Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters

Country/Region as subject
Affiliation country
Publication year range
1.
Cytokine ; 89: 98-104, 2017 01.
Article in English | MEDLINE | ID: mdl-27599390

ABSTRACT

Cancer metastasis to the lymph nodes is indicative of a poor prognosis. An endobronchial ultrasound-guided fine needle aspiration (EBUS-FNA) biopsy is increasingly being used to sample paratracheal lymph nodes for simultaneous cancer diagnosis and staging. In this prospective, single-center study, we collected dedicated EBUS-FNA biopsies from 27 patients with enlarged paratracheal and hilar lymph nodes. Cytokines were assayed using Bio-Plex Pro human cancer biomarker panels (34 cytokines), in a Bio-Rad 200 suspension array system. A mean cytokine value was taken from each subject with more than 1 lymph node station EBUS-FNA biopsies. Malignant and benign histologic diagnoses were established in 16 and 12 patients, respectively. An initial analysis using the Kruskal-Wallis test with Sidak correction for multiple comparisons, showed significant elevation of sVEGFR-1, IL-6, VEGF-A, Angiopoeintin-2, uPA, sHER-2/neu and PLGF in malignant lymph node samples compared to benign samples. The univariate logistic regression analyses revealed that 6 cytokines were significant predictors and 1 cytokine (PLGF) was marginally significant for discrimination between benign and malignant samples. The prediction power of these cytokines as biomarkers were very high according to the area under the ROC curve. Multiple logistic regression for subsets of the seven cytokine combined; provided an almost complete discrimination between benign and malignant samples (AUC=0.989). For screening and diagnostic purposes, we presented the optimal discrimination cut-off for each cytokine: sVEGFR-1 (2124.5pg/mL), IL-6 (40.2pg/mL), VEGF-A (1060.1pg/mL), Angiopoeintin-2 (913.7pg/mL), uPA (248.1pg/mL), sHER-2/neu (5010pg/mL) and PLGF (93.4pg/mL). For the very first time, a novel cytokine profile associated with cancer metastasis to the paratracheal lymph nodes were reported.


Subject(s)
Cytokines/metabolism , Mediastinal Neoplasms/metabolism , Mediastinal Neoplasms/secondary , Neoplasm Proteins/metabolism , Aged , Aged, 80 and over , Biopsy, Fine-Needle , Female , Humans , Lymph Nodes/metabolism , Lymph Nodes/pathology , Lymphatic Metastasis , Male , Mediastinal Neoplasms/pathology , Mediastinum/pathology , Middle Aged , Pilot Projects , Prospective Studies
2.
Kidney Int ; 88(4): 804-14, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25993322

ABSTRACT

A systems biology approach was used to comprehensively examine the impact of renal disease and hemodialysis (HD) on patient response during critical illness. To achieve this, we examined the metabolome, proteome, and transcriptome of 150 patients with critical illness, stratified by renal function. Quantification of plasma metabolites indicated greater change as renal function declined, with the greatest derangements in patients receiving chronic HD. Specifically, 6 uremic retention molecules, 17 other protein catabolites, 7 modified nucleosides, and 7 pentose phosphate sugars increased as renal function declined, consistent with decreased excretion or increased catabolism of amino acids and ribonucleotides. Similarly, the proteome showed increased levels of low-molecular-weight proteins and acute-phase reactants. The transcriptome revealed a broad-based decrease in mRNA levels among patients on HD. Systems integration revealed an unrecognized association between plasma RNASE1 and several RNA catabolites and modified nucleosides. Further, allantoin, N1-methyl-4-pyridone-3-carboxamide, and N-acetylaspartate were inversely correlated with the majority of significantly downregulated genes. Thus, renal function broadly affected the plasma metabolome, proteome, and peripheral blood transcriptome during critical illness; changes were not effectively mitigated by hemodialysis. These studies allude to several novel mechanisms whereby renal dysfunction contributes to critical illness.


Subject(s)
Acute Kidney Injury/blood , Blood Proteins/metabolism , Kidney/metabolism , RNA, Messenger/blood , Systemic Inflammatory Response Syndrome/blood , Systems Biology , Acute Kidney Injury/diagnosis , Acute Kidney Injury/genetics , Acute Kidney Injury/physiopathology , Acute Kidney Injury/therapy , Adult , Aged , Aged, 80 and over , Biomarkers/blood , Critical Illness , Female , Gene Expression Profiling , Gene Expression Regulation , Humans , Kidney/physiopathology , Kidney Function Tests , Male , Metabolomics , Middle Aged , Proteomics , Renal Dialysis , Systemic Inflammatory Response Syndrome/diagnosis , Systemic Inflammatory Response Syndrome/genetics , Systemic Inflammatory Response Syndrome/therapy , Systems Integration , Time Factors , Treatment Outcome , United States
3.
Am J Respir Crit Care Med ; 190(4): 445-55, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-25054455

ABSTRACT

RATIONALE: Sepsis is a leading cause of morbidity and mortality. Currently, early diagnosis and the progression of the disease are difficult to make. The integration of metabolomic and transcriptomic data in a primate model of sepsis may provide a novel molecular signature of clinical sepsis. OBJECTIVES: To develop a biomarker panel to characterize sepsis in primates and ascertain its relevance to early diagnosis and progression of human sepsis. METHODS: Intravenous inoculation of Macaca fascicularis with Escherichia coli produced mild to severe sepsis, lung injury, and death. Plasma samples were obtained before and after 1, 3, and 5 days of E. coli challenge and at the time of killing. At necropsy, blood, lung, kidney, and spleen samples were collected. An integrative analysis of the metabolomic and transcriptomic datasets was performed to identify a panel of sepsis biomarkers. MEASUREMENTS AND MAIN RESULTS: The extent of E. coli invasion, respiratory distress, lethargy, and mortality was dependent on the bacterial dose. Metabolomic and transcriptomic changes characterized severe infections and death, and indicated impaired mitochondrial, peroxisomal, and liver functions. Analysis of the pulmonary transcriptome and plasma metabolome suggested impaired fatty acid catabolism regulated by peroxisome-proliferator activated receptor signaling. A representative four-metabolite model effectively diagnosed sepsis in primates (area under the curve, 0.966) and in two human sepsis cohorts (area under the curve, 0.78 and 0.82). CONCLUSIONS: A model of sepsis based on reciprocal metabolomic and transcriptomic data was developed in primates and validated in two human patient cohorts. It is anticipated that the identified parameters will facilitate early diagnosis and management of sepsis.


Subject(s)
Bacteremia/blood , Bacteremia/diagnosis , Metabolomics/methods , Systemic Inflammatory Response Syndrome/blood , Systemic Inflammatory Response Syndrome/diagnosis , Transcriptome/physiology , Animals , Biomarkers/blood , Cohort Studies , Disease Models, Animal , Early Diagnosis , Female , Humans , Macaca , Male
4.
Toxicol Appl Pharmacol ; 274(2): 200-8, 2014 Jan 15.
Article in English | MEDLINE | ID: mdl-24269878

ABSTRACT

Sarin is an organophosphate nerve agent that is among the most lethal chemical toxins known to mankind. Because of its vaporization properties and ease and low cost of production, sarin is the nerve agent with a strong potential for use by terrorists and rouge nations. The primary route of sarin exposure is through inhalation and, depending on the dose, sarin leads to acute respiratory failure and death. The mechanism(s) of sarin-induced respiratory failure is poorly understood. Sarin irreversibly inhibits acetylcholine esterase, leading to excessive synaptic levels of acetylcholine and, we have previously shown that sarin causes marked ventilatory changes including weakened response to hypoxia. We now show that LD50 sarin inhalation causes severe bronchoconstriction in rats, leading to airway resistance, increased hypoxia-induced factor-1α, and severe lung epithelium injury. Transferring animals into 60% oxygen chambers after sarin exposure improved the survival from about 50% to 75% at 24h; however, many animals died within hours after removal from the oxygen chambers. On the other hand, if LD50 sarin-exposed animals were administered the bronchodilator epinephrine, >90% of the animals survived. Moreover, while both epinephrine and oxygen treatments moderated cardiorespiratory parameters, the proinflammatory cytokine surge, and elevated expression of hypoxia-induced factor-1α, only epinephrine consistently reduced the sarin-induced bronchoconstriction. These data suggest that severe bronchoconstriction is a critical factor in the mortality induced by LD50 sarin inhalation, and epinephrine may limit the ventilatory, inflammatory, and lethal effects of sarin.


Subject(s)
Bronchoconstriction/drug effects , Chemical Warfare Agents/toxicity , Epinephrine/pharmacology , Lung Diseases/drug therapy , Oxygen/pharmacology , Sarin/toxicity , Acute Disease , Administration, Inhalation , Airway Resistance/drug effects , Animals , Cholinesterase Inhibitors/toxicity , Dose-Response Relationship, Drug , Enzyme Precursors/metabolism , Gelatinases/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Lethal Dose 50 , Lung/drug effects , Lung/pathology , Lung Diseases/chemically induced , Male , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Rats , Rats, Inbred F344 , Sarin/administration & dosage
5.
J Immunol ; 187(9): 4542-52, 2011 Nov 01.
Article in English | MEDLINE | ID: mdl-21930963

ABSTRACT

Parental, particularly maternal, smoking increases the risk for childhood allergic asthma and infection. Similarly, in a murine allergic asthma model, prenatal plus early postnatal exposure to secondhand cigarette smoke (SS) exacerbates airways hyperreactivity and Th2 responses in the lung. However, the mechanism and contribution of prenatal versus early postnatal SS exposure on allergic asthma remain unresolved. To identify the effects of prenatal and/or early postnatal SS on allergic asthma, BALB/c dams and their offspring were exposed gestationally and/or 8-10 wk postbirth to filtered air or SS. Prenatal, but not postnatal, SS strongly increased methacholine and allergen (Aspergillus)-induced airway resistance, Th2 cytokine levels, and atopy and activated the Th2-polarizing pathway GATA3/Lck/ERK1/2/STAT6. Either prenatal and/or early postnatal SS downregulated the Th1-specific transcription factor T-bet and, surprisingly, despite high levels of IL-4/IL-13, dramatically blocked the allergen-induced mucous cell metaplasia, airway mucus formation, and the expression of mucus-related genes/proteins: Muc5ac, γ-aminobutyric acid A receptors, and SAM pointed domain-containing Ets-like factor. Given that SS/nicotine exposure of normal adult mice promotes mucus formation, the results suggested that fetal and neonatal lung are highly sensitive to cigarette smoke. Thus, although the gestational SS promotes Th2 polarization/allergic asthma, it may also impair and/or delay the development of fetal and neonatal lung, affecting mucociliary clearance and Th1 responses. Together, this may explain the increased susceptibility of children from smoking parents to allergic asthma and childhood respiratory infections.


Subject(s)
Cell Differentiation/immunology , Cell Polarity/immunology , Goblet Cells/immunology , Mucus/immunology , Prenatal Exposure Delayed Effects/immunology , Respiratory Mucosa/immunology , Th2 Cells/immunology , Tobacco Smoke Pollution/adverse effects , Animals , Cell Differentiation/drug effects , Cell Polarity/drug effects , Down-Regulation/immunology , Female , Goblet Cells/drug effects , Goblet Cells/pathology , Humans , Male , Mice , Mice, Inbred BALB C , Mucus/metabolism , Pregnancy , Respiratory Mucosa/embryology , Respiratory Mucosa/pathology , Risk Factors , Th2 Cells/drug effects , Th2 Cells/pathology , Up-Regulation/immunology
6.
J Allergy Clin Immunol ; 130(3): 770-780.e11, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22578901

ABSTRACT

BACKGROUND: Airway mucus hypersecretion is a key pathophysiologic feature in a number of lung diseases. Cigarette smoke/nicotine and allergens are strong stimulators of airway mucus; however, the mechanism of mucus modulation is unclear. OBJECTIVES: We sought to characterize the pathway by which cigarette smoke/nicotine regulates airway mucus and identify agents that decrease airway mucus. METHODS: IL-13 and γ-aminobutyric acid type A receptors (GABA(A)Rs) are implicated in airway mucus. We examined the role of IL-13 and GABA(A)Rs in nicotine-induced mucus formation in normal human bronchial epithelial (NHBE) and A549 cells and secondhand cigarette smoke-induced, ovalbumin-induced, or both mucus formation in vivo. RESULTS: Nicotine promotes mucus formation in NHBE cells; however, the nicotine-induced mucus formation is independent of IL-13 but sensitive to the GABA(A)R antagonist picrotoxin. Airway epithelial cells express α7-, α9-, and α10-nicotinic acetylcholine receptors (nAChRs), and specific inhibition or knockdown of α7- but not α9/α10-nAChRs abrogates mucus formation in response to nicotine and IL-13. Moreover, addition of acetylcholine or inhibition of its degradation increases mucus in NHBE cells. Nicotinic but not muscarinic receptor antagonists block allergen- or nicotine/cigarette smoke-induced airway mucus formation in NHBE cells, murine airways, or both. CONCLUSIONS: Nicotine-induced airway mucus formation is independent of IL-13, and α7-nAChRs are critical in airway mucous cell metaplasia/hyperplasia and mucus production in response to various promucoid agents, including IL-13. In the absence of nicotine, acetylcholine might be the biological ligand for α7-nAChRs to trigger airway mucus formation. α7-nAChRs are downstream of IL-13 but upstream of GABA(A)Rα2 in the MUC5AC pathway. Acetylcholine and α7-nAChRs might serve as therapeutic targets to control airway mucus.


Subject(s)
Acetylcholine/physiology , Bronchi/metabolism , Bronchi/pathology , Mucus/physiology , Receptors, Nicotinic/physiology , Epithelial Cells/pathology , Humans , Hyperplasia , Interleukin-13/pharmacology , Metaplasia , Mucus/cytology , Nicotine/pharmacology , Receptors, GABA-A/physiology , alpha7 Nicotinic Acetylcholine Receptor
7.
J Immunol ; 185(1): 588-96, 2010 Jul 01.
Article in English | MEDLINE | ID: mdl-20505147

ABSTRACT

Smokers are less likely to develop some inflammatory and allergic diseases. In Brown-Norway rats, nicotine inhibits several parameters of allergic asthma, including the production of Th2 cytokines and the cysteinyl leukotriene LTC(4). Cysteinyl leukotrienes are primarily produced by mast cells, and these cells play a central role in allergic asthma. Mast cells express a high-affinity receptor for IgE (FcepsilonRI). Following its cross-linking, cells degranulate and release preformed inflammatory mediators (early phase) and synthesize and secrete cytokines/chemokines and leukotrienes (late phase). The mechanism by which nicotine modulates mast cell activation is unclear. Using alpha-bungarotoxin binding and quantitative PCR and PCR product sequencing, we showed that the rat mast/basophil cell line RBL-2H3 expresses nicotinic acetylcholine receptors (nAChRs) alpha7, alpha9, and alpha10; exposure to exceedingly low concentrations of nicotine (nanomolar), but not the biologically inactive metabolite cotinine, for > or = 8 h suppressed the late phase (leukotriene/cytokine production) but not degranulation (histamine and hexosaminidase release). These effects were unrelated to those of nicotine on intracellular free calcium concentration but were causally associated with the inhibition of cytosolic phospholipase A(2) activity and the PI3K/ERK/NF-kappaB pathway, including phosphorylation of Akt and ERK and nuclear translocation of NF-kappaB. The suppressive effect of nicotine on the late-phase response was blocked by the alpha7/alpha9-nAChR antagonists methyllycaconitine and alpha-bungarotoxin, as well as by small interfering RNA knockdown of alpha7-, alpha9-, or alpha10-nAChRs, suggesting a functional interaction between alpha7-, alpha9-, and alpha10-nAChRs that might explain the response of RBL cells to nanomolar concentrations of nicotine. This "hybrid" receptor might serve as a target for novel antiallergic/antiasthmatic therapies.


Subject(s)
Cell Degranulation/immunology , Cysteine/antagonists & inhibitors , Cytokines/antagonists & inhibitors , Mast Cells/metabolism , Nicotine/pharmacology , Receptors, IgE/antagonists & inhibitors , Receptors, Nicotinic/physiology , Animals , Basophils/drug effects , Basophils/immunology , Basophils/metabolism , Cell Degranulation/drug effects , Cell Line, Tumor , Cysteine/biosynthesis , Cytokines/biosynthesis , Cytosol/drug effects , Cytosol/enzymology , Cytosol/immunology , Down-Regulation/drug effects , Down-Regulation/immunology , Leukotrienes/biosynthesis , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/immunology , Mast Cells/drug effects , Mast Cells/immunology , Phospholipase A2 Inhibitors , Phospholipases A2/physiology , Rats , Rats, Inbred BN , Receptors, IgE/physiology , Tobacco Smoke Pollution/adverse effects , alpha7 Nicotinic Acetylcholine Receptor
8.
J Immunol ; 183(3): 2115-21, 2009 Aug 01.
Article in English | MEDLINE | ID: mdl-19596983

ABSTRACT

Airway hyperreactivity (AHR), lung inflammation, and atopy are clinical signs of allergic asthma. Gestational exposure to cigarette smoke (CS) markedly increases the risk for childhood allergic asthma. Muscarinic receptors regulate airway smooth muscle tone, and asthmatics exhibit increased AHR to muscarinic agonists. We have previously reported that in a murine model of bronchopulmonary aspergillosis, maternal exposure to mainstream CS increases AHR after acute intratracheal administration of Aspergillus fumigatus extract. However, the mechanism by which gestational CS induces allergic asthma is unclear. We now show for the first time that, compared with controls, mice exposed prenatally to secondhand CS exhibit increased lung inflammation (predominant infiltration by eosinophils and polymorphs), atopy, and airway resistance, and produce proinflammatory cytokines (IL-4, IL-5, IL-6, and IL-13, but not IL-2 or IFN-gamma). These changes, which occur only after an allergen (A. fumigatus extract) treatment, are correlated with marked up-regulated lung expression of M1, M2, and M3 muscarinic receptors and phosphodiesterase (PDE)4D5 isozyme. Interestingly, the PDE4-selective inhibitor rolipram attenuates the increase in AHR, muscarinic receptors, and PDE4D5, but fails to down-regulate lung inflammation, Th2 cytokines, or serum IgE levels. Thus, the fetus is extraordinarily sensitive to CS, inducing allergic asthma after postnatal exposure to allergens. Although the increased AHR might reflect increased PDE4D5 and muscarinic receptor expression, the mechanisms underlying atopy and lung inflammation are unrelated to the PDE4 activity. Thus, PDE4 inhibitors might ease AHR, but are unlikely to attenuate lung inflammation and atopy associated with childhood allergic asthma.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 4/immunology , Maternal Exposure , Prenatal Exposure Delayed Effects , Th2 Cells/immunology , Tobacco Smoke Pollution/adverse effects , Airway Resistance , Allergens/adverse effects , Animals , Bronchial Hyperreactivity , Cyclic Nucleotide Phosphodiesterases, Type 4/genetics , Cytokines/analysis , Female , Humans , Isoenzymes/genetics , Isoenzymes/immunology , Mice , Pneumonia/etiology , Pregnancy , Receptors, Muscarinic/analysis , Rolipram/pharmacology
9.
J Toxicol Environ Health A ; 74(19): 1261-79, 2011.
Article in English | MEDLINE | ID: mdl-21830856

ABSTRACT

Silicosis, a fibrotic granulomatous lung disease, may occur through accidental high-dose or occupational inhalation of silica, leading to acute/accelerated and chronic silicosis, respectively. While chronic silicosis has a long asymptomatic latency, lung inflammation and apoptosis are hallmarks of acute silicosis. In animal models, histiocytic granulomas develop within days after high-dose intratracheal (IT) silica instillation. However, following chronic inhalation of occupationally relevant doses of silica, discrete granulomas resembling human silicosis arise months after the final exposure without significant lung inflammation/apoptosis. To identify molecular events associated with chronic silicosis, lung RNA samples from controls or subchronic silica-exposed rats were analyzed by Affymetrix at 28 wk after silica exposures. Results suggested a significant upregulation of 144 genes and downregulation of 7 genes. The upregulated genes included complement cascade, chemokines/chemokine receptors, G-protein signaling components, metalloproteases, and genes associated with oxidative stress. To examine the kinetics of gene expression relevant to silicosis, quantitative polymerase chain reaction (qPCR), enzyme-linked immunosorbent assay (ELISA), Luminex-bead assays, Western blotting, and/or zymography were performed on lung tissues from 4 d, 28 wk, and intermediate times after subchronic silica exposure and compared with 14-d acute silicosis samples. Results indicated that genes regulating fibrosis (secreted phosphoprotein-1, Ccl2, and Ccl7), redox enzymes (superoxide dismutase-2 and arginase-1), and the enzymatic activities of matrix metalloproteinases 2 and 9 were upregulated in acute and chronic silicosis models. However, proinflammatory cytokines were strongly upregulated only in acute silicosis. Thus, inflammatory cytokines are associated with acute but not chronic silicosis. Data suggest that genes regulating fibrosis, oxidative stress, and metalloproteases may contribute to both acute and chronic silicosis.


Subject(s)
Lung/drug effects , Lung/metabolism , Oxidative Stress/drug effects , Silicosis/metabolism , Silicosis/pathology , Up-Regulation/drug effects , Animals , Arginase/genetics , Arginase/metabolism , Disease Models, Animal , Fibrosis , Gelatinases/genetics , Gelatinases/metabolism , Gene Expression Profiling , Lung/immunology , Lung/pathology , Male , Monocyte Chemoattractant Proteins/genetics , Monocyte Chemoattractant Proteins/metabolism , Oligonucleotide Array Sequence Analysis , Osteopontin/genetics , Osteopontin/metabolism , RNA, Messenger/metabolism , Rats , Rats, Inbred Lew , Silicosis/immunology , Specific Pathogen-Free Organisms , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism
10.
J Toxicol Environ Health A ; 73(10): 669-83, 2010.
Article in English | MEDLINE | ID: mdl-20391111

ABSTRACT

Chronic human silicosis results primarily from continued occupational exposure to silica and exhibits a long asymptomatic latency. Similarly, continued exposure of Lewis rats to low doses of silica is known to cause delayed granuloma formation with limited lung inflammation and injury. On the other hand, intratracheal exposure to large doses of silica induces acute silicosis characterized by granuloma-like formations in the lung associated with apoptosis, severe alveolitis, and alveolar lipoproteinosis. To ascertain similarities/differences between acute and chronic silicosis, in this communication, we compared cellular and molecular changes in established rat models of acute and chronic silicosis. In Lewis rats, acute silicosis was induced by intratracheal instillation of 35 mg silica, and chronic silicosis through inhalation of aerosolized silica (6.2 mg/m(3), 5 d/wk for 6 wk). Animals exposed to acute high-dose silica were sacrificed at 14 d after silica instillation while chronically silica-treated animals were sacrificed between 4 d and 28 wk after silica exposure. The lung granulomas formation in acute silicosis was associated with strong inflammation, presence of TUNEL-positive cells, and increases in caspase-3 activity and other molecular markers of apoptosis. On the other hand, lungs from chronically silica-exposed animals exhibited limited inflammation and increased expression of anti-apoptotic markers, including dramatic increases in Bcl-2 and procaspase-3, and lower caspase-3 activity. Moreover, chronic silicotic lungs were TUNEL-negative and overexpressed Bcl-3 and NF-kappaB-p50 but not NF-kappaB-p65 subunits. These results suggest that, unlike acute silicosis, chronic exposures to occupationally relevant doses of silica cause significantly lower lung inflammation and elevated expression of anti-apoptotic rather than proapoptotic markers in the lung that might result from interaction between NF-kappaB-p50 and Bcl-3.


Subject(s)
Apoptosis , Granuloma, Respiratory Tract/pathology , Lung/pathology , Silicon Dioxide/toxicity , Silicosis/pathology , Acute Disease , Animals , B-Cell Lymphoma 3 Protein , Biomarkers/analysis , Bronchoalveolar Lavage Fluid/chemistry , Bronchoalveolar Lavage Fluid/cytology , Caspase 3/metabolism , Chronic Disease , Disease Models, Animal , Dose-Response Relationship, Drug , Granuloma, Respiratory Tract/chemically induced , Granuloma, Respiratory Tract/metabolism , In Situ Nick-End Labeling , Inhalation Exposure , Intubation, Intratracheal , Lung/drug effects , Lung/metabolism , Male , NF-kappa B p50 Subunit/genetics , NF-kappa B p50 Subunit/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Rats , Rats, Inbred Lew , Silicosis/etiology , Silicosis/metabolism , Specific Pathogen-Free Organisms , Transcription Factors/genetics , Transcription Factors/metabolism , Up-Regulation/drug effects
11.
Front Immunol ; 11: 1628, 2020.
Article in English | MEDLINE | ID: mdl-32849552

ABSTRACT

Rationale: Gestational cigarette smoke (CS) impairs lung angiogenesis and alveolarization, promoting transgenerational development of asthma and bronchopulmonary dysplasia (BPD). Hydrogen sulfide (H2S), a proangiogenic, pro-alveolarization, and anti-asthmatic gasotransmitter is synthesized by cystathionine-γ-lyase (CSE), cystathionine-ß-synthase (CBS), and 3-mercaptopyruvate sulfur transferase (3MST). Objective: Determine if gestational CS exposure affected the expression of H2S synthesizing enzymes in the mouse lung and human placenta. Methods: Mice were exposed throughout gestational period to secondhand CS (SS) at approximating the dose of CS received by a pregnant woman sitting in a smoking bar for 3 h/days during pregnancy. Lungs from 7-days old control and SS-exposed pups and human placenta from mothers who were either non-smokers or smokers during pregnancy were analyzed for expression of the enzymes. Measurements: Mouse lungs and human placentas were examined for the expression of CSE, CBS, and 3MST by immunohistochemical staining, qRT-PCR and/or Western blot (WB) analyses. Results: Compared to controls, mouse lung exposed gestationally to SS had significantly lower levels of CSE, CBS, and 3MST. Moreover, the SS-induced suppression of CSE and CBS in F1 lungs was transmitted to the F2 generation without significant change in the magnitude of the suppression. These changes were associated with impaired epithelial-mesenchymal transition (EMT)-a process required for normal lung angiogenesis and alveolarization. Additionally, the placentas from mothers who smoked during pregnancy, expressed significantly lower levels of CSE, CBS, and 3MST, and the effects were partially moderated by quitting smoking during the first trimester. Conclusions: Lung H2S synthesizing enzymes are downregulated by gestational CS and the effects are transmitted to F2 progeny. Smoking during pregnancy decreases H2S synthesizing enzymes is human placentas, which may correlate with the increased risk of asthma/BPD in children.


Subject(s)
Gasotransmitters/biosynthesis , Hydrogen Sulfide/metabolism , Maternal Exposure/adverse effects , Prenatal Exposure Delayed Effects , Tobacco Smoking/adverse effects , Animals , Disease Models, Animal , Epithelial-Mesenchymal Transition , Female , Fluorescent Antibody Technique , Gene Expression Regulation, Enzymologic , Humans , Hydrogen Sulfide/adverse effects , Immunohistochemistry , Lung/metabolism , Lung/pathology , Maternal-Fetal Exchange , Mice , Models, Biological , Placenta/metabolism , Pregnancy
12.
J Neuroimmunol ; 194(1-2): 83-8, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18190972

ABSTRACT

Leukocytes contain both nicotinic and muscarinic receptors, and while activation of nicotinic receptors suppresses immune/inflammatory responses, the role of muscarinic receptors in immunity is unclear. We examined the effects of a muscarinic receptor antagonist (atropine) and agonist (oxotremorine), administered chronically through miniosmotic pumps, on immune/inflammatory responses in the rat. Results show that while oxotremorine stimulated, atropine inhibited the antibody and T-cell proliferative responses. Moreover, atropine also suppressed the turpentine-induced leukocytic infiltration and tissue injury, and inhibited chemotaxis of leukocytes toward neutrophil and monocyte/lymphocyte chemoattractants. Thus, activation of nicotinic and muscarinic receptors has opposite effects on the immune/inflammatory responses.


Subject(s)
Antibody Formation/physiology , Atropine/pharmacology , Inflammation/physiopathology , Lymphocyte Activation/physiology , Muscarinic Agonists/pharmacology , Muscarinic Antagonists/pharmacology , Oxotremorine/pharmacology , Receptors, Muscarinic/physiology , T-Lymphocytes/immunology , Abscess/chemically induced , Abscess/immunology , Animals , Antibody Formation/drug effects , Cells, Cultured/drug effects , Chemokine CCL2/pharmacology , Chemotaxis, Leukocyte/drug effects , Lymphocyte Activation/drug effects , Male , Rats , Rats, Inbred Lew , Receptors, Muscarinic/drug effects , Receptors, Nicotinic/physiology , Specific Pathogen-Free Organisms , T-Lymphocytes/drug effects , Turpentine/toxicity
13.
Toxicol Appl Pharmacol ; 232(3): 440-7, 2008 Nov 01.
Article in English | MEDLINE | ID: mdl-18706921

ABSTRACT

Sarin, a highly toxic nerve gas, is believed to cause bronchoconstriction and even death primarily through respiratory failure; however, the mechanism underlying the respiratory failure is not fully understood. The goals of this study were to ascertain whether sarin affects baseline ventilation (VE) and VE chemoreflexes as well as airway resistance and, if so, whether these changes are reversible. Four groups of F344 rats were exposed to vehicle (VEH) or sarin at 2.5, 3.5, and 4.0 mg h m(-3) (SL, SM, and SH, respectively). VE and VE responses to hypercapnia (7% CO2) or hypoxia (10% O2) were measured by plethysmography at 2 h and 1, 2, and 5 days after VEH or sarin exposure. Total pulmonary resistance (RL) also was measured in anesthetized VEH- and SH-exposed animals 2 h after exposure. Our results showed that within 2 h after exposure 11% of the SM- and 52% of the SH- exposed groups died. Although the SM and SH significantly decreased hypercapnic and hypoxic VE to similar levels (64 and 69%), SH induced greater respiratory impairment, characterized by lower baseline VE (30%; P<0.05), and total loss of the respiratory frequency response to hypercapnia and hypoxia. VE impairment recovered within 1-2 days after sarin exposure; interestingly, SH did not significantly affect baseline RL. Moreover, sarin induced body tremors that were unrelated to the changes in the VE responses. Thus, LC50 sarin causes a reversible impairment of VE that is not dependent on the sarin-induced body tremors and not associated with changes in RL.


Subject(s)
Chemical Warfare Agents/toxicity , Hypercapnia/physiopathology , Hypoxia/physiopathology , Respiration/drug effects , Sarin/toxicity , Airway Resistance/drug effects , Animals , Carbon Dioxide/blood , Inhalation Exposure , Male , Rats , Rats, Inbred F344
14.
Toxicol Sci ; 97(1): 181-8, 2007 May.
Article in English | MEDLINE | ID: mdl-17301067

ABSTRACT

Inhalation of subclinical doses of sarin suppresses the antibody-forming cell (AFC) response, T-cell mitogenesis, and serum corticosterone (CORT) levels, and high doses of sarin cause lung inflammation. However, the duration of these changes is not known. In these studies, rats were exposed to a subclinical dose of sarin (0.4 mg/m3/h/day) for 1 or 5 days, and immune and inflammatory parameters were assayed up to 8 weeks before sarin exposure. Our results showed that the effects of a 5-day sarin exposure on the AFC response and T-cell receptor (TCR)-mediated Ca2+ response disappeared within 2-4 weeks after sarin exposure, whereas the CORT and adrenocorticotropin hormone (ACTH) levels remained significantly decreased. Pretreatment of rats with chlorisondamine attenuated the effects of sarin on the AFC and the TCR-mediated Ca2+ response, implicating the autonomic nervous system (ANS) in the sarin-induced changes in T-cell function. Moreover, exposure to a single or five repeated subclinical doses of sarin upregulated the mRNA expression of proinflammatory cytokines in the lung, which is associated with the activation of NFkappaB in bronchoalveolar lavage cells. These effects were lost within 2 weeks of sarin inhalation. Our results suggest that while sarin-induced changes in T cells and cytokine gene expression were short lived, suppression of CORT and ACTH levels were relatively long lived and might represent biomarkers of sarin exposure. Moreover, while the effects of sarin on T-cell function were regulated by the ANS, the decreased CORT levels by sarin might result from its effects on the hypothalamus-pituitary-adrenal axis.


Subject(s)
Autonomic Nervous System/drug effects , Chemical Warfare Agents/toxicity , Cholinesterase Inhibitors/toxicity , Inflammation/chemically induced , Inhalation Exposure , Neuroimmunomodulation/drug effects , Neurosecretory Systems/drug effects , Sarin/toxicity , Adrenocorticotropic Hormone/blood , Animals , Autonomic Nervous System/metabolism , Biomarkers/blood , Calcium/metabolism , Chlorisondamine/pharmacology , Cholinesterase Inhibitors/administration & dosage , Corticosterone/blood , Cytokines/genetics , Cytokines/metabolism , Ganglionic Blockers/pharmacology , Hypothalamo-Hypophyseal System/drug effects , Hypothalamo-Hypophyseal System/metabolism , Inflammation/metabolism , Inflammation/pathology , Lung/drug effects , Lung/metabolism , Lung/pathology , Male , NF-kappa B/metabolism , Neurosecretory Systems/metabolism , Pituitary-Adrenal System/drug effects , Pituitary-Adrenal System/metabolism , RNA, Messenger/metabolism , Rats , Rats, Inbred F344 , Receptors, Antigen, T-Cell/drug effects , Receptors, Antigen, T-Cell/metabolism , Sarin/administration & dosage , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , Time Factors , Transcription, Genetic/drug effects
15.
Toxicol Lett ; 167(3): 231-7, 2006 Dec 15.
Article in English | MEDLINE | ID: mdl-17113252

ABSTRACT

Annually, approximately two million babies are exposed to cigarette smoke in utero and postnatally through cigarette smoking of their mothers. Exposure to mainstream cigarette smoke is known to impair both innate and adaptive immunities, and it has been hypothesized that the effects of in utero exposure to cigarette smoke on children's health might primarily stem from the adverse effects of cigarette smoke on the immune system. To simulate the environment that babies from smoking mothers encounter, we examined the effects of prenatal mainstream and postnatal sidestream cigarette smoke on spleen cell responses. Results show that postnatal exposure of newborn Balb/c mouse pups to sidestream cigarette smoke through the first 6 weeks of life strongly suppresses the antibody response of spleen cells to the T-cell-dependent antigen, sheep red blood cells. The reduction in the antibody response seen within 6 weeks of postnatal smoke exposure is much quicker than the published data on the time 25 weeks) required to establish reproducible immunosuppression in adult rats and mice. Moreover, the immunosuppression is not associated with significant changes in T-cell numbers or subset distribution. While the postnatal exposure to cigarette smoke did not affect the mitogenic response of T and B cells, the exposure inhibited the T cell receptor-mediated rise in the intracellular calcium concentration. These results suggest that the early postnatal period is highly sensitive to the immunosuppressive effects of environmental tobacco smoke, and the effects are causally associated with impaired antigen-mediated signaling in T cells.


Subject(s)
Antigens/immunology , Immune Tolerance , Nicotiana/adverse effects , Smoke/adverse effects , Spleen/immunology , T-Lymphocytes/immunology , Animals , Calcium/metabolism , Female , Fetus/immunology , Lymphocyte Activation , Male , Mice , Mice, Inbred BALB C , Receptors, Antigen, T-Cell/physiology
16.
PLoS One ; 10(9): e0137757, 2015.
Article in English | MEDLINE | ID: mdl-26361040

ABSTRACT

RATIONALE: Smoking during pregnancy increases the risk of bronchopulmonary dysplasia (BPD) and, in mice, gestational exposure to sidestream cigarette smoke (SS) induces BPD-like condition characterized by alveolar simplification, impaired angiogenesis, and suppressed surfactant protein production. Normal fetal development occurs in a hypoxic environment and nicotinic acetylcholine receptors (nAChRs) regulate the hypoxia-inducible factor (HIF)-1α that controls apoptosis and angiogenesis. To understand SS-induced BPD, we hypothesized that gestational SS affected alveolar development through HIF-1α. METHODS: Pregnant BALB/c mice were exposed to air (control) or SS throughout the gestational period and the 7-day-old lungs of the progeny were examined. RESULTS: Gestational SS increased apoptosis of alveolar and airway epithelial cells. This response was associated with increased alveolar volumes, higher levels of proapoptotic factors (FOXO3a, HIPK2, p53, BIM, BIK, and BAX) and the antiangiogenic factor (GAX), and lower levels of antiapoptotic factors (Akt-PI3K, NF-κB, HIF-1α, and Bcl-2) in the lung. Although gestational SS increased the cells containing the proangiogenic bombesin-like-peptide, it markedly decreased the expression of its receptor GRPR in the lung. The effects of SS on apoptosis were attenuated by the nAChR antagonist mecamylamine. CONCLUSIONS: Gestational SS-induced BPD is potentially regulated by nAChRs and associated with downregulation of HIF-1α, increased apoptosis of epithelial cells, and increased alveolar volumes. Thus, in mice, exposure to sidestream tobacco smoke during pregnancy promotes BPD-like condition that is potentially mediated through the nAChR/HIF-1α pathway.


Subject(s)
Bronchopulmonary Dysplasia/etiology , Bronchopulmonary Dysplasia/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Maternal Exposure/adverse effects , Smoking/adverse effects , Alveolar Epithelial Cells/metabolism , Angiogenesis Inhibitors/metabolism , Animals , Apoptosis , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Bronchopulmonary Dysplasia/physiopathology , Caspase 3/metabolism , Disease Models, Animal , Down-Regulation , Female , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Keratins/metabolism , Mecamylamine/pharmacology , Mice , NF-kappa B/metabolism , Pregnancy , Respiratory Mucosa/metabolism
17.
J Neuroimmunol ; 148(1-2): 140-5, 2004 Mar.
Article in English | MEDLINE | ID: mdl-14975594

ABSTRACT

Cholinergic compounds modulate the immune system; however, the mechanism of cholinergic immunotoxicity is largely unknown. Lewis rats were exposed chronically to cholinergic compounds via subcutaneous or intracerebroventricular routes. Compounds that crossed the blood-brain barrier (BBB) inhibited the antibody response when given by either route, however, poorly permeable compounds, unless given in high doses, inhibited the antibody response only by intracerebroventricular administration. Intracerebroventricular administration of cholinergic agents also reduced serum corticosterone levels, which along with the antibody response was attenuated by pretreatment with the ganglionic blocker chlorisondamine. Thus, cholinergic agents affect the neuroimmune communication and inhibit glucocorticoid production; the latter may be a biomarker for cholinergic toxicity.


Subject(s)
Chlorisondamine/pharmacology , Ganglionic Blockers/pharmacology , Immune System/drug effects , Nervous System Physiological Phenomena , Animals , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , Blood Cells/metabolism , Chlorisondamine/administration & dosage , Cholinesterase Inhibitors/pharmacology , Corticosterone/blood , Drug Administration Routes , Ganglionic Blockers/administration & dosage , Immunization , Immunosuppression Therapy , Infusion Pumps, Implantable , Male , Radioimmunoassay , Rats , Rats, Inbred Lew , Sheep , Spleen/cytology , Spleen/drug effects , T-Lymphocytes/drug effects
18.
PLoS One ; 8(10): e77160, 2013.
Article in English | MEDLINE | ID: mdl-24155926

ABSTRACT

Lung diseases such as chronic obstructive pulmonary disease (COPD), asthma, and lung infections are major causes of morbidity and mortality among HIV-infected patients even in the era of antiretroviral therapy (ART). Many of these diseases are strongly associated with smoking and smoking is more common among HIV-infected than uninfected people; however, HIV is an independent risk factor for chronic bronchitis, COPD, and asthma. The mechanism by which HIV promotes these diseases is unclear. Excessive airway mucus formation is a characteristic of these diseases and contributes to airway obstruction and lung infections. HIV gp120 plays a critical role in several HIV-related pathologies and we investigated whether HIV gp120 promoted airway mucus formation in normal human bronchial epithelial (NHBE) cells. We found that NHBE cells expressed the HIV-coreceptor CXCR4 but not CCR5 and produced mucus in response to CXCR4-tropic gp120. The gp120-induced mucus formation was blocked by the inhibitors of CXCR4, α7-nicotinic acetylcholine receptor (α7-nAChR), and γ-aminobutyric acid (GABA)AR but not the antagonists of CCR5 and epithelial growth factor receptor (EGFR). These results identify two distinct pathways (α7-nAChR-GABAAR and EGFR) for airway mucus formation and demonstrate for the first time that HIV-gp120 induces and regulates mucus formation in the airway epithelial cells through the CXCR4-α7-nAChR-GABAAR pathway. Interestingly, lung sections from HIV ± ART and simian immunodeficiency virus (SIV) ± ART have significantly more mucus and gp120-immunoreactivity than control lung sections from humans and macaques, respectively. Thus, even after ART, lungs from HIV-infected patients contain significant amounts of gp120 and mucus that may contribute to the higher incidence of obstructive pulmonary diseases in this population.


Subject(s)
Bronchi/pathology , Epithelial Cells/metabolism , HIV Envelope Protein gp120/metabolism , Mucus/metabolism , Receptors, CXCR4/metabolism , alpha7 Nicotinic Acetylcholine Receptor/metabolism , Animals , Antiretroviral Therapy, Highly Active , Bronchi/virology , Epithelial Cells/pathology , Epithelial Cells/virology , HIV Infections/metabolism , HIV Infections/pathology , HIV Infections/therapy , HIV Infections/virology , Humans , Macaca mulatta/virology , Mucin 5AC/metabolism , Receptors, CXCR5/metabolism , Receptors, GABA-A/metabolism , Simian Acquired Immunodeficiency Syndrome/metabolism , Simian Acquired Immunodeficiency Syndrome/pathology , Simian Acquired Immunodeficiency Syndrome/virology
19.
Environ Health Perspect ; 121(8): 957-64, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23757602

ABSTRACT

BACKGROUND: Cigarette smoke (CS) exposure during gestation may increase the risk of bronchopulmonary dysplasia (BPD)-a developmental lung condition primarily seen in neonates that is characterized by hypoalveolarization, decreased angiogenesis, and diminished surfactant protein production and may increase the risk of chronic obstructive pulmonary disease. OBJECTIVE: We investigated whether gestational exposure to secondhand CS (SS) induced BPD and sought to ascertain the role of nicotinic acetylcholine receptors (nAChRs) in this response. METHODS: We exposed BALB/c and C57BL/6 mice to filtered air (control) or SS throughout the gestation period or postnatally up to 10 weeks. Lungs were examined at 7 days, 10 weeks, and 8 months after birth. RESULTS: Gestational but not postnatal exposure to SS caused a typical BPD-like condition: suppressed angiogenesis [decreased vascular endothelial growth factor (VEGF), VEGF receptor, and CD34/CD31 (hematopoietic progenitor cell marker/endothelial cell marker)], irreversible hypoalveolarization, and significantly decreased levels of Clara cells, Clara cell secretory protein, and surfactant proteins B and C, without affecting airway ciliated cells. Importantly, concomitant exposure to SS and the nAChR antagonist mecamylamine during gestation blocked the development of BPD. CONCLUSIONS: Gestational exposure to SS irreversibly disrupts lung development leading to a BPD-like condition with hypoalveolarization, decreased angiogenesis, and diminished lung secretory function. Nicotinic receptors are critical in the induction of gestational SS-induced BPD, and the use of nAChR antagonists during pregnancy may block CS-induced BPD.


Subject(s)
Air Pollutants/toxicity , Bronchopulmonary Dysplasia/chemically induced , Lung/drug effects , Mecamylamine/metabolism , Nicotinic Antagonists/metabolism , Prenatal Exposure Delayed Effects/chemically induced , Tobacco Smoke Pollution/adverse effects , Air Pollutants/analysis , Animals , Bronchoalveolar Lavage Fluid/chemistry , Bronchopulmonary Dysplasia/pathology , Bronchopulmonary Dysplasia/physiopathology , Female , Lung/pathology , Lung/physiopathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Pregnancy , Prenatal Exposure Delayed Effects/pathology , Prenatal Exposure Delayed Effects/physiopathology , Pulmonary Alveoli/drug effects , Pulmonary Alveoli/pathology , Pulmonary Alveoli/physiopathology , RNA/analysis , Real-Time Polymerase Chain Reaction , Specific Pathogen-Free Organisms
20.
Int Immunopharmacol ; 13(1): 101-8, 2012 May.
Article in English | MEDLINE | ID: mdl-22465472

ABSTRACT

Sulfur mustard (SM) is a highly toxic chemical warfare agent that remains a threat to human health. The immediate symptoms of pulmonary distress may develop into chronic lung injury characterized by progressive lung fibrosis, the major cause of morbidity among the surviving SM victims. Although SM has been intensely investigated, little is known about the mechanism(s) by which SM induces chronic lung pathology. Increasing evidence suggests that IL-17(+) cells are critical in fibrosis, including lung fibrotic diseases. In this study we exposed F344 rats and cynomolgus monkeys to SM via inhalation and determined the molecular and cellular milieu in their lungs at various times after SM exposure. In rats, SM induced a burst of pro-inflammatory cytokines/chemokines within 72 h, including IL-1ß, TNF-α, IL-2, IL-6, CCL2, CCL3, CCL11, and CXCL1 that was associated with neutrophilic infiltration into the lung. At 2 wks and beyond (chronic phase), lymphocytic infiltration and continued elevated expression of cytokines/chemokines were sustained. TGF-ß, which was undetectable in the acute phase, was strongly upregulated in the chronic phase; these conditions persisted until the animals were sacrificed. The chronic phase was also associated with myofibroblast proliferation, collagen deposition, and presence of IL-17(+) cells. At ≥30 days, SM inhalation promoted the accumulation of IL-17(+) cells in the inflamed areas of monkey lungs. Thus, SM inhalation causes acute and chronic inflammatory responses; the latter is characterized by the presence of TGF-ß, fibrosis, and IL-17(+) cells in the lung. IL-17(+) cells likely play an important role in the pathogenesis of SM-induced lung injury.


Subject(s)
Chemical Warfare Agents/toxicity , Inhalation Exposure , Lung Injury/chemically induced , Mustard Gas/toxicity , Pulmonary Fibrosis/chemically induced , Th17 Cells/drug effects , Animals , Apoptosis/drug effects , Bronchoalveolar Lavage Fluid/cytology , Bronchoalveolar Lavage Fluid/immunology , Chemotaxis, Leukocyte/drug effects , Chemotaxis, Leukocyte/immunology , Cytokines/genetics , Female , Immunohistochemistry , In Situ Nick-End Labeling , Lung/drug effects , Lung/immunology , Lung/pathology , Lung Injury/immunology , Lung Injury/pathology , Lymphocyte Count , Macaca fascicularis , Pulmonary Fibrosis/immunology , Pulmonary Fibrosis/pathology , RNA, Messenger/genetics , Rats , Rats, Inbred F344 , Real-Time Polymerase Chain Reaction , Th17 Cells/immunology
SELECTION OF CITATIONS
SEARCH DETAIL