Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
J Asthma Allergy ; 16: 1097-1113, 2023.
Article in English | MEDLINE | ID: mdl-37822519

ABSTRACT

Background: The complex nature of asthma has resulted in a poor understanding of its epidemiology, particularly in low-and middle-income countries (LMIC). Clinical subgroups, such as patients with severe asthma, eosinophilic asthma, allergic rhinitis, or nasal polyps, experience additional barriers to care. Methods: Prevalence estimates for asthma and key clinical subgroups were extracted from the Global Burden of Diseases, Injuries, and Risk Factors Study 2019 and from a targeted literature review conducted through PubMed in October of 2021. National estimates were calculated and the roles of potential explanatory factors were explored through qualitative analysis. Results: In total, 162 publications from 69 countries were included. Across continents, asthma prevalence values ranged from 3.44% (Asia), 3.67% (Africa), 4.90% (South America), 5.69% (Europe), 8.29% (North America), to 8.33% (Oceania). Globally, of those with asthma, 26.70% had severe asthma, 30.99% had eosinophilic asthma, 48.95% had allergic rhinitis, and 7.0% to 25.40% had nasal polyps. Countries with higher air quality, income status, and healthcare access and quality reported a higher asthma prevalence. Conclusion: Asthma prevalence values were low in LMICs, potentially indicating health system deficiencies resulting in low diagnosis and reporting. The prevalence of eosinophilic asthma and severe asthma phenotypes was high in many countries, although the prevalence estimates of all asthma subgroups were quite variable.

2.
Expert Rev Gastroenterol Hepatol ; 16(10): 967-980, 2022 Oct.
Article in English | MEDLINE | ID: mdl-36254610

ABSTRACT

INTRODUCTION: Erosive esophagitis (EE) occurs when refluxate from the stomach causes T-lymphocyte infiltration of the esophageal mucosa, resulting in mucosal breaks. Currently, therapy with proton-pump inhibitors (PPIs) is the standard treatment for EE in the United States, but few comprehensive reviews exist on the efficacy of PPIs in US populations. Here, we present the most contemporary, thorough analysis of PPI efficacy rates, and identify and characterize patient subgroups at risk for poor healing outcomes. AREAS COVERED: We searched the literature to identify studies reporting rates of endoscopic healing and maintained healing of EE with PPI therapies in the US and found a paucity of recent evidence and real-world evidence. Twenty-two studies from 2009 and earlier were included in the final dataset. EXPERT OPINION: Rates of EE healing with PPIs were highest after 8 weeks of treatment, with over 80% of patients in most treatment arms demonstrating endoscopic healing, compared to lower efficacy (<80%) at 4 weeks. Rates of maintained healing with PPIs at 6 and 12 months were mostly lower than 80%, although the data were limited. Symptomatic patients and those with severe EE were less likely to achieve healing. Obese patients experienced similar healing rates as non-obese patients.


Subject(s)
Barrett Esophagus , Esophagitis , Peptic Ulcer , Upper Gastrointestinal Tract , Humans , United States/epidemiology , Esophagitis/diagnosis , Esophagitis/drug therapy , Esophagitis/complications , Proton Pump Inhibitors/adverse effects , Barrett Esophagus/etiology
3.
eNeuro ; 2022 Jul 25.
Article in English | MEDLINE | ID: mdl-35882549

ABSTRACT

There are currently no Food and Drug Administration (FDA)-approved delta opioid receptor (DOR)-selective agonists, despite having fewer side effects in rodents and non-human primates compared to traditional mu opioid receptor (MOR) therapeutics (Vanderah, 2010). Targeting peripheral receptors is an attractive strategy to reduce abuse potential. However, peripheral opioid receptors do not readily respond to agonists unless primed by inflammation, which would limit their efficacy in non-inflammatory pain patients (Stein et al., 1989). It was recently identified that G protein-coupled receptor kinase 2 (GRK2) maintains DOR incompetence in non-inflamed nociceptors (Brackley et al., 2016; Brackley et al., 2017). Here, we report that paroxetine, a selective serotonin reuptake inhibitor and potent GRK2 inhibitor (Thal et al., 2012), reduces chronic GRK2 association with membrane DOR, thereby enhancing peripheral DOR-mediated analgesic competence in the absence of inflammation. Interestingly, paroxetine's effects on GRK2 in vivo are limited to peripheral tissues in the male rat. The effects of paroxetine on DOR competence are notably antagonized by GRK2 overexpression. This is the first study to suggest that paroxetine induces peripheral DOR analgesic competence through a GRK2-dependent mechanism, improving analgesic efficacy in non-inflamed tissue. Because paroxetine targets the protein that governs peripheral opioid receptor responsiveness, and does so in the absence of inflammation, we propose that paroxetine may be suitable as a co-therapy with peripherally-restrictive doses of opioids to improve analgesic efficacy in non-inflammatory pain conditions.Significance StatementOpioids that target MOR represent the gold-standard for analgesic healthcare, despite widespread abuse potential and the ongoing opioid-epidemic. Work herein uncovers the therapeutic potential of targeting peripheral DOR for analgesic utility with an FDA-approved GRK2 inhibitor paroxetine to boost efficacy and reduce side effect profiles. Analgesic pain management targeting DOR with increased efficacy through adjuvant paroxetine treatment could reduce over-reliance on MOR agonist opioids for pain relief and usher in new options for analgesia.

4.
Environ Entomol ; 51(5): 1030-1039, 2022 10 21.
Article in English | MEDLINE | ID: mdl-35866523

ABSTRACT

All holometabolous insects undergo a pupal life stage, a transformative period during which the insects are immobile and thus particularly vulnerable to both natural enemies and harmful abiotic conditions. For multivoltine species like the silver-spotted skipper [Epargyreus clarus (Cramer) (Lepidoptera: Hesperiidae)], which produces both diapausing and nondiapausing generations throughout much of its range, both the duration of the pupal stage and the ecological challenges faced by pupae can differ among generations. We conducted a set of field experiments to investigate the seasonal and annual variation in pupal mortality for E. clarus pupae experiencing different biotic and abiotic conditions. We also examined the behavioral and ecological factors influencing the construction and persistence of pupal shelters by prepupal larvae. Surprisingly, measures of both cumulative and daily pupal predation were significantly higher during the relatively short (10-14 d) nondiapausing (summer) generations, compared with the diapausing (winter) generations, despite a nearly 20-fold longer pupal duration recorded for the latter. Indirect evidence from field censuses suggested that this intergenerational difference in mortality was due to seasonal variation in consumption of pupae by generalist vertebrate predators. The presence of a shelter increased survival in summer, though not in winter, perhaps because winter pupae were likely to be buried under autumnal leaf litter, regardless of initial shelter status. When constructing their shelters, prepupal E. clarus larvae did not prefer host leaves over nonhost leaves, suggesting that induced preferences are unlikely to play an important role in this process. Despite finding marked differences in the decomposition rates of shelter leaves derived from host vs. nonhost plants, several lines of evidence suggest that these differences are unlikely to impact E. clarus pupal mortality during either the summer or winter generations.


Subject(s)
Butterflies , Animals , Pupa , Larva , Seasons , North America
5.
J Neurochem ; 162(2): 156-165, 2022 07.
Article in English | MEDLINE | ID: mdl-35526109

ABSTRACT

Inflammatory hyperalgesia represents a nociceptive phenotype that can become persistent in nature through dynamic protein modifications. However, a large gap in knowledge exists concerning how the integration of intracellular signaling molecules coordinates a persistent inflammatory phenotype. Herein, we demonstrate that Raf Kinase Anchoring Protein (RKIP) interrupts a vital canonical desensitization pathway to maintain bradykinin (BK) receptor activation in primary afferent neurons. Biochemical analyses of primary neuronal cultures indicate bradykinin-stimulated PKC phosphorylation of RKIP at Ser153. Furthermore, BK exposure increases G-protein Receptor Kinase 2 (GRK2) binding to RKIP, inhibiting pharmacological desensitization of the BK receptor. Additional studies found that molecular RKIP down-regulation increases BK receptor desensitization in real-time imaging of primary afferent neurons, identifying a key pathway integrator in the desensitization process that controls multiple GRK2-sensitive G-protein coupled receptors. Therefore, RKIP serves as an integral scaffolding protein that inhibits BK receptor desensitization.


Subject(s)
Bradykinin , Receptors, Bradykinin , Bradykinin/pharmacology , Phosphorylation , Signal Transduction , Transcription Factors , raf Kinases
6.
J Pharmacol Exp Ther ; 378(2): 96-107, 2021 08.
Article in English | MEDLINE | ID: mdl-33990416

ABSTRACT

Opioid overdose intervention by naloxone, a high affinity receptor antagonist, reverses opioid-induced respiratory depression (OIRD) and analgesia by displacing opioids. Systemic naloxone stimulates release of the hypothalamic neuropeptide oxytocin, which has analgesic properties and participates in cardiorespiratory homeostasis. To test the hypothesis that oxytocin can reverse OIRD, we assessed the rescue potential of graded doses (0, 0.1, 2, 5, 10, 50 nmol/kg, i.v.) of oxytocin to counter fentanyl (60 nmol/kg, i.v.)-induced depression of neural inspiration indexed by recording phrenic nerve activity (PNA) in anesthetized (urethane/α-chloralose), vagotomized, and artificially ventilated rats. Oxytocin dose-dependently rescued fentanyl OIRD by almost immediately reversing PNA burst arrest (P = 0.0057) and restoring baseline burst frequency (P = 0.0016) and amplitude (P = 0.0025) at low but not high doses, resulting in inverted bell-shaped dose-response curves. Oxytocin receptor antagonism (40 nmol/kg, i.v.) prevented oxytocin reversal of OIRD (arrest: P = 0.0066, frequency: P = 0.0207, amplitude: P = 0.0022). Vasopressin 1A receptor (V1aR) antagonism restored high-dose oxytocin efficacy to rescue OIRD (P = 0.0170 to P < 0.0001), resulting in classic sigmoidal dose-response curves, and prevented (P = 0.0135) transient hypertension from V1aR cross-activation (P = 0.0275). Alone, vasopressin (5 nmol/kg, i.v.) failed to reverse fentanyl respiratory arrest (P = 0.6184). The nonpeptide oxytocin receptor agonist WAY-267464 (75 nmol/kg, i.v.), which has V1aR antagonist properties, quickly reversed fentanyl OIRD (P < 0.0001), with rapid recovery of PNA frequency (P = 0.0011) and amplitude (P = 0.0044) without adverse hemodynamic consequences (P = 0.9991). Findings indicate that peptide and nonpeptide agonist activation of oxytocin receptors without V1aR cross-activation rescues fentanyl OIRD. Oxytocin receptor agonists could be lifesaving resuscitation agents that enhance rather than interrupt opioid analgesia. SIGNIFICANCE STATEMENT: Oxytocin receptor activation produces analgesia. Here, we demonstrate that activation by the US Food and Drug Administration-approved agonist oxytocin and the nonpeptide partial agonist WAY-267464 can each reverse fentanyl cardiorespiratory depression. Selective targeting of oxytocin receptors for resuscitation from opioid overdose, alone or in combination with an opioid antagonist, could eliminate or attenuate negative side effects associated with traditional opioid receptor antagonism.


Subject(s)
Oxytocin , Receptors, Opioid , Animals , Fentanyl , Rats , Receptors, Oxytocin
7.
J Physiol ; 598(15): 3239-3257, 2020 08.
Article in English | MEDLINE | ID: mdl-32415789

ABSTRACT

KEY POINTS: Sleep apnoea increases susceptibility to opioid-induced respiratory depression (OIRD). Endogenous opioids are implicated as a contributing factor in sleep apnoea. Rats exposed to sleep-phase chronic intermittent hypercapnic hypoxia (CIHH) for 7 days exhibited exaggerated OIRD to systemic fentanyl both while anaesthetized and artificially ventilated and while conscious and breathing spontaneously, implicating heightened CNS inhibitory efficacy of fentanyl. CIHH also induced tonic endogenous opioid suppression of neural inspiration. Sleep-related episodes of hypercapnic hypoxia, as in sleep apnoea, promote hypersensitivity to OIRD, with tonic respiratory depression by endogenous opioids implicated as a potential underlying cause. ABSTRACT: Sleep apnoea (SA) increases opioid-induced respiratory depression (OIRD) and lethality. To test the hypothesis that this results from chronic intermittent bouts of hypercapnic hypoxia (CIHH) accompanying SA, we compared OIRD across continuously normoxic control rats and rats exposed to sleep-phase (8 h/day) CIHH for 1 week. OIRD sensitivity was first assessed in anaesthetized (urethane/α-chloralose), vagotomized and artificially ventilated rats by recording phrenic nerve activity (PNA) to index neural inspiration and quantify PNA burst inhibition to graded doses (0, 2, 20, 50 µg kg-1 , i.v.) of the synthetic opioid fentanyl. Fentanyl dose-dependently reduced PNA burst frequency (P = 0.0098-0.0001), while increasing the duration of burst quiescence at 50 µg kg-1 (P < 0.0001, n = 5-6/group/dose). CIHH shifted the fentanyl dose-phrenic burst frequency response curve to the left (P = 0.0163) and increased the duration of burst quiescence (P < 0.0001). During fentanyl recovery, PNA burst width was increased relative to baseline in normoxic and CIHH rats. Systemic naloxone (1 mg kg-1 , i.v.) reversed fentanyl-induced PNA arrest in both groups (P = 0.0002), and increased phrenic burst amplitude above baseline (P = 0.0113) in CIHH rats only. Differential sensitivity to anaesthesia as a cause of CIHH-related OIRD hypersensitivity was excluded by observing in conscious spontaneously breathing rats that fentanyl at 20 µg kg-1 (i.v.), which silenced PNA in anaesthetized rats, differentially increased breathing variability in normoxic versus CIHH rats (P = 0.0427), while significantly reducing breathing frequency (P < 0.0001) and periodicity (P = 0.0003) in CIHH rats only. Findings indicate that CIHH increased OIRD sensitivity, with tonic inspiratory depression by endogenous opioids as a likely contributing cause.


Subject(s)
Respiratory Hypersensitivity , Respiratory Insufficiency , Analgesics, Opioid , Animals , Fentanyl/toxicity , Hypoxia , Rats , Rats, Sprague-Dawley
8.
Pain Rep ; 3(3): e658, 2018 May.
Article in English | MEDLINE | ID: mdl-29922747

ABSTRACT

INTRODUCTION: Chronic metabotropic glutamate receptor activation in nociceptive afferents may upregulate A-Kinase Anchoring Protein 150 (AKAP150) expression and/or function. OBJECTIVES: To quantify transcriptional changes in AKAP150 expression and/or function after long-term mGluR5 agonist exposure, and identify transcriptional elements responsible. METHODS: Dorsal root ganglia (DRG) were dissected from Sprague-Dawley rats and cultured for biochemical analysis of AKAP150 expression after prolonged mGluR5 agonist exposure. Serum response factor (SRF) expression was knocked down through siRNA in cultures to demonstrate significance to AKAP150 upregulation. Serum response factor was also knocked down in vivo through intrathecal injections of specifically targeted oligonucleotides to demonstrate significance to hyperalgesic priming behavior in persistent mechanical hypersensitivity. RESULTS: Serum response factor and AKAP150 are coexpressed in TRPV1(+) DRG neurons in intact DRG. Prolonged mGluR5 agonist exposure increases SRF-dependent transcription and AKAP150 expression in a manner sensitive to protein kinase C inhibition and SRF knock down. Serum response factor in vivo knock down reduces mechanical hyperalgesic priming. CONCLUSION: Serum response factor transcription plays an important role in transcriptional upregulation of AKAP and hyperalgesic priming behavior, and may contribute to the increased role of AKAP150 in the transition from acute to chronic pain.

9.
PLoS One ; 12(8): e0182102, 2017.
Article in English | MEDLINE | ID: mdl-28797041

ABSTRACT

Blast-associated sensory and cognitive trauma sustained by military service members is an area of extensively studied research. Recent studies in our laboratory have revealed that low-level blast exposure increased expression of transient receptor potential vanilloid 1 (TRPV1) and endothelin-1 (ET-1), proteins well characterized for their role in mediating pain transmission, in the cornea. Determining the functional consequences of these alterations in protein expression is critical to understanding blast-related sensory trauma. Thus, the purpose of this study was to examine TRPV1 and ET-1 expression in ocular associated sensory tissues following primary and tertiary blast. A rodent model of blast injury was used in which anesthetized animals, unrestrained or restrained, received a single or repeat blast (73.8 ± 5.5 kPa) from a compressed air shock tube once or daily for five consecutive days, respectively. Behavioral and functional analyses were conducted to assess blast effects on nocifensive behavior and TRPV1 activity. Immunohistochemistry and Western Blot were also performed with trigeminal ganglia (TG) to determine TRPV1, ET-1 and glial fibrillary associated protein (GFAP) expression following blast. Increased TRPV1, ET-1 and GFAP were detected in the TG of animals exposed to repeat blast. Increased nocifensive responses were also observed in animals exposed to repeat, tertiary blast as compared to single blast and control. Moreover, decreased TRPV1 desensitization was observed in TG neurons exposed to repeat blast. Repeat, tertiary blast resulted in increased TRPV1, ET-1 and GFAP expression in the TG, enhanced nociception and decreased TRPV1 desensitization.


Subject(s)
Blast Injuries/metabolism , Endothelin-1/metabolism , Neurons/metabolism , TRPV Cation Channels/metabolism , Trigeminal Ganglion/metabolism , Animals , Male , Rats , Rats, Long-Evans
10.
Sci Rep ; 7(1): 1842, 2017 05 12.
Article in English | MEDLINE | ID: mdl-28500286

ABSTRACT

Mechanical pain serves as a base clinical symptom for many of the world's most debilitating syndromes. Ion channels expressed by peripheral sensory neurons largely contribute to mechanical hypersensitivity. Transient Receptor Potential A 1 (TRPA1) is a ligand-gated ion channel that contributes to inflammatory mechanical hypersensitivity, yet little is known as to the post-translational mechanism behind its somatosensitization. Here, we utilize biochemical, electrophysiological, and behavioral measures to demonstrate that metabotropic glutamate receptor-induced sensitization of TRPA1 nociceptors stimulates targeted modification of the receptor. Type 1 mGluR5 activation increases TRPA1 receptor agonist sensitivity in an AKA-dependent manner. As a scaffolding protein for Protein Kinases A and C (PKA and PKC, respectively), AKAP facilitates phosphorylation and sensitization of TRPA1 in ex vivo sensory neuronal preparations. Furthermore, hyperalgesic priming of mechanical hypersensitivity requires both TRPA1 and AKAP. Collectively, these results identify a novel AKAP-mediated biochemical mechanism that increases TRPA1 sensitivity in peripheral sensory neurons, and likely contributes to persistent mechanical hypersensitivity.


Subject(s)
A Kinase Anchor Proteins/metabolism , Receptors, Metabotropic Glutamate/metabolism , TRPA1 Cation Channel/metabolism , A Kinase Anchor Proteins/chemistry , A Kinase Anchor Proteins/genetics , Animals , CHO Cells , Calcium/metabolism , Chromatography, Liquid , Cricetulus , Male , Mice , Mice, Knockout , Molecular Imaging , Phosphorylation , Rats , Receptors, Metabotropic Glutamate/chemistry , TRPA1 Cation Channel/chemistry , TRPA1 Cation Channel/genetics , Tandem Mass Spectrometry
11.
J Biol Chem ; 292(21): 8762-8772, 2017 05 26.
Article in English | MEDLINE | ID: mdl-28381559

ABSTRACT

µ-Opioid receptor (MOR) agonists are often used to treat severe pain but can result in adverse side effects. To circumvent systemic side effects, targeting peripheral opioid receptors is an attractive alternative treatment for severe pain. Activation of the δ-opioid receptor (DOR) produces similar analgesia with reduced side effects. However, until primed by inflammation, peripheral DOR is analgesically incompetent, raising interest in the mechanism. We recently identified a novel role for G-protein-coupled receptor kinase 2 (GRK2) that renders DOR analgesically incompetent at the plasma membrane. However, the mechanism that maintains constitutive GRK2 association with DOR is unknown. Protein kinase A (PKA) phosphorylation of GRK2 at Ser-685 targets it to the plasma membrane. Protein kinase A-anchoring protein 79/150 (AKAP), residing at the plasma membrane in neurons, scaffolds PKA to target proteins to mediate downstream signal. Therefore, we sought to determine whether GRK2-mediated DOR desensitization is directed by PKA via AKAP scaffolding. Membrane fractions from cultured rat sensory neurons following AKAP siRNA transfection and from AKAP-knock-out mice had less PKA activity, GRK2 Ser-685 phosphorylation, and GRK2 plasma membrane targeting than controls. Site-directed mutagenesis revealed that GRK2 Ser-685 phosphorylation drives the association of GRK2 with plasma membrane-associated DOR. Moreover, overexpression studies with AKAP mutants indicated that impaired AKAP-mediated PKA scaffolding significantly reduces DOR-GRK2 association at the plasma membrane and consequently increases DOR activity in sensory neurons without a priming event. These findings suggest that AKAP scaffolds PKA to increase plasma membrane targeting and phosphorylation of GRK2 to maintain DOR analgesic incompetence in peripheral sensory neurons.


Subject(s)
Cell Membrane/metabolism , Receptors, Opioid, delta/metabolism , Sensory Receptor Cells/metabolism , Signal Transduction , A Kinase Anchor Proteins/genetics , A Kinase Anchor Proteins/metabolism , Animals , Cattle , Cell Membrane/genetics , Cyclic AMP-Dependent Protein Kinases/genetics , Cyclic AMP-Dependent Protein Kinases/metabolism , G-Protein-Coupled Receptor Kinase 2/genetics , G-Protein-Coupled Receptor Kinase 2/metabolism , Male , Mice , Phosphorylation/genetics , Rats , Rats, Sprague-Dawley , Receptors, Opioid, delta/agonists , Receptors, Opioid, delta/genetics , Sensory Receptor Cells/pathology
12.
Cell Rep ; 16(10): 2686-2698, 2016 09 06.
Article in English | MEDLINE | ID: mdl-27568556

ABSTRACT

Opioids remain the standard for analgesic care; however, adverse effects of systemic treatments contraindicate long-term administration. While most clinical opioids target mu opioid receptors (MOR), those that target the delta class (DOR) also demonstrate analgesic efficacy. Furthermore, peripherally restrictive opioids represent an attractive direction for analgesia. However, opioid receptors including DOR are analgesically incompetent in the absence of inflammation. Here, we report that G protein-coupled receptor kinase 2 (GRK2) naively associates with plasma membrane DOR in peripheral sensory neurons to inhibit analgesic agonist efficacy. This interaction prevents optimal Gß subunit association with the receptor, thereby reducing DOR activity. Importantly, bradykinin stimulates GRK2 movement away from DOR and onto Raf kinase inhibitory protein (RKIP). protein kinase C (PKC)-dependent RKIP phosphorylation induces GRK2 sequestration, restoring DOR functionality in sensory neurons. Together, these results expand the known function of GRK2, identifying a non-internalizing role to maintain peripheral DOR in an analgesically incompetent state.


Subject(s)
G-Protein-Coupled Receptor Kinase 2/metabolism , Receptors, Opioid, delta/metabolism , Analgesics, Opioid/pharmacology , Animals , Bradykinin/pharmacology , GTP-Binding Proteins/metabolism , Male , Nociception/drug effects , Phosphatidylethanolamine Binding Protein/metabolism , Protein Kinase C/metabolism , Rats, Sprague-Dawley , Sensory Receptor Cells/drug effects , Sensory Receptor Cells/metabolism , Signal Transduction/drug effects , Type C Phospholipases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...