMs and their lipid precursors exhibit anti-viral activity at nanogram doses in the setting of influenza without being immunosuppressive. SPMs also promote anti-viral B cell antibodies and lymphocyte activity, highlighting their potential use in the treatment of COVID-19. Soluble epoxide hydrolase (sEH) inhibitors stabilize arachidonic acid-derived epoxyeicosatrienoic acids (EETs), which also stimulate inflammation resolution by promoting the production of pro-resolution mediators, activating anti-inflammatory processes, and preventing the cytokine storm. Both resolvins and EETs also attenuate pathological thrombosis and promote clot removal, which is emerging as a key pathology of COVID-19 infection. Thus, both SPMs and sEH inhibitors may promote the resolution of inflammation in COVID-19, thereby reducing acute respiratory distress syndrome (ARDS) and other life-threatening complications associated with robust viral-induced inflammation. While most COVID-19 clinical trials focus on "anti-viral" and "anti-inflammatory" strategies, stimulating inflammation resolution is a novel host-centric therapeutic avenue. Importantly, SPMs and sEH inhibitors are currently in clinical trials for other inflammatory diseases and could be rapidly translated for the management of COVID-19 via debris clearance and inflammatory cytokine suppression. Here, we discuss using pro-resolution mediators as a potential complement to current anti-viral strategies for COVID-19.


Asunto(s)
Antiinflamatorios no Esteroideos/uso terapéutico , Antivirales/uso terapéutico , Betacoronavirus/inmunología , Infecciones por Coronavirus/tratamiento farmacológico , Síndrome de Liberación de Citoquinas/tratamiento farmacológico , Neumonía Viral/tratamiento farmacológico , Síndrome de Dificultad Respiratoria/terapia , Antiinflamatorios no Esteroideos/farmacología , Betacoronavirus/aislamiento & purificación , COVID-19 , Ensayos Clínicos como Asunto , Infecciones por Coronavirus/complicaciones , Infecciones por Coronavirus/inmunología , Infecciones por Coronavirus/virología , Síndrome de Liberación de Citoquinas/inmunología , Citocinas/inmunología , Citocinas/metabolismo , Eicosanoides/inmunología , Eicosanoides/metabolismo , Epóxido Hidrolasas/antagonistas & inhibidores , Epóxido Hidrolasas/metabolismo , Humanos , Macrófagos/inmunología , Macrófagos/metabolismo , Pandemias , Neumonía Viral/complicaciones , Neumonía Viral/inmunología , Neumonía Viral/virología , Alveolos Pulmonares/inmunología , Alveolos Pulmonares/metabolismo , Alveolos Pulmonares/virología , Síndrome de Dificultad Respiratoria/inmunología , SARS-CoV-2
9.
Transfusion ; 60(7): 1579-1589, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32415759

RESUMEN

BACKGROUND: Platelet transfusion is associated with logistical problems with the national storage guidelines of platelets. This results in decreased function in vivo as a result of the platelet storage lesion, and complications such as allergic or hemolytic reactions and thrombosis. We evaluated a new, freshly prepared platelet modified lysate (PML) product designed to be more procoagulant than fresh and stored platelets. METHODS: Fresh platelets were concentrated, sonicated, and centrifuged to produce PML. Samples of both washed and unwashed PML were evaluated for particle size, concentration, and activity, and then tested for clot kinetics and thrombin generation. PML samples were also stored at various temperatures for durations up to 6 months and evaluated for clot kinetics and thrombin generation throughout. RESULTS: PML showed significantly higher concentration of platelet microparticles, increased procoagulant properties, and increased thrombin generation as compared to fresh and stored platelets. In addition, PML maintained its clot kinetics over a 6-month storage period with variable storage conditions. CONCLUSIONS: The newly proposed PML product is more procoagulant, stable, and has additional potential applications than currently available platelet products. Further studies will be performed to assess its functions in vivo and to assess thrombotic potential.


Asunto(s)
Coagulación Sanguínea/efectos de los fármacos , Plaquetas/química , Micropartículas Derivadas de Células/química , Coagulantes , Coagulantes/química , Coagulantes/farmacología , Evaluación Preclínica de Medicamentos , Humanos , Transfusión de Plaquetas
10.
J Occup Environ Med ; 61 Suppl 12: S5-S14, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31800446

RESUMEN

OBJECTIVE: Review advances in exposure assessment offered by the exposome concept and new -omics and sensor technologies. METHODS: Narrative review of advances, including current efforts and potential future applications by the US military. RESULTS: Exposure assessment methods from both bottom-up and top-down exposomics approaches are advancing at a rapid pace, and the US military is engaged in developing both approaches. Top-down approaches employ various -omics technologies to identify biomarkers of internal exposure and biological effect. Bottom-up approaches use new sensor technology to better measure external dose. Key challenges of both approaches are largely centered around how to integrate, analyze, and interpret large datasets that are multidimensional and disparate. CONCLUSIONS: Advances in -omics and sensor technologies may dramatically enhance exposure assessment and improve our ability to characterize health risks related to occupational and environmental exposures, including for the US military.


Asunto(s)
Exposición a Riesgos Ambientales/estadística & datos numéricos , Personal Militar/estadística & datos numéricos , Monitoreo Biológico , Epigenómica , Humanos , Metabolómica , Estados Unidos
11.
J Occup Environ Med ; 61 Suppl 12: S45-S54, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31800450

RESUMEN

OBJECTIVE: The potential health risks of deployment to sites with open burn pits remain poorly understood, in part, because personal exposure monitoring was not performed. Here, we investigated whether postdeployment serum samples contain biomarkers associated with exposure to burn pits. METHODS: A total of 237 biomarkers were measured in 800 serum samples from deployed and never-deployed subjects. We used a regression model and a supervised vector machine to identify serum biomarkers with significant associations with exposures and deployment. RESULTS: We identified 101 serum biomarkers associated with polycyclic aromatic hydrocarbons, dioxins or furans, and 54 biomarkers associated with deployment. Twenty-six of these biomarkers were shared in common by the exposure and deployment groups. CONCLUSIONS: We identify a potential signature of exposure to open burn pits, and provide a framework for using postexposure sera to identify exposures when contemporaneous monitoring was inadequate.


Asunto(s)
Contaminantes Atmosféricos/sangre , Exposición por Inhalación/análisis , Personal Militar/estadística & datos numéricos , Campaña Afgana 2001- , Contaminantes Atmosféricos/efectos adversos , Biomarcadores/sangre , Dioxinas/efectos adversos , Dioxinas/sangre , Femenino , Furanos/efectos adversos , Furanos/sangre , Humanos , Incineración/estadística & datos numéricos , Exposición por Inhalación/efectos adversos , Exposición por Inhalación/normas , Guerra de Irak 2003-2011 , Aprendizaje Automático , Masculino , Espectrometría de Masas , MicroARNs/sangre , Hidrocarburos Policíclicos Aromáticos/efectos adversos , Hidrocarburos Policíclicos Aromáticos/sangre , Estados Unidos , Instalaciones de Eliminación de Residuos/estadística & datos numéricos
12.
J Occup Environ Med ; 61 Suppl 12: S55-S64, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31800451

RESUMEN

OBJECTIVE: To develop an approach for a retrospective analysis of post-exposure serum samples using diverse molecular profiles. METHODS: The 236 molecular profiles from 800 de-identified human serum samples from the Department of Defense Serum Repository were classified as smokers or non-smokers based on direct measurement of serum cotinine levels. A machine-learning pipeline was used to classify smokers and non-smokers from their molecular profiles. RESULTS: The refined supervised support vector machines with recursive feature elimination predicted smokers and non-smokers with 78% accuracy on the independent held-out set. Several of the identified classifiers of smoking status have previously been reported and four additional miRNAs were validated with experimental tobacco smoke exposure in mice, supporting the computational approach. CONCLUSIONS: We developed and validated a pipeline that shows retrospective analysis of post-exposure serum samples can identify environmental exposures.


Asunto(s)
Cotinina/sangre , Exposición a Riesgos Ambientales/estadística & datos numéricos , Aprendizaje Automático , Adolescente , Adulto , Factores de Edad , Animales , Biomarcadores/sangre , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Ratones Endogámicos C57BL , Factores Sexuales , Fumar/efectos adversos , Fumar/epidemiología , Máquina de Vectores de Soporte , Contaminación por Humo de Tabaco/efectos adversos , Contaminación por Humo de Tabaco/estadística & datos numéricos , Adulto Joven
13.
J Occup Environ Med ; 61 Suppl 12: S82-S89, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31800454

RESUMEN

OBJECTIVE: Benzo(ghi)perylene (BghiP) and 1,2,3,4,6,7,8-Heptachlorodibenzo-p-dioxin (HpCDD) were elevated in serum from personnel deployed to sites with open burn pits. Here, we investigated the ability of BghiP and HpCDD to regulate microRNA (miRNA) expression through the aryl hydrocarbon receptor (AHR). METHODS: Human lung fibroblasts (HLFs) were exposed to BghiP and HpCDD. AHR activity was measured by reporter assay and gene expression. Deployment related miRNA were measured by quantitative polymerase chain reaction. AHR expression was depleted using siRNA. RESULTS: BghiP displayed weak AHR agonist activity. HpCDD induced AHR activity in a dose-dependent manner. Let-7d-5p, miR-103-3p, miR-107, and miR-144-3p levels were significantly altered by HpCDD. AHR knockdown attenuated these effects. CONCLUSIONS: These studies reveal that miRNAs previously identified in sera from personnel deployed to sites with open burn pits are altered by HpCDD exposure in HLFs.


Asunto(s)
Fibroblastos/efectos de los fármacos , Pulmón/efectos de los fármacos , MicroARNs/metabolismo , Dibenzodioxinas Policloradas/efectos adversos , Fibroblastos/metabolismo , Humanos , Exposición por Inhalación/efectos adversos , Pulmón/citología , Pulmón/metabolismo
14.
PLoS One ; 14(9): e0222596, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31527897

RESUMEN

Proliferative vitreoretinopathy (PVR) is characterized by membranes that form in the vitreous cavity and on both surfaces of the retina, which results in the formation of tractional membranes that can cause retinal detachment and intrinsic fibrosis of the retina, leading to retina foreshortening. Currently, there are no pharmacologic therapies that are effective in inhibiting or preventing PVR formation. One of the key aspects of PVR pathogenesis is retinal pigment epithelial (RPE) cell epithelial mesenchymal transition (EMT). Here we show that the polyether ionophore compound salinomycin (SNC) effectively inhibits TGFß-induced EMT of RPE cells. SNC blocks the activation of TGFß-induced downstream targets alpha smooth muscle actin (αSMA) and collagen 1 (Col1A1). Additionally, SNC inhibits TGFß-induced RPE cell migration and contraction. We show that SNC functions to inhibit RPE EMT by targeting both the pTAK1/p38 and Smad2 signaling pathways upon TGFß stimulation. Additionally, SNC is able to inhibit αSMA and Col1A1 expression in RPE cells that have already undergone TGFß-induced EMT. Together, these results suggest that SNC could be an effective therapeutic compound in both the prevention and treatment of PVR.


Asunto(s)
Éter/farmacología , Piranos/farmacología , Transducción de Señal/efectos de los fármacos , Vitreorretinopatía Proliferativa/tratamiento farmacológico , Actinas/metabolismo , Línea Celular , Movimiento Celular/efectos de los fármacos , Colágeno Tipo I/metabolismo , Transición Epitelial-Mesenquimal/efectos de los fármacos , Humanos , Epitelio Pigmentado de la Retina/efectos de los fármacos , Epitelio Pigmentado de la Retina/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Vitreorretinopatía Proliferativa/metabolismo
15.
PLoS One ; 14(9): e0222779, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31536596

RESUMEN

Thyroid eye disease (TED) can lead to scar formation and tissue remodeling in the orbital space. In severe cases, the scarring process leads to sight-threatening pathophysiology. There is no known effective way to prevent scar formation in TED patients, or to reverse scarring once it occurs. In this study, we show that the proton pump inhibitors (PPIs), esomeprazole and lansoprazole, can prevent transforming growth factor beta (TGFß)-mediated differentiation of TED orbital fibroblasts to myofibroblasts, a critical step in scar formation. Both PPIs prevent TGFß-induced increases in alpha-smooth muscle actin (αSMA), calponin, and collagen production and reduce TED orbital fibroblast cell proliferation and migration. Esomeprazole and lansoprazole exert these effects through an aryl hydrocarbon receptor (AHR)-dependent pathway that includes reducing ß-catenin/Wnt signaling. We conclude that PPIs are potentially useful therapies for preventing or treating TED by reducing the myofibroblast accumulation that occurs in the disease.


Asunto(s)
Oftalmopatía de Graves/metabolismo , Miofibroblastos/efectos de los fármacos , Inhibidores de la Bomba de Protones/farmacología , Receptores de Hidrocarburo de Aril/metabolismo , Actinas/metabolismo , Proteínas de Unión al Calcio/metabolismo , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Cicatriz/metabolismo , Cicatriz/patología , Cicatriz/prevención & control , Colágeno/metabolismo , Esomeprazol/farmacología , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Oftalmopatía de Graves/patología , Células HEK293 , Humanos , Lansoprazol/farmacología , Proteínas de Microfilamentos/metabolismo , Miofibroblastos/metabolismo , Receptores de Hidrocarburo de Aril/genética , Factor de Crecimiento Transformador beta/metabolismo , Vía de Señalización Wnt/efectos de los fármacos , Calponinas
16.
Transfusion ; 59(6): 2007-2015, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30811035

RESUMEN

BACKGROUND: Relationships between red blood cell (RBC) transfusion, circulating cell-free heme, and clinical outcomes in critically ill transfusion recipients are incompletely understood. The goal of this study was to determine whether total plasma heme increases after RBC transfusion and predicts mortality in critically ill patients. STUDY DESIGN AND METHODS: This was a prospective cohort study of 111 consecutive medical intensive care patients requiring RBC transfusion. Cell-free heme was measured in RBC units before transfusion and in the patients' plasma before and after transfusion. RESULTS: Total plasma heme levels increased in response to transfusion, from a median (interquartile range [IQR]) of 35 (26-76) µmol/L to 47 (35-73) µmol/L (p < 0.001). Posttransfusion total plasma heme was higher in nonsurvivors (54 [35-136] µmol/L) versus survivors (44 [31-65] µmol/L, p = 0.03). Posttransfusion total plasma heme predicted hospital mortality (odds ratio [95% confidence interval] per quartile increase in posttransfusion plasma heme, 1.76 [1.17-2.66]; p = 0.007). Posttransfusion total plasma heme was not correlated with RBC unit storage duration and weakly correlated with RBC unit cell-free heme concentration. CONCLUSIONS: Total plasma heme concentration increases in critically ill patients after RBC transfusion and is independently associated with mortality. This transfusion-associated increase in total plasma heme is not fully explained by RBC unit storage age or cell-free heme content. Additional studies are warranted to define mechanisms of transfusion-related plasma heme accumulation and test prevention strategies.


Asunto(s)
Enfermedad Crítica/mortalidad , Enfermedad Crítica/terapia , Transfusión de Eritrocitos/efectos adversos , Hemo/metabolismo , Adulto , Anciano , Estudios de Cohortes , Transfusión de Eritrocitos/métodos , Transfusión de Eritrocitos/mortalidad , Femenino , Hemo/análisis , Mortalidad Hospitalaria , Humanos , Unidades de Cuidados Intensivos , Masculino , Persona de Mediana Edad , Pronóstico , Estudios Prospectivos , Resultado del Tratamiento
17.
Plast Reconstr Surg Glob Open ; 7(9): e2430, 2019 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-31942393

RESUMEN

Capsular contracture is a devastating complication that occurs in patients undergoing implant-based breast reconstruction. Ionizing radiation drives and exacerbates capsular contracture in part by activating cytokines, including transforming growth factor-beta (TGF-ß). TGF-ß promotes myofibroblast differentiation and proliferation, leading to excessive contractile scar formation. Therefore, targeting the TGF-ß pathway may attenuate capsular contracture. METHODS: A 20,000 small molecule library was screened for anti-TGF-ß activity. Structurally diverse anti-TGF-ß agents were identified and then tested on primary human capsular fibroblasts. Fibroblasts were irradiated or not, and then treated with both TGF-ß and candidate molecules. Resulting cells were then analyzed for myofibroblast activity using myofibroblast markers including alpha-smooth muscle actin, collagen I, Thy1, and periostin, using Western Blot, quantitative real-time polymerase chain reaction, and immunofluorescence. RESULTS: Human capsular fibroblasts treated with TGF-ß showed a significant increase in alpha-smooth muscle actin, collagen I, and periostin levels (protein and/or mRNA). Interestingly, fibroblasts treated with latent TGF-ß and 10 Gy radiation also showed significantly increased levels of myofibroblast markers. Cells that were treated with the novel small molecules showed a significant reduction in myofibroblast activation, even in the presence of radiation. CONCLUSIONS: Several novel small molecules with anti-TGF-ß activity can effectively prevent human capsular fibroblast to myofibroblast differentiation in vitro, even in the presence of radiation. These results highlight novel therapeutic options that may be utilized in the future to prevent radiation-induced capsular contracture.

18.
Am J Respir Cell Mol Biol ; 60(3): 269-278, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30265126

RESUMEN

The differentiation of interstitial lung fibroblasts into contractile myofibroblasts that proliferate and secrete excessive extracellular matrix is critical for the pathogenesis of pulmonary fibrosis. Certain lipid signaling molecules, such as prostaglandins (PGs), can inhibit myofibroblast differentiation. However, the sources and delivery mechanisms of endogenous PGs are undefined. Activated primary human lung fibroblasts (HLFs) produce PGs such as PGE2. We report that activation of primary HLFs with IL-1ß inhibited transforming growth factor ß-induced myofibroblast differentiation in both the IL-1ß-treated cells themselves (autocrine signal) and adjacent naive HLFs in cocultures (paracrine signal). Additionally, we demonstrate for the first time that at least some of the antifibrotic effect of activated fibroblasts on nearby naive fibroblasts is carried by exosomes and other extracellular vesicles that contain several PGs, including high levels of the antifibrotic PGE2. Thus, activated fibroblasts communicate with surrounding cells to limit myofibroblast differentiation and maintain homeostasis. This work opens the way for future research into extracellular vesicle-mediated intercellular signaling in the lung and may inform the development of novel therapies for fibrotic lung diseases.


Asunto(s)
Antifibrinolíticos/farmacología , Vesículas Extracelulares/efectos de los fármacos , Fibroblastos/efectos de los fármacos , Prostaglandinas/farmacología , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Dinoprostona/metabolismo , Exosomas/efectos de los fármacos , Exosomas/metabolismo , Matriz Extracelular/efectos de los fármacos , Matriz Extracelular/metabolismo , Vesículas Extracelulares/metabolismo , Femenino , Fibroblastos/metabolismo , Humanos , Interleucina-1beta/metabolismo , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Masculino , Miofibroblastos/efectos de los fármacos , Miofibroblastos/metabolismo , Fibrosis Pulmonar/tratamiento farmacológico , Fibrosis Pulmonar/metabolismo , Transducción de Señal/efectos de los fármacos , Factor de Crecimiento Transformador beta/metabolismo
19.
FASEB J ; 33(3): 3353-3363, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30376360

RESUMEN

The obesity epidemic is developing into the most costly health problem facing the world. Obesity, characterized by excessive adipogenesis and enlarged adipocytes, promotes morbidities, such as diabetes, cardiovascular disease, and cancer. Regulation of adipogenesis is critical to our understanding of how fat cell formation causes obesity and associated health problems. Thy1 (also called CD90), a widely used stem cell marker, blocks adipogenesis and reduces lipid accumulation. Thy1-knockout mice are prone to diet-induced obesity. Although the importance of Thy1 in adipogenesis and obesity is now evident, how its expression is regulated is not. We hypothesized that DNA methylation has a role in promoting adipogenesis and affects Thy1 expression. Using the methylation inhibitor 5-aza-2'-deoxycytidine (5-aza-dC), we investigated whether DNA methylation alters Thy1 expression during adipogenesis in both mouse 3T3-L1 preadipocytes and mouse mesenchymal stem cells. Thy1 protein and mRNA levels were decreased dramatically during adipogenesis. However, 5-aza-dC treatment prevented that phenomenon. Methylation-sensitive pyrosequencing analysis showed that CpG sites at the Thy1 locus have increased methylation during adipogenesis, as well as increased methylation in adipose tissue from diet-induced obese mice. These new findings highlight the potential role of Thy1 and DNA methylation in adipogenesis and obesity.-Flores, E. M., Woeller, C. F., Falsetta, M. L., Susiarjo, M., Phipps, R. P. Thy1 (CD90) expression is regulated by DNA methylation during adipogenesis.


Asunto(s)
Adipogénesis/genética , Metilación de ADN/genética , Antígenos Thy-1/genética , Células 3T3-L1 , Adipocitos/fisiología , Tejido Adiposo/fisiología , Animales , Diferenciación Celular/genética , Línea Celular , Células Madre Mesenquimatosas/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Obesos , Obesidad/genética , ARN Mensajero/genética , Células Madre/fisiología
20.
J Immunol ; 201(11): 3343-3351, 2018 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-30348736

RESUMEN

Vaccination has been the most effective way to prevent or reduce infectious diseases; examples include the eradication of smallpox and attenuation of tetanus and measles. However, there is a large segment of the population that responds poorly to vaccines, in part because they are immunocompromised because of disease, age, or pharmacologic therapy and are unable to generate long-term protection. Specialized proresolving mediators are endogenously produced lipids that have potent proresolving and anti-inflammatory activities. Lipoxin B4 (LXB4) is a member of the lipoxin family, with its proresolving effects shown in allergic airway inflammation. However, its effects on the adaptive immune system, especially on human B cells, are not known. In this study, we investigated the effects of LXB4 on human B cells using cells from healthy donors and donors vaccinated against influenza virus in vitro. LXB4 promoted IgG Ab production in memory B cells and also increased the number of IgG-secreting B cells. LXB4 enhanced expression of two key transcription factors involved in plasma cell differentiation, BLIMP1 and XBP1. Interestingly, LXB4 increased expression of cyclooxygenase-2 (COX2), an enzyme that is required for efficient B cell Ab production. The effects of LXB4 are at least partially COX2-dependent as COX2 inhibitors attenuated LXB4-stimulated BLIMP1 and Xpb-1 expression as well as IgG production. Thus, our study reveals for the first time, to our knowledge, that LXB4 boosts memory B cell activation through COX2 and suggests that LXB4 can serve as a new vaccine adjuvant.


Asunto(s)
Adyuvantes Inmunológicos/metabolismo , Anticuerpos Antivirales/metabolismo , Linfocitos B/inmunología , Ciclooxigenasa 2/metabolismo , Vacunas contra la Influenza/inmunología , Gripe Humana/inmunología , Lipoxinas/metabolismo , Inmunidad Adaptativa , Formación de Anticuerpos , Diferenciación Celular , Células Cultivadas , Ciclooxigenasa 2/genética , Humanos , Memoria Inmunológica , Factor 1 de Unión al Dominio 1 de Regulación Positiva/genética , Factor 1 de Unión al Dominio 1 de Regulación Positiva/metabolismo , Regulación hacia Arriba , Vacunación , Proteína 1 de Unión a la X-Box/genética , Proteína 1 de Unión a la X-Box/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...