Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Emerg Microbes Infect ; 10(1): 638-650, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-33704016

RESUMEN

SARS-CoV-2 is the causative agent of COVID-19 and responsible for the current global pandemic. We and others have previously demonstrated that cats are susceptible to SARS-CoV-2 infection and can efficiently transmit the virus to naïve cats. Here, we address whether cats previously exposed to SARS-CoV-2 can be re-infected with SARS-CoV-2. In two independent studies, SARS-CoV-2-infected cats were re-challenged with SARS-CoV-2 at 21 days post primary challenge (DPC) and necropsies performed at 4, 7 and 14 days post-secondary challenge (DP2C). Sentinels were co-mingled with the re-challenged cats at 1 DP2C. Clinical signs were recorded, and nasal, oropharyngeal, and rectal swabs, blood, and serum were collected and tissues examined for histologic lesions. Viral RNA was transiently shed via the nasal, oropharyngeal and rectal cavities of the re-challenged cats. Viral RNA was detected in various tissues of re-challenged cats euthanized at 4 DP2C, mainly in the upper respiratory tract and lymphoid tissues, but less frequently and at lower levels in the lower respiratory tract when compared to primary SARS-CoV-2 challenged cats at 4 DPC. Viral RNA and antigen detected in the respiratory tract of the primary SARS-CoV-2 infected cats at early DPCs were absent in the re-challenged cats. Naïve sentinels co-housed with the re-challenged cats did not shed virus or seroconvert. Together, our results indicate that cats previously infected with SARS-CoV-2 can be experimentally re-infected with SARS-CoV-2; however, the levels of virus shed was insufficient for transmission to co-housed naïve sentinels. We conclude that SARS-CoV-2 infection in cats induces immune responses that provide partial, non-sterilizing immune protection against re-infection.


Asunto(s)
Anticuerpos Antivirales/sangre , COVID-19/transmisión , Susceptibilidad a Enfermedades/inmunología , Reinfección/veterinaria , Esparcimiento de Virus , Animales , Anticuerpos Neutralizantes/sangre , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , COVID-19/veterinaria , Gatos , Línea Celular , Chlorocebus aethiops , ARN Viral/aislamiento & purificación , Reinfección/inmunología , Reinfección/virología , SARS-CoV-2/inmunología , Células Vero , Carga Viral
2.
Free Radic Biol Med ; 143: 25-46, 2019 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-31356870

RESUMEN

Elevation of blood triglycerides, primarily triglyceride-rich lipoproteins (TGRL), is an independent risk factor for cardiovascular disease and vascular dementia (VaD). Accumulating evidence indicates that both atherosclerosis and VaD are linked to vascular inflammation. However, the role of TGRL in vascular inflammation, which increases risk for VaD, remains largely unknown and its underlying mechanisms are still unclear. We strived to determine the effects of postprandial TGRL exposure on brain microvascular endothelial cells, the potential risk factor of vascular inflammation, resulting in VaD. We showed in Aung et al., J Lipid Res., 2016 that postprandial TGRL lipolysis products (TL) activate mitochondrial reactive oxygen species (ROS) and increase the expression of the stress-responsive protein, activating transcription factor 3 (ATF3), which injures human brain microvascular endothelial cells (HBMECs) in vitro. In this study, we deployed high-throughput sequencing (HTS)-based RNA sequencing methods and mito stress and glycolytic rate assays with an Agilent Seahorse XF analyzer and profiled the differential expression of transcripts, constructed signaling pathways, and measured mitochondrial respiration, ATP production, proton leak, and glycolysis of HBMECs treated with TL. Conclusions: TL potentiate ROS by mitochondria which activate mitochondrial oxidative stress, decrease ATP production, increase mitochondrial proton leak and glycolysis rate, and mitochondria DNA damage. Additionally, CPT1A1 siRNA knockdown suppresses oxidative stress and prevents mitochondrial dysfunction and vascular inflammation in TL treated HBMECs. TL activates ATF3-MAPKinase, TNF, and NRF2 signaling pathways. Furthermore, the NRF2 signaling pathway which is upstream of the ATF3-MAPKinase signaling pathway, is also regulated by the mitochondrial oxidative stress. We are the first to report differential inflammatory characteristics of transcript variants 4 (ATF3-T4) and 5 (ATF3-T5) of the stress responsive gene ATF3 in HBMECs induced by postprandial TL. Specifically, our data indicates that ATF3-T4 predominantly regulates the TL-induced brain microvascular inflammation and TNF signaling. Both siRNAs of ATF3-T4 and ATF3-T5 suppress cells apoptosis and lipotoxic brain microvascular endothelial cells. These novel signaling pathways triggered by oxidative stress-responsive transcript variants, ATF3-T4 and ATF3-T5, in the brain microvascular inflammation induced by TGRL lipolysis products may contribute to pathophysiological processes of vascular dementia.


Asunto(s)
Factor de Transcripción Activador 3/genética , Factor de Transcripción Activador 3/metabolismo , Encéfalo/patología , Microvasos/lesiones , Mitocondrias/metabolismo , Estrés Oxidativo , Apoptosis , Lesiones Encefálicas/metabolismo , Daño del ADN , Células Endoteliales/citología , Células Endoteliales/metabolismo , Variación Genética , Glucólisis , Humanos , Inflamación , Lipólisis , Microvasos/metabolismo , Consumo de Oxígeno , Periodo Posprandial , Protones , ARN Interferente Pequeño/metabolismo , RNA-Seq , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Superóxidos/metabolismo
3.
Food Funct ; 9(12): 6245-6256, 2018 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-30402637

RESUMEN

Our lab previously demonstrated that triglyceride-rich lipoprotein (TGRL) lipolysis products induce lipid droplet formation and pro-inflammatory gene expression in monocytes. We hypothesized that the inhibition of perilipin 2 expression in THP-1 monocytes would reduce lipid droplet formation and suppress pro-inflammatory gene expression induced by TGRL lipolysis products. In the current study, we use microarray analysis to identify gene expression altered by TGRL lipolysis products in THP-1 monocytes. We confirmed the expression of selected genes by quantitative reverse transcription PCR and characterized lipid droplet formation in these cells after exposure to TGRL lipolysis products. Using siRNA inhibition of perilipin 2 expression, we examined the role of perilipin 2 in the response of THP-1 monocytes to TGRL lipolysis products. We found that perilipin 2 siRNA increased the intracellular triglyceride content, increased the size of lipid droplets, and reduced pro-atherogenic and pro-inflammatory gene expression. We saw a reduction of serum/glucocorticoid kinase 1, v-maf musculoaponeurotic fibrosarcoma oncogene homolog F (avian), chemokine (C-C motif) ligand 3, and interleukin 8 gene expression induced by TGRL lipolysis products. This study supports previous findings that reduction of perilipin 2 expression is protective against atherogenesis, while finding an unexpected increase in lipid droplet size with reduced perilipin 2 expression.


Asunto(s)
Quimiocina CCL3/genética , Interleucina-8/genética , Gotas Lipídicas/metabolismo , Monocitos/efectos de los fármacos , Perilipina-2/genética , Adulto , Quimiocina CCL3/metabolismo , Regulación hacia Abajo , Femenino , Humanos , Interleucina-8/metabolismo , Gotas Lipídicas/química , Lipólisis , Lipoproteínas/metabolismo , Masculino , Monocitos/metabolismo , Perilipina-2/metabolismo , Triglicéridos/metabolismo , Adulto Joven
4.
Toxicol Sci ; 163(1): 140-151, 2018 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-29394414

RESUMEN

Particulate matter (PM) and ozone (O3) are dominant air pollutants that contribute to development and exacerbation of multiple cardiopulmonary diseases. Mature adults with cardiovascular disease (CVD) are particularly susceptible to air pollution-related cardiopulmonary morbidities and mortalities. The aim was to investigate the biologic potency of ultrafine particulate matter (UFPM) combined with O3 in the lungs of mature adult normotensive and spontaneously hypertensive (SH) Wistar-Kyoto rats. Conscious, mature adult male normal Wistar-Kyoto (NW) and SH rats were exposed to one of the following atmospheres: filtered air (FA); UFPM (∼ 250 µg/m3); O3 (1.0 ppm); or UFPM + O3 (∼ 250 µg/m3 + 1.0 ppm) combined for 6 h, followed by an 8 h FA recovery period. Lung sections were evaluated for lesions in the large airways, terminal bronchiolar/alveolar duct regions, alveolar parenchyma, and vasculature. NW and SH rats were similarly affected by the combined-pollutant exposure, displaying severe injury in both large and small airways. SH rats were particularly susceptible to O3 exposure, exhibiting increased injury scores in terminal bronchioles and epithelial degeneration in large airways. UFPM-exposure groups had minimal histologic changes. The chemical composition of UFPM was altered by the addition of O3, indicating that ozonolysis promoted compound degradation. O3 increased the biologic potency of UFPM, resulting in greater lung injury following exposure. Pathologic manifestations of CVD may confer susceptibility to air pollution by impairing normal lung defenses and responses to exposure.


Asunto(s)
Contaminantes Atmosféricos/toxicidad , Enfermedades Cardiovasculares/complicaciones , Lesión Pulmonar/inducido químicamente , Pulmón/efectos de los fármacos , Ozono/toxicidad , Material Particulado/toxicidad , Animales , Enfermedades Cardiovasculares/patología , Exposición por Inhalación , Pulmón/patología , Lesión Pulmonar/complicaciones , Lesión Pulmonar/patología , Masculino , Ozono/administración & dosificación , Ozono/química , Tamaño de la Partícula , Material Particulado/administración & dosificación , Material Particulado/química , Ratas Endogámicas SHR , Ratas Endogámicas WKY
5.
PLoS One ; 13(2): e0191909, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29444171

RESUMEN

Recent work suggests that diet affects brain metabolism thereby impacting cognitive function. Our objective was to determine if a western diet altered brain metabolism, increased blood-brain barrier (BBB) transport and inflammation, and induced cognitive impairment in C57BL/6 (WT) mice and low-density lipoprotein receptor null (LDLr -/-) mice, a model of hyperlipidemia and cognitive decline. We show that a western diet and LDLr -/- moderately influence cognitive processes as assessed by Y-maze and radial arm water maze. Also, western diet significantly increased BBB transport, as well as microvessel factor VIII in LDLr -/- and microglia IBA1 staining in WT, both indicators of activation and neuroinflammation. Interestingly, LDLr -/- mice had a significant increase in 18F- fluorodeoxyglucose uptake irrespective of diet and brain 1H-magnetic resonance spectroscopy showed increased lactate and lipid moieties. Metabolic assessments of whole mouse brain by GC/MS and LC/MS/MS showed that a western diet altered brain TCA cycle and ß-oxidation intermediates, levels of amino acids, and complex lipid levels and elevated proinflammatory lipid mediators. Our study reveals that the western diet has multiple impacts on brain metabolism, physiology, and altered cognitive function that likely manifest via multiple cellular pathways.


Asunto(s)
Barrera Hematoencefálica , Encéfalo/metabolismo , Cognición , Dieta Occidental , Receptores de LDL/genética , Animales , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
6.
PLoS One ; 12(1): e0169976, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28081568

RESUMEN

Systemic inflammation co-activates coagulation, which unchecked culminates in a lethal syndrome of multi-organ microvascular thrombosis known as disseminated intravascular coagulation (DIC). We studied an endotoxin-induced inflammatory state in rats to identify biomarkers of hemostatic imbalance favoring hypercoagulability. Intraperitoneal injection of LPS at 15 mg/kg body weight resulted in peripheral leukopenia and widespread neutrophilic sequestration characteristic of an acute systemic inflammatory response. Early indicators of hemostatic pathway activation developed within 4 hours, including increased circulating concentrations of procoagulant extracellular vesicles (EVs), EVs expressing endothelial cell and platelet membrane markers, and high concentration of soluble intercellular adhesion molecule-1 (sICAM-1), plasminogen activator inhibitor-1 (PAI-1), and D-dimers. Inflammation persisted throughout the 48-hour observation period; however, increases were found in a subset of serum microRNA (miRNA) that coincided with gradual resolution of hemostatic protein abnormalities and reduction in EV counts. Dose-adjusted LPS treatment in rats provides a time-course model to develop biomarker profiles reflecting procoagulant imbalance and rebalance under inflammatory conditions.


Asunto(s)
Lipopolisacáridos , Trombofilia/inducido químicamente , Trombofilia/fisiopatología , Enfermedad Aguda , Animales , Biomarcadores/sangre , Coagulación Sanguínea/efectos de los fármacos , Plaquetas/metabolismo , Modelos Animales de Enfermedad , Células Endoteliales/metabolismo , Vesículas Extracelulares/metabolismo , Productos de Degradación de Fibrina-Fibrinógeno/metabolismo , Molécula 1 de Adhesión Intercelular/metabolismo , Leucopenia/inducido químicamente , Masculino , MicroARNs/sangre , Neutrófilos/metabolismo , Neutrófilos/patología , Inhibidor 1 de Activador Plasminogénico/metabolismo , Ratas , Ratas Wistar , Trombofilia/inmunología , Factores de Tiempo
7.
Am J Physiol Cell Physiol ; 312(4): C500-C516, 2017 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-28077357

RESUMEN

Elevation of blood triglycerides, primarily as triglyceride-rich lipoproteins (TGRL), has been linked to cerebrovascular inflammation, vascular dementia, and Alzheimer's disease (AD). Brain microvascular endothelial cells and astrocytes, two cell components of the neurovascular unit, participate in controlling blood-brain barrier (BBB) permeability and regulating neurovascular unit homeostasis. Our studies showed that infusion of high physiological concentrations of TGRL lipolysis products (TGRL + lipoprotein lipase) activate and injure brain endothelial cells and transiently increase the BBB transfer coefficient (Ki = permeability × surface area/volume) in vivo. However, little is known about how blood lipids affect astrocyte lipid accumulation and inflammation. To address this, we first demonstrated TGRL lipolysis products increased lipid droplet formation in cultured normal human astrocytes. We then evaluated the transcriptional pathways activated in astrocytes by TGRL lipolysis products and found upregulated stress and inflammatory-related genes including activating transcription factor 3 (ATF3), macrophage inflammatory protein-3α (MIP-3α), growth differentiation factor-15 (GDF15), and prostaglandin-endoperoxide synthase 2 (COX2). TGRL lipolysis products also activated the JNK/cJUN/ATF3 pathway, induced endoplasmic reticulum stress protein C/EBP homologous protein (CHOP), and the NF-κB pathway, while increasing secretion of MIP-3α, GDF15, and IL-8. Thus our results demonstrate TGRL lipolysis products increase the BBB transfer coefficient (Ki), induce astrocyte lipid droplet formation, activate cell stress pathways, and induce secretion of inflammatory cytokines. Our observations are consistent with evidence for lipid-induced neurovascular injury and inflammation, and we, therefore, speculate that lipid-induced astrocyte injury could play a role in cognitive decline.


Asunto(s)
Astrocitos/metabolismo , Barrera Hematoencefálica/fisiología , Gotas Lipídicas/metabolismo , Lipólisis/fisiología , Lipoproteínas/metabolismo , Estrés Oxidativo/fisiología , Triglicéridos/metabolismo , Animales , Células Cultivadas , Masculino , Ratones , Ratones Endogámicos C57BL
8.
PLoS One ; 10(12): e0145523, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26709509

RESUMEN

Studies have suggested a link between the transforming growth factor beta 1 (TGF-ß1) signaling cascade and the stress-inducible activating transcription factor 3 (ATF3). We have demonstrated that triglyceride-rich lipoproteins (TGRL) lipolysis products activate MAP kinase stress associated JNK/c-Jun pathways resulting in up-regulation of ATF3, pro-inflammatory genes and induction of apoptosis in human aortic endothelial cells. Here we demonstrate increased release of active TGF-ß at 15 min, phosphorylation of Smad2 and translocation of co-Smad4 from cytosol to nucleus after a 1.5 h treatment with lipolysis products. Activation and translocation of Smad2 and 4 was blocked by addition of SB431542 (10 µM), a specific inhibitor of TGF-ß-activin receptor ALKs 4, 5, 7. Both ALK receptor inhibition and anti TGF-ß1 antibody prevented lipolysis product induced up-regulation of ATF3 mRNA and protein. ALK inhibition prevented lipolysis product-induced nuclear accumulation of ATF3. ALKs 4, 5, 7 inhibition also prevented phosphorylation of c-Jun and TGRL lipolysis product-induced p53 and caspase-3 protein expression. These findings demonstrate that TGRL lipolysis products cause release of active TGF-ß and lipolysis product-induced apoptosis is dependent on TGF-ß signaling. Furthermore, signaling through the stress associated JNK/c-Jun pathway is dependent on TGF-ß signaling suggesting that TGF-ß signaling is necessary for nuclear accumulation of the ATF3/cJun transcription complex and induction of pro-inflammatory responses.


Asunto(s)
Factor de Transcripción Activador 3/biosíntesis , Células Endoteliales/metabolismo , Lipoproteínas/metabolismo , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Triglicéridos/metabolismo , Transporte Activo de Núcleo Celular , Animales , Aorta/metabolismo , Apoptosis , Caspasa 3/metabolismo , Células Cultivadas , Humanos , Lipólisis , Lipoproteínas/genética , Ratones , Fosforilación , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transducción de Señal , Proteína Smad2/metabolismo , Proteína Smad4/metabolismo , Estrés Fisiológico , Factor de Crecimiento Transformador beta1/metabolismo , Triglicéridos/genética , Proteína p53 Supresora de Tumor/metabolismo
9.
Phys Chem Chem Phys ; 16(39): 21595-21601, 2014 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-25189785

RESUMEN

Activating transcription factor 3 (ATF3) is a member of the mammalian activation transcription factor/cAMP, physiologically important in the regulation of pro- and anti-inflammatory target genes. We compared the induction of ATF3 protein as measured by Western blot analysis with single-molecule localization microscopy dSTORM to quantify the dynamics of accumulation of intranuclear ATF3 of triglyceride-rich (TGRL) lipolysis product-treated HAEC (Human Aortic Endothelial Cells). The ATF3 expression rate within the first three hours after treatment with TGRL lipolysis products is about 3500 h(-1). After three hours we detected 33,090 ± 3491 single-molecule localizations of ATF3. This was accompanied by significant structural changes in the F-actin network of the cells at ∼3-fold increased localization precision compared to widefield microscopy after treatment. Additionally, we discovered a cluster size of approximately 384 nanometers of ATF3 molecules. We show for the first time the time course of ATF3 accumulation in the nucleus undergoing lipotoxic injury. Furthermore, we demonstrate ATF3 accumulation associated with increased concentrations of TGRL lipolysis products occurs in large aggregates.


Asunto(s)
Factor de Transcripción Activador 3/biosíntesis , Factor de Transcripción Activador 3/metabolismo , Aorta/citología , Aorta/metabolismo , Células Cultivadas , Células Endoteliales/citología , Células Endoteliales/metabolismo , Humanos , Lipólisis
10.
Arterioscler Thromb Vasc Biol ; 33(9): 2088-96, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23868936

RESUMEN

OBJECTIVE: Elevation of triglyceride-rich lipoproteins (TGRLs) contributes to the risk of atherosclerotic cardiovascular disease. Our work has shown that TGRL lipolysis products in high physiological to pathophysiological concentrations cause endothelial cell injury; however, the mechanisms remain to be delineated. APPROACH AND RESULTS: We analyzed the transcriptional signaling networks in arterial endothelial cells exposed to TGRL lipolysis products. When human aortic endothelial cells in culture were exposed to TGRL lipolysis products, activating transcription factor 3 (ATF3) was identified as a principal response gene. Induction of ATF3 mRNA and protein was confirmed by quantitative reverse-transcription polymerase chain reaction and Western blot respectively. Immunofluorescence analysis showed that ATF3 accumulated in the nuclei of cells treated with lipolysis products. Nuclear expression of phosphorylated c-Jun N-terminal kinase (JNK), previously shown to be an initiator of the ATF3 signaling cascade, also was demonstrated. Small interfering RNA (siRNA)-mediated inhibition of ATF3 blocked lipolysis products-induced transcription of E-selectin and interleukin-8, but not interleukin-6 or nuclear factor-κB. c-Jun, a downstream protein in the JNK pathway, was phosphorylated, whereas expression of nuclear factor-κB-dependent JunB was downregulated. Additionally, JNK siRNA suppressed ATF3 and p-c-Jun protein expression, suggesting that JNK is upstream of the ATF3 signaling pathway. In vivo studies demonstrated that infusion of TGRL lipolysis products into wild-type mice induced nuclear ATF3 accumulation in carotid artery endothelium. ATF3(-/-) mice were resistant to vascular apoptosis precipitated by treatment with TGRL lipolysis products. Also peripheral blood monocytes isolated from postprandial humans had increased ATF3 expression as compared with fasting monocytes. CONCLUSIONS: This study demonstrates that TGRL lipolysis products activate ATF3-JNK transcription factor networks and induce endothelial cells inflammatory response.


Asunto(s)
Factor de Transcripción Activador 3/metabolismo , Apoptosis , Células Endoteliales/metabolismo , Inflamación/metabolismo , Lipoproteínas/metabolismo , Triglicéridos/metabolismo , Factor de Transcripción Activador 3/deficiencia , Factor de Transcripción Activador 3/genética , Animales , Western Blotting , Células Cultivadas , Selectina E/metabolismo , Células Endoteliales/inmunología , Células Endoteliales/patología , Activación Enzimática , Técnica del Anticuerpo Fluorescente , Perfilación de la Expresión Génica/métodos , Regulación de la Expresión Génica , Humanos , Inflamación/genética , Inflamación/inmunología , Inflamación/patología , Mediadores de Inflamación/metabolismo , Interleucina-8/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Leucocitos Mononucleares/metabolismo , Lipólisis , Lipoproteína Lipasa/metabolismo , Lipoproteínas/sangre , Lipoproteínas/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Análisis de Secuencia por Matrices de Oligonucleótidos , Fosforilación , Interferencia de ARN , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transfección , Triglicéridos/sangre
11.
Toxicol Pathol ; 41(6): 880-92, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23129576

RESUMEN

Dilated cardiomyopathy (DCM) in A/J mice homozygous for the spontaneous thrombocytopenia and cardiomyopathy (trac) mutation results from a single base pair change in the Abcg5 gene. A similar mutation in humans causes sitosterolemia with high plant sterol levels, hypercholesterolemia, and early onset atherosclerosis. Analyses of CD3+ and Mac-3+ cells and stainable collagen in hearts showed inflammation and myocyte degeneration in A/J-trac/trac mice beginning postweaning and progressed to marked dilative and fibrosing cardiomyopathy by 140 days. Transmission electron microscopy (TEM) demonstrated myocyte vacuoles consistent with swollen endoplasmic reticulum (ER). Myocytes with cytoplasmic glycogen and irregular actinomyosin filament bundles formed mature intercalated disks with normal myocytes suggesting myocyte repair. A/J-trac/trac mice fed lifelong phytosterol-free diets did not develop cardiomyopathy. BALB/cByJ-trac/trac mice had lesser inflammatory infiltrates and later onset DCM. BALB/cByJ-trac/trac mice changed from normal to phytosterol-free diets had lesser T cell infiltrates but persistent monocyte infiltrates and equivalent fibrosis to mice on normal diets. B- and T-cell-deficient BALB/cBy-Rag1(null) trac/trac mice fed normal diets did not develop inflammatory infiltrates or DCM. We conclude that the trac/trac mouse has many features of inflammatory DCM and that the reversibility of myocardial T cell infiltration provides a novel model for investigating the progression of myocardial fibrosis.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/deficiencia , Cardiomiopatía Dilatada/metabolismo , Inflamación/metabolismo , Lipoproteínas/deficiencia , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 5 , Transportadoras de Casetes de Unión a ATP/genética , Animales , Cardiomiopatía Dilatada/genética , Cardiomiopatía Dilatada/patología , Modelos Animales de Enfermedad , Ecocardiografía , Femenino , Fibrosis/metabolismo , Fibrosis/patología , Histocitoquímica , Inflamación/genética , Inflamación/patología , Lipoproteínas/genética , Masculino , Ratones , Ratones Transgénicos , Microscopía Electrónica , Monocitos/metabolismo , Monocitos/patología , Miocardio/química , Miocardio/metabolismo , Miocardio/patología , Miofibrillas/metabolismo , Miofibrillas/patología , Fitosteroles/farmacología , Linfocitos T/metabolismo , Linfocitos T/patología
12.
Inhal Toxicol ; 24(8): 506-17, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22746400

RESUMEN

Increasing evidence suggests a role for a systemic pro-coagulant state in the pathogenesis of cardiac dysfunction subsequent to inhalation of airborne particulate matter (PM). We evaluated platelet activation, systemic cytokines and pulmonary gene expression in mice exposed to concentrated ambient particulate matter (CAPs) in the summer of 2008 (S08) and winter of 2009 (W09) from the San Joaquin Valley of California, a region with severe PM pollution episodes. Additionally, we characterized the PM from both exposures including organic compounds, metals, and polycyclic aromatic hydrocarbons. Mice were exposed to an average of 39.01 µg/m(3) of CAPs in the winter and 21.7 µg/m3 CAPs in the summer, in a size range less than 2.5 µm for 6 h/day for 5 days per week for 2 weeks. Platelets were analyzed by flow cytometry for relative size, shape, CD41, P-selectin and lysosomal associated membrane protein-1 (LAMP-1) expression. Platelets from W09 CAPs-exposed animals had a greater response to thrombin stimulation than platelets from S08 CAPs-exposed animals. Serum cytokines were analyzed by bead based immunologic assays. W09 CAPs-exposed mice had elevations in IL-2, MIP-1α, and TNFα. Laser capture microdissection (LCM) of pulmonary vasculature, parenchyma and airways all showed increases in CYP1a1 gene expression. Pulmonary vasculature showed increased expression of ICAM-1 and Nox-2. Our findings demonstrate that W09 CAPs exposure generated a greater systemic pro-inflammatory and pro-coagulant response to inhalation of environmentally derived fine and ultrafine PM. Changes in platelet responsiveness to agonists, seen in both exposures, strongly suggests a role for platelet activation in the cardiovascular and respiratory effects of particulate air pollution.


Asunto(s)
Contaminantes Atmosféricos/toxicidad , Citocinas/sangre , Regulación de la Expresión Génica/efectos de los fármacos , Material Particulado/toxicidad , Activación Plaquetaria/efectos de los fármacos , Estaciones del Año , Animales , California , Monitoreo del Ambiente , Perfilación de la Expresión Génica , Exposición por Inhalación , Pulmón/irrigación sanguínea , Pulmón/efectos de los fármacos , Pulmón/inmunología , Pulmón/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Tamaño de la Partícula
13.
Physiol Genomics ; 43(15): 917-29, 2011 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-21652769

RESUMEN

Epidemiologic studies associate exposure to ambient particulate matter (APM) with increased cardiovascular mortality. Since both pulmonary inflammation and systemic circulation of ultrafine particles are hypothesized as initiating cardiovascular effects, we examined responses of potential target cells in vitro. Human aortic endothelial cells (HAEC) were exposed to 10 µg/ml fine and ultrafine APM collected in an urban setting in summer 2006 or winter 2007 in the San Joaquin Valley, California. RNA isolated after 3 h was analyzed with high-density oligonucleotide arrays. Summer APM treatment affected genes involved in xenobiotic and oxidoreductase activity, transcription factors, and inflammatory responses in HAEC, while winter APM had a robust xenobiotic but lesser inflammatory response. Real-time polymerase chain reaction analysis confirmed that particulate matter (PM)-treated HAEC increased mRNA levels of xenobiotic response enzymes CYP1A1, ALDH1A3, and TIPARP and cellular stress response transcription factor ATF3. Inflammatory response genes included E-selectin, PTGS2, CXCL-2 (MIP-2α), and CCL-2 (MCP-1). Multiplex protein assays showed secretion of IL-6 and MCP-1 by HAEC. Since induction of CYP1A1 is mediated through the ligand-activated aryl hydrocarbon receptor (AhR), we demonstrated APM induced AhR nuclear translocation by immunofluorescence and Western blotting and activation of the AhR response element using a luciferase reporter construct. Inhibitor studies suggest differential influences of polycyclic aromatic hydrocarbon signaling, ROS-mediated responses and endotoxin alter stress and proinflammatory endothelial cell responses. Our findings demonstrate gene responses correlated with current concepts that systemic inflammation drives cardiovascular effects of particulate air pollution. We also demonstrate a unique pattern of gene responses related to xenobiotic metabolism in PM-exposed HAEC.


Asunto(s)
Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Material Particulado/toxicidad , Apoptosis/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Quimiocina CCL2/genética , Quimiocina CCL2/metabolismo , Quimiocina CXCL2/genética , Citocromo P-450 CYP1A1/genética , Sistema Enzimático del Citocromo P-450/genética , Selectina E/genética , Humanos , Interleucina-6/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena de la Polimerasa , Receptores de Hidrocarburo de Aril/genética
14.
Chem Res Toxicol ; 23(9): 1504-13, 2010 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-20695460

RESUMEN

Protein-xenobiotic adducts are byproducts of xenobiotic metabolism. While there is a correlation between protein adduction and target organ toxicity, a cause and effect relationship is not often clear. Naphthoquinone (NQ) and monocrotaline pyrrole (MCTP) are two pneumotoxic electrophiles that form covalent adducts with a similar select group of proteins rich in reactive thiols. In this study, we treated human pulmonary artery endothelial cells (HPAEC) with NQ, MCTP, or preformed NQ or MCTP adducts to the protein galectin-1 (gal-1) and examined indicators of reactive oxygen species (ROS) oxidative injury, markers of apoptosis (caspase-3 and annexin V), and gene responses of cellular stress. ROS production was assayed fluorescently using CM-H(2)DCFDA. NQ adducts to gal-1 (NQ-gal) produced 183% more intracellular ROS than gal-1 alone (p < 0.0001). Caspase-3 activity and annexin V staining of phosphatidylserine were used to assess apoptotic activity in treated cells. HPAEC exposed to MCTP-gal had increases in both caspase-3 activation and membrane translocation of annexin V relative to gal-1 alone (p < 0.0001). Direct application of NQ produced significantly more ROS and induced significant caspase-3 activation, whereas MCTP did not. Human bronchial epithelial cells were also exposed to MCTP-gal and found to have significant increases in both caspase-3 activation and annexin V staining in comparison to that of gal-1 (p < 0.05). Western blot analysis showed that both NQ and MCTP significantly induced the Nrf2 mediated stress response pathway despite differences in ROS generation. ER stress was not induced by either adducts or parent compounds as seen by quantitative RT-PCR, but HOX-1 expression was significantly induced by NQ-gal and MCTP alone. Electrophile adduction to gal-1 produces different cytotoxic effects specific to each reactive intermediate.


Asunto(s)
Galectina 1/química , Monocrotalina/análogos & derivados , Naftoquinonas/química , Anexina A5/metabolismo , Apoptosis , Caspasa 3/metabolismo , Línea Celular , Femenino , Colorantes Fluorescentes/química , Galectina 1/metabolismo , Humanos , Monocrotalina/química , Monocrotalina/toxicidad , Factor 2 Relacionado con NF-E2/metabolismo , Naftoquinonas/toxicidad , Especies Reactivas de Oxígeno/metabolismo
15.
Nat Rev Nephrol ; 6(6): 361-70, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20440276

RESUMEN

Diabetic nephropathy is an increasingly important cause of morbidity and mortality worldwide. A large body of evidence suggests that dyslipidemia has an important role in the progression of kidney disease in patients with diabetes. Lipids may induce renal injury by stimulating TGF-beta, thereby inducing the production of reactive oxygen species and causing damage to the glomeruli and glomerular glycocalyx. Findings from basic and clinical studies strongly suggest that excess amounts of a variety of lipoproteins and lipids worsens diabetes-associated microvascular and macrovascular disease, increases glomerular injury, increases tubulointerstitial fibrosis, and accelerates the progression of diabetic nephropathy. The increasing prevalence of obesity, type 2 diabetes mellitus, and diabetic nephropathy means that interventions that can interrupt the pathophysiological cascade of events induced by lipoproteins and lipids could enable major life and cost savings. This Review discusses the structural, cellular, and microscopic findings associated with diabetic nephropathy and the influence of lipoproteins, specifically triglyceride-rich lipoproteins (TGRLs), on the development and perpetuation of diabetic nephropathy. Some of the accepted and hypothesized mechanisms of renal injury relating to TGRLs are also described.


Asunto(s)
Nefropatías Diabéticas/metabolismo , Nefropatías Diabéticas/fisiopatología , Dislipidemias/metabolismo , Dislipidemias/fisiopatología , Lipoproteínas/metabolismo , Triglicéridos/metabolismo , Animales , Nefropatías Diabéticas/tratamiento farmacológico , Dislipidemias/tratamiento farmacológico , Humanos , Hipolipemiantes/uso terapéutico , Lipoproteínas/efectos de los fármacos
16.
J Aerosol Med Pulm Drug Deliv ; 23(4): 197-206, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19803732

RESUMEN

BACKGROUND: Osteosarcoma is the most common skeletal malignancy in the dog and in young humans. Although chemotherapy improves survival time, death continues to be attributed to metastases. Aerosol delivery can provide a strategy with which to improve the lung drug delivery while reducing systemic toxicity. The purpose of this study is to assess the safety of a regional aerosol approach to chemotherapy delivery in osteosarcoma-bearing dogs, and second, to evaluate the effect of gemcitabine on Fas expression in the pulmonary metastasis. METHODS: We examined the systemic and local effects of aerosol gemcitabine on lung and pulmonary metastasis in this relevant large-animal tumor model using serial laboratory and arterial blood gas analysis and histopathology and immunohistochemistry, respectively. RESULTS AND CONCLUSIONS: Six hundred seventy-two 1-h doses of aerosol gemcitabine were delivered. The treatment was well tolerated by these subjects with osteosarcoma (n = 20). Aerosol-treated subjects had metastatic foci that demonstrated extensive, predominately central, intratumoral necrosis. Fas expression was decreased in pulmonary metastases compared to the primary tumor (p = 0.008). After aerosol gemcitabine Fas expression in the metastatic foci was increased compared to lung metastases before treatment (p = 0.0075), and even was higher than the primary tumor (p = 0.025). Increased apoptosis (TUNEL) staining was also detected in aerosol gemcitabine treated metastasis compared to untreated controls (p = 0.028). The results from this pivotal translational study support the concept that aerosol gemcitabine may be useful against pulmonary metastases of osteosarcoma. Additional studies that evaluate the aerosol route of administration of gemcitabine in humans should be safe and are warranted.


Asunto(s)
Antimetabolitos Antineoplásicos/uso terapéutico , Neoplasias Óseas/tratamiento farmacológico , Desoxicitidina/análogos & derivados , Neoplasias Pulmonares/tratamiento farmacológico , Osteosarcoma/tratamiento farmacológico , Aerosoles , Animales , Antimetabolitos Antineoplásicos/administración & dosificación , Antimetabolitos Antineoplásicos/efectos adversos , Neoplasias Óseas/patología , Desoxicitidina/administración & dosificación , Desoxicitidina/efectos adversos , Desoxicitidina/uso terapéutico , Perros , Proteína Ligando Fas/fisiología , Neoplasias Pulmonares/secundario , Osteosarcoma/secundario , Receptor fas/fisiología , Gemcitabina
17.
Toxicol Pathol ; 36(2): 311-20, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18367643

RESUMEN

Mutations in the bone morphogenetic protein receptor type II (BMPrII) gene have been implicated in the development of familial pulmonary artery hypertension (PAH). The function of BMP signal transduction within the pulmonary vasculature and the role BMPrII mutations have in the development of PAH are incompletely understood. We used the monocrotaline (MCT) model of PAH to examine alterations in Smad signal transduction pathways in vivo. Lungs harvested from Sprague-Dawley rats treated with a single 60-mg/kg intraperitoneal (IP) injection of MCT were compared to saline-treated controls 2 weeks following treatment. Smad 4 was localized by immunohistochemistry to endothelial nuclei of the intra-acinar vessels undergoing remodeling. Smad 4, common to both BMP and transforming growth factor beta (TGFbeta) signaling, and BMP-specific Smad 1 were significantly decreased in western blot from whole lungs of treated animals, while no change was found for TGFbeta-specific Smad 2. MCT-treated rats also had increased expression of phosphorylated Smad 1 (P-Smad 1) but not phosphorylated Smad 2 (P-Smad 2). There was a decrease in the expression of the full BMPrII protein but not its short form variant in MCT-treated rat lungs. The type I receptor Alk1 had increased expression. Collectively, our data indicate that vascular remodeling in the MCT model is associated with alterations in BMP receptors and persistent endothelial Smad 1 signaling.


Asunto(s)
Receptores de Activinas/metabolismo , Receptores de Proteínas Morfogenéticas Óseas de Tipo II/metabolismo , Hipertensión Pulmonar/metabolismo , Monocrotalina/toxicidad , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Proteína Smad4/metabolismo , Animales , Proteínas Morfogenéticas Óseas/metabolismo , Modelos Animales de Enfermedad , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/metabolismo , Endotelio Vascular/patología , Hipertensión Pulmonar/inducido químicamente , Inyecciones Intraperitoneales , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Pulmón/patología , Masculino , Ratas , Ratas Sprague-Dawley , Transducción de Señal , Proteína Smad1/metabolismo , Proteína Smad2/metabolismo , Factor de Crecimiento Transformador beta/metabolismo
18.
Am J Physiol Heart Circ Physiol ; 292(6): H2745-53, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17259442

RESUMEN

Products generated from lipoprotein lipase-mediated hydrolysis of triglyceride-rich lipoproteins (TGRL) are reported to increase endothelial layer permeability. We hypothesize that these increases in permeability result from the active rearrangement and dissolution of the junctional barrier in human aortic endothelial cells, as well as induction of the apoptotic cascade. Human aortic endothelial cells were treated with TGRL lipolysis products generated from coincubation of human TGRL plus lipoprotein lipase. Measurement of transendothelial electrical resistance demonstrated a time-dependent decrease in endothelial barrier function in response to TGRL lipolysis products. Immunofluorescent localization of zonula occludens-1 (ZO-1) showed radial rearrangement along cell borders after 1.5 h of treatment with lipolysis products. A concurrent redistribution of F-actin from the cell body to the cell margins was observed via rhodamine phalloidin staining. Immunofluorescent imaging for occludin and vascular endothelial cadherin showed that these proteins relocalize as well, although these changes are less prominent than for ZO-1. Western analysis of cells exposed to lipolysis products for 3 h revealed the fragmentation of ZO-1, a reduction in occludin, and no change of vascular endothelial cadherin. Lipolysis products also increased caspase-3 activity and induced nuclear fragmentation. Treatments did not cause oncosis in cells at any point during the incubation. These results demonstrate that TGRL lipolysis products play an important role in the regulation of endothelial permeability, the organization of the actin cytoskeleton, the localization and expression of junctional proteins, especially ZO-1, and the induction of apoptosis.


Asunto(s)
Actinas/metabolismo , Apoptosis , Permeabilidad Capilar , Células Endoteliales/metabolismo , Lipólisis , Lipoproteínas/metabolismo , Proteínas de la Membrana/metabolismo , Fosfoproteínas/metabolismo , Uniones Estrechas/metabolismo , Triglicéridos/metabolismo , Antígenos CD/metabolismo , Aorta/citología , Aorta/metabolismo , Aorta/patología , Apoptosis/efectos de los fármacos , Cadherinas/metabolismo , Permeabilidad Capilar/efectos de los fármacos , Caspasa 3/metabolismo , Células Cultivadas , Impedancia Eléctrica , Células Endoteliales/efectos de los fármacos , Células Endoteliales/patología , Activación Enzimática , Humanos , Hidrólisis , Lipoproteína Lipasa/metabolismo , Lipoproteínas/farmacología , Ocludina , Uniones Estrechas/efectos de los fármacos , Factores de Tiempo , Triglicéridos/farmacología , Proteína de la Zonula Occludens-1
19.
J Vasc Surg ; 44(2): 364-71, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16890870

RESUMEN

BACKGROUND: We explored the role of angiotensin II in determining the histomorphometric features of plaque stability in apolipoprotein E-deficient mice submitted to ligation of the carotid artery. METHODS: Six-month-old apolipoprotein E-deficient mice underwent ligation of the common left carotid artery and were immediately assigned to receive either angiotensin II (1.4 mg . kg(-1) . d(-1) subcutaneously) or vehicle (phosphate-buffered saline; control) via a subcutaneous osmotic minipump for 4 weeks. RESULTS: Ligated arteries from control animals developed intimal lesions composed of macrophage foam cell plaques, which accumulated adjacent to the internal elastic lamina and were surrounded by a fibromuscular layer. Angiotensin II-treated mice had a greater intimal area (threefold), which was accompanied by a fivefold increase in the foam cell area. Lesions from angiotensin II-treated mice also displayed complex morphology characterized by intralesional neovasculature and hemorrhage. The content of active matrix metalloproteinase 2, mainly colocalized with macrophage foam cells, and the production of the inflammatory mediators monocyte chemoattractant protein 1 and vascular cell adhesion molecule 1 were also increased by angiotensin II treatment. Although angiotensin II induced vessel expansion and lumen loss to a similar extent, only vessel enlargement correlated with intimal area. CONCLUSIONS: Taken together, this study's results support a role of angiotensin II in plaque vulnerability by promoting intraplaque neovascularization/hemorrhage, inflammation, and expansive remodeling.


Asunto(s)
Angiotensina II , Aterosclerosis/patología , Arterias Carótidas/patología , Enfermedades de las Arterias Carótidas/patología , Animales , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Aterosclerosis/etiología , Aterosclerosis/metabolismo , Arterias Carótidas/efectos de los fármacos , Arterias Carótidas/cirugía , Enfermedades de las Arterias Carótidas/etiología , Enfermedades de las Arterias Carótidas/metabolismo , Quimiocina CCL2/metabolismo , Modelos Animales de Enfermedad , Células Espumosas/efectos de los fármacos , Células Espumosas/patología , Inmunohistoquímica , Ligadura , Masculino , Metaloproteinasa 2 de la Matriz/metabolismo , Ratones , Ratones Noqueados , Túnica Íntima/efectos de los fármacos , Túnica Íntima/metabolismo , Túnica Íntima/patología , Molécula 1 de Adhesión Celular Vascular/metabolismo
20.
Arterioscler Thromb Vasc Biol ; 26(4): 765-72, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16456092

RESUMEN

BACKGROUND: Endothelial NO deficiency (endothelial NO synthase [eNOS]-knockout [KO]) enhanced smooth muscle cell (SMC)-rich neointimal lesion formation in a mouse model of carotid artery ligation (CAL). Recent evidence indicated that stromal cell-derived factor-1alpha (SDF-1alpha)-mediated recruitment of circulating SMC progenitor cells substantially contributed to the SMC-rich neointimal hyperplasia induced by vascular injury. The goal of this study was to investigate the effects of eNOS deficiency on the expression of SDF-1alpha and mobilization of circulating SMC progenitor cells in CAL model. METHODS AND RESULTS: Two- to 3-month-old C57BL/6J wild-type (WT) and eNOS-KO mice were evaluated 1, 2, or 4 weeks after CAL. CAL-induced expression of SDF-1alpha, as detected by immunohistochemical staining and further quantified by ELISA in the ligated carotid arteries, was moderate and transient with a peak at 1 week in WT mice. SDF-1alpha expression was significantly higher at 1 week and persisted through 2 weeks in eNOS-KO mice. CAL was associated with increased circulating stem cell antigen-1(+) (Sca-1(+))/c-Kit(-)/Lin- cells (interpreted as SMC progenitor cells), which peaked at 1 week in WT mice. This effect was also significantly greater and longer-lasting in eNOS-KO than WT mice. The number of circulating Sca-1(+)/c-Kit(-)/Lin- cells was positively correlated with the expression of SDF-1alpha but not vascular endothelial growth factor in the ligated carotid arteries. Furthermore, immunostaining showed abundant Sca-1-positive cells in the adventitia of the 1-week ligated carotid arteries from eNOS-KO mice but not in WT mice. We also determined that eNOS deficiency enhanced CAL-induced intimal cell proliferation in the ligated arteries as detected by proliferating cell nuclear antigen staining but did not induce cell apoptosis as detected by staining for active caspase-3. CONCLUSIONS: Our results indicate that eNOS deficiency exacerbates CAL-induced expression of SDF-1alpha and its receptor CXCR4. This is correlated with an increase in Sca-1(+) cells in peripheral blood and adventitia, which may contribute to vascular remodeling and SMC-rich neointimal lesion formation. This suggests that constitutive eNOS inhibits SDF-1alpha expression and provides an important vasculoprotective mechanism for intact endothelium to limit SMC proliferation and recruitment in response to vascular injury.


Asunto(s)
Traumatismos de las Arterias Carótidas/enzimología , Traumatismos de las Arterias Carótidas/patología , Quimiocinas CXC/biosíntesis , Músculo Liso Vascular/patología , Óxido Nítrico Sintasa de Tipo III/deficiencia , Túnica Íntima/patología , Animales , Arterias Carótidas/patología , Arterias Carótidas/fisiopatología , Traumatismos de las Arterias Carótidas/fisiopatología , Diferenciación Celular , Proliferación Celular , Quimiocina CXCL12 , Técnicas In Vitro , Metaloproteinasa 2 de la Matriz/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células Madre/patología , Células Madre/fisiología , Túnica Íntima/fisiopatología , Regulación hacia Arriba , Factores de Crecimiento Endotelial Vascular/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...