Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
1.
Nat Commun ; 14(1): 7112, 2023 11 06.
Article in English | MEDLINE | ID: mdl-37932311

ABSTRACT

An unresolved issue in contemporary biomedicine is the overwhelming number and diversity of complex images that require annotation, analysis and interpretation. Recent advances in Deep Learning have revolutionized the field of computer vision, creating algorithms that compete with human experts in image segmentation tasks. However, these frameworks require large human-annotated datasets for training and the resulting "black box" models are difficult to interpret. In this study, we introduce Kartezio, a modular Cartesian Genetic Programming-based computational strategy that generates fully transparent and easily interpretable image processing pipelines by iteratively assembling and parameterizing computer vision functions. The pipelines thus generated exhibit comparable precision to state-of-the-art Deep Learning approaches on instance segmentation tasks, while requiring drastically smaller training datasets. This Few-Shot Learning method confers tremendous flexibility, speed, and functionality to this approach. We then deploy Kartezio to solve a series of semantic and instance segmentation problems, and demonstrate its utility across diverse images ranging from multiplexed tissue histopathology images to high resolution microscopy images. While the flexibility, robustness and practical utility of Kartezio make this fully explicable evolutionary designer a potential game-changer in the field of biomedical image processing, Kartezio remains complementary and potentially auxiliary to mainstream Deep Learning approaches.


Subject(s)
Algorithms , Image Processing, Computer-Assisted , Humans , Image Processing, Computer-Assisted/methods , Microscopy , Biological Evolution , Semantics
2.
Mucosal Immunol ; 14(4): 912-922, 2021 07.
Article in English | MEDLINE | ID: mdl-33846533

ABSTRACT

Acute exacerbations (AE) of asthma, remain one of the biggest concerns for patients living with asthma. As such, identifying the causes, the molecular mechanisms involved and new therapeutic interventions to prevent AE is a high priority. Immunity to intestinal helminths involves the reactivation of type-2 immune responses leading to smooth muscle contraction and mucus hypersecretion-physiological processes very similar to acute exacerbations in the airways following allergen exposure. In this study, we employed a murine model of intestinal helminth infection, using Heligmosomoides polygyrus, to identify miRNAs during active expulsion, as a system for the identification of miRNAs that may contribute to AE in the airways. Concomitant with type-2 immunity and expulsion of H. polygyrus, we identified miR-99a-5p, miR-148a-3p and miR-155-5p that were differentially regulated. Systemic inhibition of these miRNAs, alone or in combination, had minimal impact on expulsion of H. polygyrus, but inhibition of miR-99a-5p or miR-155-5p significantly reduced house dust mite (HDM)-driven acute inflammation, modelling human acute exacerbations. Immunological, pathological and transcriptional analysis identified that miR-155-5p or miR-99a-5p contribute significantly to HDM-driven AE and that transient inhibition of these miRNAs may provide relief from allergen-driven AE, without compromising anti-helminth immunity in the gut.


Subject(s)
Allergens/immunology , Asthma/etiology , Immunologic Memory , MicroRNAs/genetics , Animals , Asthma/metabolism , Asthma/pathology , Biomarkers , Disease Progression , Disease Susceptibility , Gene Expression Profiling , Helminthiasis, Animal/complications , Helminthiasis, Animal/immunology , Helminthiasis, Animal/parasitology , Host-Parasite Interactions , Immunity, Innate , Mice
3.
Emerg Microbes Infect ; 10(1): 638-650, 2021 Dec.
Article in English | MEDLINE | ID: mdl-33704016

ABSTRACT

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.


Subject(s)
Antibodies, Viral/blood , COVID-19/transmission , Disease Susceptibility/immunology , Reinfection/veterinary , Virus Shedding , Animals , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , COVID-19/veterinary , Cats , Cell Line , Chlorocebus aethiops , RNA, Viral/isolation & purification , Reinfection/immunology , Reinfection/virology , SARS-CoV-2/immunology , Vero Cells , Viral Load
4.
Retin Cases Brief Rep ; 14(1): 20-22, 2020.
Article in English | MEDLINE | ID: mdl-28644176

ABSTRACT

PURPOSE: To describe a case of central retinal vein occlusion in a young patient presenting with symptomatic malignant hypertension because of Conn syndrome. METHODS: Single interventional case report. RESULTS: A 44-year-old man presented with a 1-day history of headache and vision loss in his right eye on a background of malignant hypertension. He was diagnosed with right central retinal vein occlusion. Further investigation of his malignant hypertension revealed Conn syndrome because of an aldosterone-secreting adenoma in the left adrenal gland. CONCLUSION: This is the first reported case of Conn syndrome presenting as central retinal vein occlusion. Conn syndrome should be suspected in young patients with central retinal vein occlusion because it is a common cause of hypertension and carries significant cardiovascular risk if left untreated.


Subject(s)
Adrenal Glands/diagnostic imaging , Fluorescein Angiography/methods , Hyperaldosteronism/complications , Retinal Vein Occlusion/etiology , Retinal Vessels/pathology , Adult , Diagnosis, Differential , Fundus Oculi , Humans , Hyperaldosteronism/diagnosis , Male , Retinal Vein Occlusion/diagnosis , Tomography, X-Ray Computed
5.
Free Radic Biol Med ; 143: 25-46, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31356870

ABSTRACT

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.


Subject(s)
Activating Transcription Factor 3/genetics , Activating Transcription Factor 3/metabolism , Brain/pathology , Microvessels/injuries , Mitochondria/metabolism , Oxidative Stress , Apoptosis , Brain Injuries/metabolism , DNA Damage , Endothelial Cells/cytology , Endothelial Cells/metabolism , Genetic Variation , Glycolysis , Humans , Inflammation , Lipolysis , Microvessels/metabolism , Oxygen Consumption , Postprandial Period , Protons , RNA, Small Interfering/metabolism , RNA-Seq , Reactive Oxygen Species/metabolism , Signal Transduction , Superoxides/metabolism
6.
Food Funct ; 9(12): 6245-6256, 2018 Dec 13.
Article in English | MEDLINE | ID: mdl-30402637

ABSTRACT

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.


Subject(s)
Chemokine CCL3/genetics , Interleukin-8/genetics , Lipid Droplets/metabolism , Monocytes/drug effects , Perilipin-2/genetics , Adult , Chemokine CCL3/metabolism , Down-Regulation , Female , Humans , Interleukin-8/metabolism , Lipid Droplets/chemistry , Lipolysis , Lipoproteins/metabolism , Male , Monocytes/metabolism , Perilipin-2/metabolism , Triglycerides/metabolism , Young Adult
7.
Toxicol Sci ; 163(1): 140-151, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29394414

ABSTRACT

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.


Subject(s)
Air Pollutants/toxicity , Cardiovascular Diseases/complications , Lung Injury/chemically induced , Lung/drug effects , Ozone/toxicity , Particulate Matter/toxicity , Animals , Cardiovascular Diseases/pathology , Inhalation Exposure , Lung/pathology , Lung Injury/complications , Lung Injury/pathology , Male , Ozone/administration & dosage , Ozone/chemistry , Particle Size , Particulate Matter/administration & dosage , Particulate Matter/chemistry , Rats, Inbred SHR , Rats, Inbred WKY
8.
PLoS One ; 13(2): e0191909, 2018.
Article in English | MEDLINE | ID: mdl-29444171

ABSTRACT

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.


Subject(s)
Blood-Brain Barrier , Brain/metabolism , Cognition , Diet, Western , Receptors, LDL/genetics , Animals , Mice , Mice, Inbred C57BL , Mice, Knockout
9.
PLoS One ; 12(1): e0169976, 2017.
Article in English | MEDLINE | ID: mdl-28081568

ABSTRACT

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.


Subject(s)
Lipopolysaccharides , Thrombophilia/chemically induced , Thrombophilia/physiopathology , Acute Disease , Animals , Biomarkers/blood , Blood Coagulation/drug effects , Blood Platelets/metabolism , Disease Models, Animal , Endothelial Cells/metabolism , Extracellular Vesicles/metabolism , Fibrin Fibrinogen Degradation Products/metabolism , Intercellular Adhesion Molecule-1/metabolism , Leukopenia/chemically induced , Male , MicroRNAs/blood , Neutrophils/metabolism , Neutrophils/pathology , Plasminogen Activator Inhibitor 1/metabolism , Rats , Rats, Wistar , Thrombophilia/immunology , Time Factors
10.
Am J Physiol Cell Physiol ; 312(4): C500-C516, 2017 Apr 01.
Article in English | MEDLINE | ID: mdl-28077357

ABSTRACT

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.


Subject(s)
Astrocytes/metabolism , Blood-Brain Barrier/physiology , Lipid Droplets/metabolism , Lipolysis/physiology , Lipoproteins/metabolism , Oxidative Stress/physiology , Triglycerides/metabolism , Animals , Cells, Cultured , Male , Mice , Mice, Inbred C57BL
11.
J Lipid Res ; 57(6): 955-68, 2016 06.
Article in English | MEDLINE | ID: mdl-27087439

ABSTRACT

Dysfunction of the cerebrovasculature plays an important role in vascular cognitive impairment (VCI). Lipotoxic injury of the systemic endothelium in response to hydrolyzed triglyceride-rich lipoproteins (TGRLs; TGRL lipolysis products) or a high-fat Western diet (WD) suggests similar mechanisms may be present in brain microvascular endothelium. We investigated the hypothesis that TGRL lipolysis products cause lipotoxic injury to brain microvascular endothelium by generating increased mitochondrial superoxide radical generation, upregulation of activating transcription factor 3 (ATF3)-dependent inflammatory pathways, and activation of cellular oxidative stress and apoptotic pathways. Human brain microvascular endothelial cells were treated with human TGRL lipolysis products that induced intracellular lipid droplet formation, mitochondrial superoxide generation, ATF3-dependent transcription of proinflammatory, stress response, and oxidative stress genes, as well as activation of proapoptotic cascades. Male apoE knockout mice were fed a high-fat/high-cholesterol WD for 2 months, and brain microvessels were isolated by laser capture microdissection. ATF3 gene transcription was elevated 8-fold in the hippocampus and cerebellar brain region of the WD-fed animals compared with chow-fed control animals. The microvascular injury phenotypes observed in vitro and in vivo were similar. ATF3 plays an important role in mediating brain microvascular responses to acute and chronic lipotoxic injury and may be an important preventative and therapeutic target for endothelial dysfunction in VCI.


Subject(s)
Activating Transcription Factor 3/genetics , Cerebrovascular Trauma/genetics , Cognitive Dysfunction/genetics , Inflammation/genetics , Lipoproteins/metabolism , Triglycerides/metabolism , Activating Transcription Factor 3/biosynthesis , Animals , Cerebellum/blood supply , Cerebellum/metabolism , Cerebellum/pathology , Cerebrovascular Trauma/metabolism , Cerebrovascular Trauma/physiopathology , Cognitive Dysfunction/metabolism , Cognitive Dysfunction/physiopathology , Diet, High-Fat/adverse effects , Diet, Western/adverse effects , Endothelium, Vascular/metabolism , Endothelium, Vascular/physiopathology , Hippocampus/blood supply , Hippocampus/metabolism , Hippocampus/pathology , Humans , Inflammation/metabolism , Inflammation/physiopathology , Mice , Oxidative Stress/genetics , Signal Transduction/genetics
12.
PLoS One ; 10(12): e0145523, 2015.
Article in English | MEDLINE | ID: mdl-26709509

ABSTRACT

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.


Subject(s)
Activating Transcription Factor 3/biosynthesis , Endothelial Cells/metabolism , Lipoproteins/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Triglycerides/metabolism , Active Transport, Cell Nucleus , Animals , Aorta/metabolism , Apoptosis , Caspase 3/metabolism , Cells, Cultured , Humans , Lipolysis , Lipoproteins/genetics , Mice , Phosphorylation , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction , Smad2 Protein/metabolism , Smad4 Protein/metabolism , Stress, Physiological , Transforming Growth Factor beta1/metabolism , Triglycerides/genetics , Tumor Suppressor Protein p53/metabolism
13.
Avian Dis ; 59(2): 344-8, 2015 Jun.
Article in English | MEDLINE | ID: mdl-26473689

ABSTRACT

In April 2014 an outbreak of low pathogenic avian influenza H5N8 North American genetic lineage was diagnosed in a commercial quail operation in Stanislaus County, California. Sudden increase in mortality prompted the submission of 20 Japanese quail hens (Coturnix c. japonica) to the California Animal Health and Food Safety Laboratory, Turlock Branch. Oropharyngeal and cloacal swabs tested positive for influenza A virus H5N8 by real-time reverse transcription-polymerase chain reaction. The virus was subsequently isolated. In vivo assay and sequencing of the hemagglutinin protein cleavage site classified the virus as a North American genetic lineage of low pathogenicity for chickens. Following the diagnosis, a rapid and coordinated response took place to contain the outbreak. The affected premise was depopulated, cleaned, and disinfected. Three areas from the affected premises-a 3 kilometer (km) radius (High Risk Zone), a 3-10 km area (Buffer Zone), and a 10-20 km (Surveillance Zone)-were established for avian influenza testing of commercial and noncommercial poultry operations. Surveillance testing and rapid control measures were successful in the control and eradication of the outbreak and revealed no area of spread of the virus from the index flock. This report describes the history, diagnosis, surveillance, and control measures applied to manage this outbreak.


Subject(s)
Coturnix , Influenza A virus/classification , Influenza in Birds/virology , Poultry Diseases/virology , Animals , California/epidemiology , Influenza in Birds/epidemiology , Poultry Diseases/epidemiology
14.
Toxicol Sci ; 143(2): 349-59, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25389146

ABSTRACT

Particulate matter (PM) exposure contributes to respiratory diseases and cardiopulmonary mortality. PM toxicity is related to sources and composition, such as abundance of polycyclic aromatic hydrocarbons (PAHs). We exposed adult male BALB/c mice, via oropharyngeal aspiration, to a range of doses of PM2.5 collected during the winter in downtown Sacramento near a major freeway interchange (SacPM). Two preparation methods (spin-down and multi-solvent extraction) were tested to remove particles from collection filters. Three doses were analyzed 24 h after treatment for (1) leukocytes and total protein in bronchoalveolar lavage fluid (BALF), (2) airway-specific and whole lobe expression of PAH-sensitive genes (CYP1B1 and CYP1A1) and IL-1 b, (3) lung histology, and (4) platelet function. Both extraction methods stimulated biological responses, but the spin-down method was more robust at producing IL-1 b and CYP1B1 gene responses and the multi-solvent extraction induced whole lung CYP1A1. Neutrophils in the BALF were increased 5- to 10-fold at the mid and high dose for both preparations. Histopathology scores indicated dose-dependent responses and increased pathology associated with spin-down-derived PM exposure. In microdissected airways, spin-down PM increased CYP1B1 gene expression significantly, but multi-solvent extracted PM did not. Platelet responses to the physiological agonist thrombin were approximately twice as potent in the spin-down preparation as in the multi-solvent extract. We conclude (1) the method of filter extraction can influence the degree of biological response, (2) for SacPM the minimal effective dose is 27.5-50 µg based on neutrophil recruitment, and (3) P450s are upregulated differently in airways and lung parenchyma in response to PAH-containing PM.


Subject(s)
Blood Platelets/drug effects , Inhalation Exposure/adverse effects , Lung/drug effects , Particulate Matter/toxicity , Platelet Activation/drug effects , Animals , Blood Platelets/metabolism , Bronchoalveolar Lavage Fluid/chemistry , Bronchoalveolar Lavage Fluid/cytology , Dose-Response Relationship, Drug , Flow Cytometry , Immunohistochemistry , Inhalation Exposure/analysis , Lung/metabolism , Lung/pathology , Male , Mice, Inbred BALB C , Particle Size , Particulate Matter/isolation & purification , Proteins/metabolism , Solvents/chemistry , Urbanization
15.
Phys Chem Chem Phys ; 16(39): 21595-21601, 2014 Oct 21.
Article in English | MEDLINE | ID: mdl-25189785

ABSTRACT

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.


Subject(s)
Activating Transcription Factor 3/biosynthesis , Activating Transcription Factor 3/metabolism , Aorta/cytology , Aorta/metabolism , Cells, Cultured , Endothelial Cells/cytology , Endothelial Cells/metabolism , Humans , Lipolysis
16.
Arterioscler Thromb Vasc Biol ; 33(9): 2088-96, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23868936

ABSTRACT

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.


Subject(s)
Activating Transcription Factor 3/metabolism , Apoptosis , Endothelial Cells/metabolism , Inflammation/metabolism , Lipoproteins/metabolism , Triglycerides/metabolism , Activating Transcription Factor 3/deficiency , Activating Transcription Factor 3/genetics , Animals , Blotting, Western , Cells, Cultured , E-Selectin/metabolism , Endothelial Cells/immunology , Endothelial Cells/pathology , Enzyme Activation , Fluorescent Antibody Technique , Gene Expression Profiling/methods , Gene Expression Regulation , Humans , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Inflammation Mediators/metabolism , Interleukin-8/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Leukocytes, Mononuclear/metabolism , Lipolysis , Lipoprotein Lipase/metabolism , Lipoproteins/blood , Lipoproteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Oligonucleotide Array Sequence Analysis , Phosphorylation , RNA Interference , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transfection , Triglycerides/blood
17.
Toxicol Pathol ; 41(6): 880-92, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23129576

ABSTRACT

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.


Subject(s)
ATP-Binding Cassette Transporters/deficiency , Cardiomyopathy, Dilated/metabolism , Inflammation/metabolism , Lipoproteins/deficiency , ATP Binding Cassette Transporter, Subfamily G, Member 5 , ATP-Binding Cassette Transporters/genetics , Animals , Cardiomyopathy, Dilated/genetics , Cardiomyopathy, Dilated/pathology , Disease Models, Animal , Echocardiography , Female , Fibrosis/metabolism , Fibrosis/pathology , Histocytochemistry , Inflammation/genetics , Inflammation/pathology , Lipoproteins/genetics , Male , Mice , Mice, Transgenic , Microscopy, Electron , Monocytes/metabolism , Monocytes/pathology , Myocardium/chemistry , Myocardium/metabolism , Myocardium/pathology , Myofibrils/metabolism , Myofibrils/pathology , Phytosterols/pharmacology , T-Lymphocytes/metabolism , T-Lymphocytes/pathology
18.
J S C Med Assoc ; 109(2): 43-4, 2013 Jun.
Article in English | MEDLINE | ID: mdl-24902389

ABSTRACT

A returning traveler presenting with fever accompanied by abdominal "pressure" and pain proved to have amebic appendicitis, amebic liver abscess, and probable recent amebic dysentery--a rare combination of findings amply illustrating the value of asking "Unde venis--from where do you come?"


Subject(s)
Amebiasis/diagnosis , Amebiasis/therapy , Appendicitis/parasitology , Adult , Amebicides/therapeutic use , Appendectomy , Combined Modality Therapy , Diagnosis, Differential , Humans , Laparoscopy/methods , Male
19.
Inhal Toxicol ; 24(8): 506-17, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22746400

ABSTRACT

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.


Subject(s)
Air Pollutants/toxicity , Cytokines/blood , Gene Expression Regulation/drug effects , Particulate Matter/toxicity , Platelet Activation/drug effects , Seasons , Animals , California , Environmental Monitoring , Gene Expression Profiling , Inhalation Exposure , Lung/blood supply , Lung/drug effects , Lung/immunology , Lung/metabolism , Male , Mice , Mice, Inbred C57BL , Particle Size
20.
Physiol Genomics ; 43(15): 917-29, 2011 Aug 16.
Article in English | MEDLINE | ID: mdl-21652769

ABSTRACT

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.


Subject(s)
Endothelial Cells/drug effects , Endothelial Cells/metabolism , Particulate Matter/toxicity , Apoptosis/drug effects , Cell Survival/drug effects , Cells, Cultured , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Chemokine CXCL2/genetics , Cytochrome P-450 CYP1A1/genetics , Cytochrome P-450 Enzyme System/genetics , E-Selectin/genetics , Humans , Interleukin-6/metabolism , Oligonucleotide Array Sequence Analysis , Polymerase Chain Reaction , Receptors, Aryl Hydrocarbon/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...