Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Alex P. Young is active.

Publication


Featured researches published by Alex P. Young.


Respiratory Physiology & Neurobiology | 2013

Co-activation of μ- and δ-opioid receptors elicits tolerance to morphine-induced ventilatory depression via generation of peroxynitrite.

Alex P. Young; Ryan B. Gruber; Joe F. Discala; Walter J. May; Dylan McLaughlin; Lisa A. Palmer; Stephen J. Lewis

We determined whether pretreatment with (1) the μ-/δ-opioid receptor (μ-/δ-OR) antagonist, naloxone, (2) the δ1,2-OR antagonist, naltrindole, or (3) the peroxynitrite scavenger, d-penicillamine, affects the development of tolerance to the ventilatory depressant effects of morphine in rats. The injection of morphine in vehicle-pretreated rats decreased minute ventilation predominantly via decreases in tidal volume. Pretreatment with naloxone blunted the responses to morphine whereas pretreatment with naltrindole or d-penicillamine did not. A second injection of morphine, given one day later, elicited markedly smaller responses in vehicle rats whereas it elicited pronounced ventilatory depression in rats that were pretreated with naloxone, naltrindole or d-penicillamine (prior to morphine) the day before. Moreover, the ventilatory responses elicited by subsequent exposure to a hypoxic-hypercapnic challenge were markedly depressed in naloxone- or d-penicillamine-pretreated rats compared to vehicle-pretreated rats. These findings suggest that activation of μ- and δ-ORs causes tolerance to the ventilatory depressant effects of morphine at least partly via the generation of peroxynitrite.


Respiratory Physiology & Neurobiology | 2014

Role of central and peripheral opiate receptors in the effects of fentanyl on analgesia, ventilation and arterial blood-gas chemistry in conscious rats

Fraser Henderson; Walter J. May; Ryan B. Gruber; Joseph F. Discala; Veljko Puskovic; Alex P. Young; Santhosh M. Baby; Stephen J. Lewis

This study determined the effects of the peripherally restricted μ-opiate receptor (μ-OR) antagonist, naloxone methiodide (NLXmi) on fentanyl (25μg/kg, i.v.)-induced changes in (1) analgesia, (2) arterial blood gas chemistry (ABG) and alveolar-arterial gradient (A-a gradient), and (3) ventilatory parameters, in conscious rats. The fentanyl-induced increase in analgesia was minimally affected by a 1.5mg/kg of NLXmi but was attenuated by a 5.0mg/kg dose. Fentanyl decreased arterial blood pH, pO2 and sO2 and increased pCO2 and A-a gradient. These responses were markedly diminished in NLXmi (1.5mg/kg)-pretreated rats. Fentanyl caused ventilatory depression (e.g., decreases in tidal volume and peak inspiratory flow). Pretreatment with NLXmi (1.5mg/kg, i.v.) antagonized the fentanyl decrease in tidal volume but minimally affected the other responses. These findings suggest that (1) the analgesia and ventilatory depression caused by fentanyl involve peripheral μ-ORs and (2) NLXmi prevents the fentanyl effects on ABG by blocking the negative actions of the opioid on tidal volume and A-a gradient.


Respiratory Physiology & Neurobiology | 2013

l-Cysteine ethyl ester reverses the deleterious effects of morphine on, arterial blood–gas chemistry in tracheotomized rats

James P. Mendoza; Rachael J. Passafaro; Santhosh M. Baby; Alex P. Young; James N. Bates; Benjamin Gaston; Stephen J. Lewis

This study determined whether the membrane-permeable ventilatory stimulant, L-cysteine ethylester (L-CYSee), reversed the deleterious actions of morphine on arterial blood-gas chemistry in isoflurane-anesthetized rats. Morphine (2 mg/kg, i.v.) elicited sustained decreases in arterial blood pH, pO₂ and sO₂, and increases in pCO₂ (all responses indicative of hypoventilation) and alveolar-arterial gradient (indicative of ventilation-perfusion mismatch). Injections of L-CYSee (100 μmol/kg, i.v.) reversed the effects of morphine in tracheotomized rats but were minimally active in non-tracheotomized rats. L-cysteine or L-serine ethylester (100 μmol/kg, i.v.) were without effect. It is evident that L-CYSee can reverse the negative effects of morphine on arterial blood-gas chemistry and alveolar-arterial gradient but that this positive activity is negated by increases in upper-airway resistance. Since L-cysteine and L-serine ethylester were ineffective, it is evident that cell penetrability and the sulfur moiety of L-CYSee are essential for activity. Due to its ready penetrability into the lungs, chest wall muscle and brain, the effects of L-CYSee on morphine-induced changes in arterial blood-gas chemistry are likely to involve both central and peripheral sites of action.


Journal of Applied Physiology | 2014

Role of nitric oxide-containing factors in the ventilatory and cardiovascular responses elicited by hypoxic challenge in isoflurane-anesthetized rats.

James P. Mendoza; Rachael J. Passafaro; Santhosh M. Baby; Alex P. Young; James N. Bates; Benjamin Gaston; Stephen J. Lewis

Exposure to hypoxia elicits changes in mean arterial blood pressure (MAP), heart rate, and frequency of breathing (fR). The objective of this study was to determine the role of nitric oxide (NO) in the cardiovascular and ventilatory responses elicited by brief exposures to hypoxia in isoflurane-anesthetized rats. The rats were instrumented to record MAP, heart rate, and fR and then exposed to 90 s episodes of hypoxia (10% O2, 90% N2) before and after injection of vehicle, the NO synthase inhibitor N(G)-nitro-L-arginine methyl ester (L-NAME), or the inactive enantiomer D-NAME (both at 50 μmol/kg iv). Each episode of hypoxia elicited a decrease in MAP, bidirectional changes in heart rate (initial increase and then a decrease), and an increase in fR. These responses were similar before and after injection of vehicle or D-NAME. In contrast, the hypoxia-induced decreases in MAP were attenuated after administration of L-NAME. The initial increases in heart rate during hypoxia were amplified whereas the subsequent decreases in heart rate were attenuated in L-NAME-treated rats. Finally, the hypoxia-induced increases in fR were virtually identical before and after administration of L-NAME. These findings suggest that NO factors play a vital role in the expression of the cardiovascular but not the ventilatory responses elicited by brief episodes of hypoxia in isoflurane-anesthetized rats. Based on existing evidence that NO factors play a vital role in carotid body and central responses to hypoxia in conscious rats, our findings raise the novel possibility that isoflurane blunts this NO-dependent signaling.


European Journal of Pharmacology | 2018

Bilateral carotid sinus nerve transection exacerbates morphine-induced respiratory depression

Santhosh M. Baby; Ryan B. Gruber; Alex P. Young; Peter M. MacFarlane; Luc J. Teppema; Stephen J. Lewis

ABSTRACT Opioid‐induced respiratory depression (OIRD) involves decreased sensitivity of ventilatory control systems to decreased blood levels of oxygen (hypoxia) and elevated levels of carbon dioxide (hypercapnia). Understanding the sites and mechanisms by which opioids elicit respiratory depression is pivotal for finding novel therapeutics to prevent and/or reverse OIRD. To examine the contribution of carotid body chemoreceptors OIRD, we used whole‐body plethysmography to evaluate hypoxic (HVR) and hypercapnic (HCVR) ventilatory responses including changes in frequency of breathing, tidal volume, minute ventilation and inspiratory drive, after intravenous injection of morphine (10mg/kg) in sham‐operated (SHAM) and in bilateral carotid sinus nerve transected (CSNX) Sprague‐Dawley rats. In SHAM rats, morphine produced sustained respiratory depression (e.g., decreases in tidal volume, minute ventilation and inspiratory drive) and reduced the HVR and HCVR responses. Unexpectedly, morphine‐induced suppression of HVR and HCVR were substantially greater in CSNX rats than in SHAM rats. This suggests that morphine did not compromise the function of the carotid body‐chemoafferent complex and indeed, that the carotid body acts to defend against morphine‐induced respiratory depression. These data are the first in vivo evidence that carotid body chemoreceptor afferents defend against rather than participate in OIRD in conscious rats. As such, drugs that stimulate ventilation by targeting primary glomus cells and/or chemoafferent terminals in the carotid bodies may help to alleviate OIRD.


Open Journal of Molecular and Integrative Physiology | 2013

Low-dose morphine elicits ventilatory excitant and depressant responses in conscious rats: Role of peripheral µ -opioid receptors

Fraser Henderson; Walter J. May; Ryan B. Gruber; Alex P. Young; Lisa A. Palmer; Benjamin Gaston; Stephen J. Lewis


Open Journal of Molecular and Integrative Physiology | 2013

Morphine has latent deleterious effects on the ventilatory responses to a hypoxic-hypercapnic challenge.

Walter J. May; Fraser Henderson; Ryan B. Gruber; Joseph F. Discala; Alex P. Young; James N. Bates; Lisa A. Palmer; Stephen J. Lewis


Archive | 2015

cardiorespiratory responses in the conscious rat Effect of nitric oxide synthase inhibition on

Yair M. Gozal; Sanford M. Littwin; Luc J. Teppema; Albert Dahan; Grégoire P. Millet; Raphael Faiss; Vincent Pialoux; Benjamin Gaston; Stephen J. Lewis; James P. Mendoza; Rachael J. Passafaro; Santhosh M. Baby; Alex P. Young; James N. Bates


Archive | 2015

elicited by hypoxic challenge in isoflurane-anesthetized rats Role of nitric oxide-containing factors in the ventilatory and cardiovascular responses

Kingman P. Strohl; Bernadette O. Erokwu; Thomas E. Dick; O. Pierrefiche; Luc J. Teppema; Albert Dahan; Benjamin Gaston; Stephen J. Lewis; James P. Mendoza; Rachael J. Passafaro; Santhosh M. Baby; Alex P. Young; James N. Bates


american thoracic society international conference | 2012

Changes In GSNOR Activity Correlate With The Biphasic Ventilatory Response To Hypoxic Challenge In Mice

Lisa A. Palmer; Michael S. Forbes; Alex P. Young; Kathleen Brown-Steinke; Kimberly deRonde; Benjamin Gaston; Stephen J. Lewis

Collaboration


Dive into the Alex P. Young's collaboration.

Top Co-Authors

Avatar

Stephen J. Lewis

Case Western Reserve University

View shared research outputs
Top Co-Authors

Avatar

Benjamin Gaston

Case Western Reserve University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Santhosh M. Baby

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge