Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Lab Anim Res ; 36: 36, 2020.
Article in English | MEDLINE | ID: mdl-33042783

ABSTRACT

BACKGROUND: Anti-parasitics are frequently used in research animal facilities to treat a multitude of common infections, with pinworms and fur mites being amongst the most common. Ivermectin and selamectin are common oral and topical treatments for these infections, respectively. Although commonly thought to be innocuous to both the research animals and any transgenic elements that the animals may carry, evidence exists that ivermectin is capable of activating the recombinase activity of at least one CreER. The goal of the current study was to determine if there was an effect of either anti-parasitic agent on the activity of CreER proteins in transgenic mice. CASE PRESENTATION: We analyzed the offspring of transgenic mice exposed to either ivermectin or selamectin during pregnancy and nursing. Through analysis of reporter genes co-expressed with multiple, independently generated transgenic CreER drivers, we report here that ivermectin and selamectin both alter recombinase activity and thus may have unintended consequences on gene inactivation studies in mice. CONCLUSIONS: Although the mechanisms by which ivermectin and selamectin affect CreER activity in the offspring of treated dams remain unclear, the implications are important nonetheless. Treatment of pregnant transgenic mice with these anti-parasitics has the potential to alter transgene activity in the offspring. Special considerations should be made when planning treatment of transgenic mice with either of these pharmacologics.

2.
J Med Primatol ; 48(6): 374-377, 2019 12.
Article in English | MEDLINE | ID: mdl-31148179

ABSTRACT

Benign and malignant pulmonary tumors have been reported in both Old World and New World monkeys but are uncommon. Hemangiomas are also rarely reported in nonhuman primates. Here we present a case of two primary neoplasms (a papillary adenocarcinoma of bronchioloalveolar origin and multiple cavernous subcutaneous hemangiomas) arising in an aged squirrel monkey (Saimiri sciureus).


Subject(s)
Adenocarcinoma of Lung/veterinary , Hemangioma, Cavernous/veterinary , Lung Neoplasms/veterinary , Monkey Diseases/pathology , Saimiri , Adenocarcinoma of Lung/diagnosis , Adenocarcinoma of Lung/pathology , Animals , Diagnosis, Differential , Hemangioma, Cavernous/diagnosis , Hemangioma, Cavernous/pathology , Lung Neoplasms/diagnosis , Lung Neoplasms/pathology , Male , Monkey Diseases/diagnosis
3.
Am J Physiol Lung Cell Mol Physiol ; 301(1): L99-L109, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21478252

ABSTRACT

Both respiratory syncytial virus (RSV) and influenza A virus induce nucleotide/P2Y purinergic receptor-mediated impairment of alveolar fluid clearance (AFC), which contributes to formation of lung edema. Although genetically dissimilar, both viruses generate double-stranded RNA replication intermediates, which act as Toll-like receptor (TLR)-3 ligands. We hypothesized that double-stranded RNA/TLR-3 signaling underlies nucleotide-mediated inhibition of amiloride-sensitive AFC in both infections. We found that addition of the synthetic double-stranded RNA analog poly-inosinic-cytidylic acid [poly(I:C)] (500 ng/ml) to the AFC instillate resulted in nucleotide/P2Y purinergic receptor-mediated inhibition of amiloride-sensitive AFC in BALB/c mice but had no effect on cystic fibrosis transmembrane regulator (CFTR)-mediated Cl(-) transport. Poly(I:C) also induced acute keratinocyte cytokine-mediated AFC insensitivity to stimulation by the ß-adrenergic agonist terbutaline. Inhibitory effects of poly(I:C) on AFC were absent in TLR-3(-/-) mice and were not replicated by addition to the AFC instillate of ligands for other TLRs except TLR-2. Intranasal poly(I:C) administration (250 µg/mouse) similarly induced nucleotide-dependent AFC inhibition 2-3 days later, together with increased lung water content and neutrophilic inflammation. Intranasal treatment of BALB/c mice with poly(I:C) did not induce airway hyperresponsiveness at day 2 but did result in insensitivity to airway bronchodilation by ß-adrenergic agonists. These findings suggest that viral double-stranded RNA replication intermediates induce nucleotide-mediated impairment of amiloride-sensitive AFC in both infections, together with ß-adrenergic agonist insensitivity. Both of these effects also occur in RSV infection. However, double-stranded RNA replication intermediates do not appear to be sufficient to induce either adenosine-mediated, CFTR-dependent Cl(-) secretion in the lung or severe, lethal hypoxemia, both of which are features of influenza infection.


Subject(s)
Lung/physiopathology , Lung/virology , RNA, Double-Stranded/pharmacology , Respiratory Syncytial Virus Infections/physiopathology , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Viruses/physiology , Administration, Intranasal , Adrenergic beta-Agonists/pharmacology , Animals , Body Fluids/drug effects , Inhalation Exposure , Ligands , Lung/drug effects , Lung Injury/complications , Lung Injury/pathology , Lung Injury/physiopathology , Mice , Mice, Inbred BALB C , Poly I-C/administration & dosage , Poly I-C/pharmacology , Pulmonary Alveoli/drug effects , Pulmonary Alveoli/metabolism , Receptors, Purinergic P2Y/metabolism , Respiratory Syncytial Virus Infections/complications , Respiratory Syncytial Viruses/drug effects , Time Factors , Toll-Like Receptor 3/metabolism
4.
Am J Physiol Lung Cell Mol Physiol ; 298(3): L437-45, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19965982

ABSTRACT

beta-Adrenergic agonists (beta-agonists) are commonly used to treat respiratory syncytial virus (RSV) bronchiolitis but are generally ineffective for unknown reasons. We have previously shown that RSV strain A2 inhibits bronchoalveolar epithelial responses to beta-agonists in a BALB/c mouse model by inducing heterologous keratinocyte cytokine (KC)/CXCR2-mediated desensitization of epithelial beta(2)-adrenergic receptors. The aim of the current study was to determine whether RSV also induces airway insensitivity to beta-agonists. Total lung resistance (R) was measured in anesthetized female BALB/c mice undergoing mechanical ventilation on a flexiVent computer-controlled piston ventilator. Data were analyzed using the single-compartment model. Infection with RSV A2 did not induce airway hyperresponsiveness to increasing doses of the nebulized cholinergic agonist methacholine (MCh) at any time point after RSV infection. Prenebulization with the beta-agonist terbutaline (100 muM) significantly attenuated bronchoconstrictive responses to 20 and 50 mg/ml MCh in uninfected mice and in mice infected with RSV 4-8 days postinfection (d.p.i.). However, in mice infected with replication-competent, but not UV-inactivated, RSV for 2 days, significant terbutaline insensitivity was found. Terbutaline insensitivity at 2 d.p.i. could be reversed by systemic preinfection treatment with neutralizing anti-CXCR2 antibodies, which reduced bronchoalveolar lavage (BAL) neutrophil counts but did not alter viral replication, BAL KC levels, or lung edema. Terbutaline insensitivity was also reversed by postinfection nebulization with neutralizing anti-KC or anti-CXCR2 antibodies and could be replicated in normal, uninfected mice by nebulization with recombinant KC. These data suggest that KC/CXCR2-mediated airway insensitivity to beta-agonists may underlie the modest utility of these drugs as bronchodilators in therapy for acute RSV bronchiolitis.


Subject(s)
Adrenergic beta-Agonists/pharmacology , Lung/immunology , Lung/virology , Respiratory Syncytial Viruses/physiology , Aerosols/pharmacology , Animals , Chemokines/pharmacology , Humans , Lung/drug effects , Methacholine Chloride/pharmacology , Mice , Mice, Inbred BALB C , Nebulizers and Vaporizers , Receptors, Interleukin-8B/antagonists & inhibitors , Respiratory Function Tests , Respiratory Mechanics/drug effects , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus Infections/physiopathology , Respiratory Syncytial Virus Infections/virology , Virus Replication/drug effects
5.
Am J Physiol Lung Cell Mol Physiol ; 297(4): L785-93, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19684202

ABSTRACT

High tidal volume ventilation is detrimental to alveolar fluid clearance (AFC), but effects of ventilation pressure (P) on AFC are unknown. In anesthetized BALB/c mice ventilated at constant tidal volume (8 ml/kg), mean AFC rate was 12.8% at 6 cmH(2)O P, but increased to 37.3% at 18 cmH(2)O P. AFC rate declined at 22 cmH(2)O P, which also induced lung damage. Increased AFC at 18 cmH(2)O P did not result from elevated plasma catecholamines, hypercapnia, or hypocapnia, but was due to augmented Na(+) and Cl(-) absorption. PKA agonists and beta-agonists stimulated AFC at 10 cmH(2)O P by upregulating amiloride-sensitive Na(+) transport. However, at 18 cmH(2)O P, PKA agonists and beta-agonists reduced AFC. At 15 cmH(2)O P, the AFC rate was intermediate (mean 26.6%), and forskolin and beta-agonists had no effect. Comparable P dependency of AFC and beta-agonist responsiveness was found in C57BL/6 mice. The effect on AFC of increasing P to 18 cmH(2)O was blocked by adenosine deaminase or an A(2b)-adenosine receptor antagonist, and could be mimicked by adenosine in mice ventilated at 10 cmH(2)O P. Modulation of adenosine signaling also resulted in altered responsiveness to beta-agonists. These findings indicate that, in the normal mouse lung, basal AFC rates and responses to beta-agonists are impacted by ventilation pressure in an adenosine-dependent manner.


Subject(s)
Adrenergic beta-2 Receptor Agonists , Adrenergic beta-Agonists/pharmacology , Lung/metabolism , Mucociliary Clearance , Pulmonary Alveoli/metabolism , Respiration, Artificial , Respiratory Mucosa/metabolism , Amiloride/pharmacology , Animals , Bronchoalveolar Lavage Fluid , Colforsin/pharmacology , Epithelial Cells/metabolism , Female , Lung/cytology , Lung/drug effects , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Oxygen/metabolism , Pulmonary Alveoli/cytology , Pulmonary Alveoli/drug effects , Respiratory Mucosa/cytology , Respiratory Mucosa/drug effects
6.
Am J Respir Crit Care Med ; 178(9): 969-76, 2008 Nov 01.
Article in English | MEDLINE | ID: mdl-18689466

ABSTRACT

RATIONALE: Pulmonary infections can impair alveolar fluid clearance (AFC), contributing to formation of lung edema. Effects of influenza A virus (IAV) on AFC are unknown. OBJECTIVES: To determine effects of IAV infection on AFC, and to identify intercellular signaling mechanisms underlying influenza-mediated inhibition of AFC. METHODS: BALB/c mice were infected intranasally with influenza A/WSN/33 (10,000 or 2,500 focus-forming units per mouse). AFC was measured in anesthetized, ventilated mice by instilling 5% bovine serum albumin into the dependent lung. MEASUREMENTS AND MAIN RESULTS: Infection with high-dose IAV resulted in a steady decline in arterial oxygen saturation and increased lung water content. AFC was significantly inhibited starting 1 hour after infection, and remained suppressed through Day 6. AFC inhibition at early time points (1-4 h after infection) did not require viral replication, whereas AFC inhibition later in infection was replication-dependent. Low-dose IAV infection impaired AFC for 10 days, but induced only mild hypoxemia. High-dose IAV infection increased bronchoalveolar lavage fluid ATP and UTP levels. Impaired AFC at Day 2 resulted primarily from reduced amiloride-sensitive AFC, mediated by increased activation of the pyrimidine-P2Y purinergic receptor axis. However, an additional component of AFC impairment was due to activation of A(1) adenosine receptors and stimulation of increased cystic fibrosis transmembrane regulator-mediated anion secretion. Finally, IAV-mediated inhibition of AFC at Day 2 could be reversed by addition of beta-adrenergic agonists to the AFC instillate. CONCLUSIONS: AFC inhibition may be an important feature of early IAV infection. Its blockade may reduce the severity of pulmonary edema and hypoxemia associated with influenza pneumonia.


Subject(s)
Bronchoalveolar Lavage Fluid/virology , Influenza A virus/metabolism , Orthomyxoviridae Infections/metabolism , Animals , Biological Transport, Active , Body Weight , Cytokines/metabolism , Disease Models, Animal , Female , Lung/physiopathology , Male , Mice , Mice, Inbred BALB C , Oxygen/blood , Permeability , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...