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

Country/Region as subject
Publication year range
1.
Proc Natl Acad Sci U S A ; 114(43): E9163-E9171, 2017 10 24.
Article in English | MEDLINE | ID: mdl-29073113

ABSTRACT

The mostly widely used bronchodilators in asthma therapy are ß2-adrenoreceptor (ß2AR) agonists, but their chronic use causes paradoxical adverse effects. We have previously determined that ß2AR activation is required for expression of the asthma phenotype in mice, but the cell types involved are unknown. We now demonstrate that ß2AR signaling in the airway epithelium is sufficient to mediate key features of the asthmatic responses to IL-13 in murine models. Our data show that inhibition of ß2AR signaling with an aerosolized antagonist attenuates airway hyperresponsiveness (AHR), eosinophilic inflammation, and mucus-production responses to IL-13, whereas treatment with an aerosolized agonist worsens these phenotypes, suggesting that ß2AR signaling on resident lung cells modulates the asthma phenotype. Labeling with a fluorescent ß2AR ligand shows the receptors are highly expressed in airway epithelium. In ß2AR-/- mice, transgenic expression of ß2ARs only in airway epithelium is sufficient to rescue IL-13-induced AHR, inflammation, and mucus production, and transgenic overexpression in WT mice exacerbates these phenotypes. Knockout of ß-arrestin-2 (ßarr-2-/-) attenuates the asthma phenotype as in ß2AR-/- mice. In contrast to eosinophilic inflammation, neutrophilic inflammation was not promoted by ß2AR signaling. Together, these results suggest ß2ARs on airway epithelial cells promote the asthma phenotype and that the proinflammatory pathway downstream of the ß2AR involves ßarr-2. These results identify ß2AR signaling in the airway epithelium as capable of controlling integrated responses to IL-13 and affecting the function of other cell types such as airway smooth muscle cells.


Subject(s)
Asthma/etiology , Eosinophils/pathology , Epithelial Cells/metabolism , Lung/pathology , Receptors, Adrenergic, beta-2/metabolism , Adrenergic beta-2 Receptor Antagonists/pharmacology , Animals , Asthma/pathology , Bronchi/cytology , Disease Models, Animal , Epinephrine/pharmacology , Epithelial Cells/drug effects , Epithelial Cells/pathology , Humans , Inflammation/metabolism , Inflammation/pathology , Interleukin-13/toxicity , Lung/cytology , Metaplasia , Mice, Inbred C57BL , Mice, Transgenic , Pneumonia/chemically induced , Pneumonia/metabolism , Receptors, Adrenergic, beta-2/genetics , Signal Transduction
2.
Brain Behav Immun ; 68: 66-75, 2018 02.
Article in English | MEDLINE | ID: mdl-29017969

ABSTRACT

Acute dynamic exercise mobilizes CD34+ hematopoietic stem cells (HSCs) to the bloodstream, potentially serving as an economical adjuvant to boost the collection of HSCs from stem cell transplant donors. The mechanisms responsible for HSC mobilization with exercise are unknown but are likely due to hemodynamic perturbations, endogenous granulocyte-colony stimulating factor (G-CSF), and/or ß2-adrenergic receptor (ß2-AR) signaling. We characterized the temporal response of HSC mobilization and plasma G-CSF following exercise, and determined the impact of in vivo ß-AR blockade on the exercise-induced mobilization of HSCs. Healthy runners (n = 15) completed, in balanced order, two single bouts of steady state treadmill running exercise at moderate (lasting 90-min) or vigorous (lasting 30-min) intensity. A separate cohort of healthy cyclists (n = 12) completed three 30-min cycling ergometer trials at vigorous intensity after ingesting: (i) 10 mg bisoprolol (ß1-AR antagonist); (ii) 80 mg nadolol (ß1 + ß2-AR antagonist); or (iii) placebo, in balanced order with a double-blind design. Blood samples collected before, during (runners only), immediately after, and at several points during exercise recovery were used to determine circulating G-CSF levels (runners only) and enumerate CD34+ HSCs by flow cytometry (runners and cyclists). Steady state vigorous but not moderate intensity exercise mobilized HSCs, increasing the total blood CD34+ count by ∼4.15 ±â€¯1.62 Δcells/µl (+202 ±â€¯92%) compared to resting conditions. Plasma G-CSF increased in response to moderate but not vigorous exercise. Relative to placebo, nadolol and bisoprolol lowered exercising heart rate and blood pressure to comparable levels. The number of CD34+ HSCs increased with exercise after the placebo and bisoprolol trials, but not the nadolol trial, suggesting ß2-AR signaling mediated the mobilization of CD34+ cells [Placebo: 2.10 ±â€¯1.16 (207 ±â€¯69.2%), Bisoprolol 1.66 ±â€¯0.79 (+163 ±â€¯29%), Nadolol: 0.68 ±â€¯0.54 (+143 ±â€¯36%) Δcells/µL]. We conclude that the mobilization of CD34+ HSCs with exercise is not dependent on circulating G-CSF and is likely due to the combined actions of ß2-AR signaling and hemodynamic shear stress.


Subject(s)
Exercise/physiology , Hematopoietic Stem Cells/physiology , Receptors, Adrenergic, beta-2/metabolism , Adrenergic beta-2 Receptor Antagonists/metabolism , Adult , Antigens, CD34/metabolism , Bisoprolol , Double-Blind Method , Female , Granulocyte Colony-Stimulating Factor/blood , Granulocyte Colony-Stimulating Factor/metabolism , Granulocyte Colony-Stimulating Factor/pharmacology , Hematopoietic Stem Cell Mobilization/methods , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/metabolism , Humans , Male , Nadolol , Peripheral Blood Stem Cells , Receptors, Adrenergic, beta-2/physiology , Signal Transduction
3.
Brain Behav Immun ; 74: 143-153, 2018 11.
Article in English | MEDLINE | ID: mdl-30172948

ABSTRACT

Acute exercise preferentially mobilizes cytotoxic T-cells, NK-cells and non-classical monocytes to the bloodstream under the influence of hemodynamic forces and/or ß2-adrenergic receptor (ß2-AR) signaling. However, the relative contribution of these mechanisms to the redeployment of the most exercise-responsive cell types is largely unknown. We determined the lymphocyte and monocyte subtypes mobilized to blood during exercise via ß2-AR signaling whilst controlling for ß1-AR mediated reductions in hemodynamic forces. In a randomized, double blind, complete cross-over design, 14 healthy cyclists exercised for 30-minutes at +10% of blood lactate threshold after ingesting: (1) a placebo, (2) a ß1-preferential antagonist (10 mg bisoprolol), or (2) a non-preferential ß1 + ß2-antagonist (80 mg nadolol) across three trials separated by >7-days. Bisoprolol was administered to reduce hemodynamic forces (heart rate and blood pressure) during exercise to levels comparable with nadolol but without blocking ß2-ARs. The mobilization of total NK-cells, terminally differentiated (CD57+) NK-cells, central memory, effector memory and CD45RA+ effector memory CD8+ T-cells; non-classical monocytes; and γδ T-cells were significantly blunted or abrogated under nadolol compared to both bisoprolol and placebo, indicating that the exercise-induced mobilization of these cell types to the blood is largely influenced by ß2-AR signaling. Nadolol failed to inhibit the mobilization of classical monocytes, CD4+ T-cells (and their subsets) or naïve CD8+ T-cells, indicating that these cell types are mobilized with exercise independently of the ß2-AR. We conclude that the preferential mobilization of NK-cells, non-classical monocytes and differentiated subsets of CD8+ T-cells with exercise is largely dependent on catecholamine signaling through the ß2-AR. These findings provide mechanistic insights by which distinct lymphocyte and monocyte subtypes are preferentially mobilized to protect the host from anticipated injury or infection in response to an acute stress response.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Exercise/physiology , Killer Cells, Natural/immunology , Monocytes/immunology , Receptors, Adrenergic, beta-2/immunology , Adult , Bisoprolol/pharmacology , CD8-Positive T-Lymphocytes/metabolism , Catecholamines/immunology , Catecholamines/metabolism , Cross-Over Studies , Double-Blind Method , Female , Healthy Volunteers , Humans , Killer Cells, Natural/metabolism , Lymphocyte Activation , Male , Monocytes/metabolism , Nadolol/pharmacology , Receptors, Adrenergic, beta-2/metabolism , Signal Transduction
4.
Pulm Pharmacol Ther ; 46: 30-40, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28729042

ABSTRACT

BACKGROUND: Our previous studies suggested certain ß-adrenoceptor blockers (ß-blockers) attenuate the asthma phenotype in ovalbumin driven murine models of asthma. However, the ovalbumin model has been criticized for lack of clinical relevance. METHODS: We tested the non-selective ß-blockers, carvedilol and nadolol, in house dust mite (HDM) driven murine asthma models where drugs were administered both pre- and post-development of the asthma phenotype. We measured inflammation, mucous metaplasia, and airway hyper-responsiveness (AHR). We also measured the effects of the ß-blockers on extracellular-signal regulated kinase (ERK 1/2) phosphorylation in lung homogenates. RESULTS: We show that nadolol, but not carvedilol, attenuated inflammation and mucous metaplasia, and had a moderate effect attenuating AHR. Following HDM exposure, ERK1/2 phosphorylation was elevated, but the level of phosphorylation was unaffected by ß-blockers, suggesting ERK1/2 phosphorylation becomes dissociated from the asthma phenotype. CONCLUSION: Our findings in HDM models administering drugs both pre- and post-development of the asthma phenotype are consistent with previous results using ovalbumin models and show differential effects for nadolol and carvedilol on the asthma phenotype. Lastly, our data suggest that ERK1/2 phosphorylation may be involved in development of the asthma phenotype, but may have a limited role in maintaining the phenotype.


Subject(s)
Adrenergic beta-Antagonists/pharmacology , Asthma/drug therapy , Carbazoles/pharmacology , Nadolol/pharmacology , Propanolamines/pharmacology , Animals , Asthma/immunology , Asthma/pathology , Carvedilol , Disease Models, Animal , Inflammation/drug therapy , Inflammation/pathology , Male , Mice , Mice, Inbred BALB C , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Ovalbumin/immunology , Phenotype , Phosphorylation , Pyroglyphidae/immunology , Respiratory Hypersensitivity/drug therapy
5.
Am J Respir Cell Mol Biol ; 55(2): 234-42, 2016 08.
Article in English | MEDLINE | ID: mdl-26909542

ABSTRACT

Mice lacking the endogenous ß2-adrenoceptor (ß2AR) agonist epinephrine (phenylethanolamine N-methyltransferase [PNMT]-knockout mice) are resistant to developing an "asthma-like" phenotype in an ovalbumin sensitization and challenge (Ova S/C) model, and chronic administration of ß2AR agonists to PNMT-KO mice restores the phenotype. Based on these and other studies showing differential effects of various ß2AR ligands on the asthma phenotype, we have speculated that the permissive effect of endogenous epinephrine and exogenous ß2AR agonists on allergic lung inflammation can be explained by qualitative ß2AR signaling. The ß2AR can signal through at least two pathways: the canonical Gαs-cAMP pathway and a ß-arrestin-dependent pathway. Previous studies suggest that ß-arrestin-2 is required for allergic lung inflammation. On the other hand, cell-based assays suggest antiinflammatory effects of Gαs-cAMP signaling. This study was designed to test whether the in vitro antiinflammatory effects of phosphodiesterase 4 inhibitors, known to increase intracellular cAMP in multiple airway cell types, attenuate the asthma-like phenotype produced by the ß2AR agonists formoterol and salmeterol in vivo in PNMT-KO mice, based on the hypothesis that skewing ß2AR signaling toward Gαs-cAMP pathway is beneficial. Airway inflammatory cells, epithelial mucus production, and airway hyperresponsiveness were quantified. In Ova S/C PNMT-KO mice, formoterol and salmeterol restored the asthma-like phenotype comparable to Ova S/C wild-type mice. However, coadministration of either roflumilast or rolipram attenuated this formoterol- or salmeterol-driven phenotype in Ova S/C PNMT-KO. These findings suggest that amplification of ß2AR-mediated cAMP by phosphodiesterase 4 inhibitors attenuates the asthma-like phenotype promoted by ß-agonists.


Subject(s)
Adrenergic beta-2 Receptor Agonists/adverse effects , Asthma/drug therapy , Phenylethanolamine N-Methyltransferase/deficiency , Phosphodiesterase 4 Inhibitors/therapeutic use , Aminopyridines/administration & dosage , Aminopyridines/pharmacology , Animals , Asthma/complications , Asthma/pathology , Asthma/physiopathology , Benzamides/administration & dosage , Benzamides/pharmacology , Bronchial Hyperreactivity/complications , Bronchial Hyperreactivity/drug therapy , Bronchial Hyperreactivity/pathology , Bronchial Hyperreactivity/physiopathology , Cyclopropanes/administration & dosage , Cyclopropanes/pharmacology , Drug Therapy, Combination , Eosinophils/drug effects , Eosinophils/pathology , Formoterol Fumarate/administration & dosage , Formoterol Fumarate/pharmacology , Inflammation/pathology , Lung/drug effects , Lung/pathology , Lung/physiopathology , Mice , Mice, Knockout , Mucus/metabolism , Phenotype , Phenylethanolamine N-Methyltransferase/metabolism , Phosphodiesterase 4 Inhibitors/administration & dosage , Phosphodiesterase 4 Inhibitors/pharmacology
7.
Handb Exp Pharmacol ; 219: 387-403, 2014.
Article in English | MEDLINE | ID: mdl-24292841

ABSTRACT

The obstructive lung disease asthma is treated by drugs that target, either directly or indirectly, G protein-coupled receptors (GPCRs). GPCRs coupled to Gq are the primary mediators of airway smooth muscle (ASM) contraction and increased airway resistance, whereas the Gs-coupled beta-2-adrenoceptor (ß2AR) promotes pro-relaxant signaling in and relaxation of ASM resulting in greater airway patency and reversal of life-threatening bronchoconstriction. In addition, GPCR-mediated functions in other cell types, including airway epithelium and hematopoietic cells, are involved in the control of lung inflammation that causes most asthma. The capacity of arrestins to regulate GPCR signaling, via either control of GPCR desensitization/resensitization or G protein-independent signaling, renders arrestins an intriguing therapeutic target for asthma and other obstructive lung diseases. This review will focus on the potential role of arrestins in those GPCR-mediated airway cell functions that are dysregulated in asthma.


Subject(s)
Arrestins/metabolism , Asthma/physiopathology , Receptors, G-Protein-Coupled/metabolism , Airway Resistance/physiology , Animals , Anti-Asthmatic Agents/pharmacology , Asthma/drug therapy , Humans , Inflammation/drug therapy , Inflammation/physiopathology , Molecular Targeted Therapy , Muscle Contraction/physiology , Muscle, Smooth/metabolism , Receptors, G-Protein-Coupled/drug effects , Signal Transduction/physiology
8.
Am J Respir Cell Mol Biol ; 48(2): 220-9, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23204390

ABSTRACT

ß(2)-Adrenoceptor (ß2AR) agonists are the most effective class of bronchodilators and a mainstay of asthma management. The first potent ß2AR agonist discovered and widely used in reversing the airway constriction associated with asthma exacerbation was the endogenous activator of the ß2AR, epinephrine. In this study, we demonstrate that activation of the ß2AR by epinephrine is paradoxically required for development of the asthma phenotype. In an antigen-driven model, mice sensitized and challenged with ovalbumin showed marked elevations in three cardinal features of the asthma phenotype: inflammatory cells in their bronchoalveolar lavage fluid, mucin over production, and airway hyperresponsiveness. However, genetic depletion of epinephrine using mice lacking the enzyme to synthesize epinephrine, phenylethanolamine N-methyltransferase, or mice that had undergone pharmacological sympathectomy with reserpine to deplete epinephrine, had complete attenuation of these three cardinal features of the asthma phenotype. Furthermore, administration of the long-acting ß2AR agonist, formoterol, a drug currently used in asthma treatment, to phenylethanolamine N-methyltransferase-null mice restored the asthma phenotype. We conclude that ß2AR agonist-induced activation is needed for pathogenesis of the asthma phenotype. These findings also rule out constitutive signaling by the ß2AR as sufficient to drive the asthma phenotype, and may help explain why chronic administration of ß2AR agonists, such as formoterol, have been associated with adverse outcomes in asthma. These data further support the hypothesis that chronic asthma management may be better served by treatment with certain "ß-blockers."


Subject(s)
Adrenergic beta-2 Receptor Agonists/pharmacology , Asthma/chemically induced , Disease Models, Animal , Ethanolamines/pharmacology , Animals , Asthma/physiopathology , Bronchi/physiopathology , Bronchoalveolar Lavage Fluid , Chromatography, High Pressure Liquid , Epinephrine/metabolism , Formoterol Fumarate , Mice , Mice, Knockout , Mucins/metabolism , Phenotype
9.
Proc Natl Acad Sci U S A ; 106(7): 2435-40, 2009 Feb 17.
Article in English | MEDLINE | ID: mdl-19171883

ABSTRACT

Chronic regular use of beta(2)-adrenoceptor (beta(2)-AR) agonists in asthma is associated with a loss of disease control and increased risk of death. Conversely, we have found that administration of beta(2)-AR inverse agonists results in attenuation of the asthma phenotype in an allergen-driven murine model. Besides antagonizing agonist-induced signaling and reducing signaling by empty receptors, beta-AR inverse agonists can also activate signaling by novel pathways. To determine the mechanism of the beta-AR inverse agonists, we compared the asthma phenotype in beta(2)-AR-null and wild-type mice. Antigen challenge of beta(2)-AR-null mice produced results similar to what was observed with chronic beta(2)-AR inverse agonist treatment, namely, reductions in mucous metaplasia, airway hyperresponsiveness (AHR), and inflammatory cells in the lungs. These results indicate that the effects of beta(2)-AR inverse agonists are caused by inhibition of beta(2)-AR signaling rather than by the induction of novel signaling pathways. Chronic administration of alprenolol, a beta-blocker without inverse agonist properties, did not attenuate the asthma phenotype, suggesting that it is signaling by empty receptors, rather than agonist-induced beta(2)-AR signaling, that supports the asthma phenotype. In conclusion, our results demonstrate that, in a murine model of asthma, beta(2)-AR signaling is required for the full development of three cardinal features of asthma: mucous metaplasia, AHR, and the presence of inflammatory cells in the lungs.


Subject(s)
Asthma/genetics , Asthma/pathology , Receptors, Adrenergic, beta-2/metabolism , Alprenolol/pharmacology , Animals , Bronchoconstrictor Agents/pharmacology , Disease Models, Animal , Epithelial Cells/metabolism , Humans , Lung/pathology , Methacholine Chloride/pharmacology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Phenotype , Signal Transduction
10.
FASEB Bioadv ; 4(12): 758-774, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36479208

ABSTRACT

The ß2AR is a prototypical G protein-coupled receptor (GPCR) known to orchestrate different cellular responses by the stimulation of specific signaling pathways. The best-established signaling pathways for the ß2AR are the canonical Gs pathway and the alternative ß arrestin 2 (ßarr2) pathway. Exploring each pathway separately remains a challenging task due to the dynamic nature of the receptor. Here, we fused the ß2AR with its cognate transducers, Gαs and ßarr2, using short linkers as a novel approach for restricting the conformation of the receptor and preferentially activating one of its two signaling pathways. We characterized the behavior of our fusion proteins ß2AR-Gαs and ß2AR-ßarr2 in HEK293 cells by measuring their constitutive activity, transducer recruitment, and pharmacological modulation. Our fusion proteins show (a) steric hindrance from the reciprocal endogenous transducers, (b) constitutive activity of the ß2AR for the signaling pathway activated by the tethered transducer, and (c) pharmacologic modulation by ß2AR ligands. Based on these characteristics, we further explored the possibility of a gain-of-function mechanism in the human lung non-tumorigenic epithelial cell line, BEAS-2B cells. This immortalized human bronchial epithelial cell line has immunomodulatory properties through cytokine release mediated by ß2AR stimulation. Our findings suggest that each signaling pathway of the ß2AR is biased toward either the Th1 or Th2 inflammatory response suggesting a role in regulating the immune phenotype of respiratory diseases. Our data imply that our fusion proteins can be used as tools to isolate the function elicited by a single signaling pathway in physiologically relevant cell types.

11.
Commun Biol ; 5(1): 212, 2022 03 08.
Article in English | MEDLINE | ID: mdl-35260793

ABSTRACT

Internalization of membrane proteins plays a key role in many physiological functions; however, highly sensitive and versatile technologies are lacking to study such processes in real-time living systems. Here we describe an assay based on bioluminescence able to quantify membrane receptor trafficking for a wide variety of internalization mechanisms such as GPCR internalization/recycling, antibody-mediated internalization, and SARS-CoV2 viral infection. This study represents an alternative drug discovery tool to accelerate the drug development for a wide range of physiological processes, such as cancer, neurological, cardiopulmonary, metabolic, and infectious diseases including COVID-19.


Subject(s)
Drug Discovery/methods , Membrane Proteins , Protein Transport/physiology , Spectrometry, Fluorescence/methods , COVID-19 , Drug Development/methods , HEK293 Cells , Humans , Luciferases/genetics , Luciferases/metabolism , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Microscopy, Fluorescence , Nanotechnology , Receptors, G-Protein-Coupled , SARS-CoV-2/chemistry , SARS-CoV-2/metabolism , Virus Internalization
12.
Front Pharmacol ; 13: 1049640, 2022.
Article in English | MEDLINE | ID: mdl-36561339

ABSTRACT

Drug repurposing can overcome both substantial costs and the lengthy process of new drug discovery and development in cancer treatment. Some Food and Drug Administration (FDA)-approved drugs have been found to have the potential to be repurposed as anti-cancer drugs. However, the progress is slow due to only a handful of strategies employed to identify drugs with repurposing potential. In this study, we evaluated GPCR-targeting drugs by high throughput screening (HTS) for their repurposing potential in triple-negative breast cancer (TNBC) and drug-resistant human epidermal growth factor receptor-2-positive (HER2+) breast cancer (BC), due to the dire need to discover novel targets and drugs in these subtypes. We assessed the efficacy and potency of drugs/compounds targeting different GPCRs for the growth rate inhibition in the following models: two TNBC cell lines (MDA-MB-231 and MDA-MB-468) and two HER2+ BC cell lines (BT474 and SKBR3), sensitive or resistant to lapatinib + trastuzumab, an effective combination of HER2-targeting therapies. We identified six drugs/compounds as potential hits, of which 4 were FDA-approved drugs. We focused on ß-adrenergic receptor-targeting nebivolol as a candidate, primarily because of the potential role of these receptors in BC and its excellent long-term safety profile. The effects of nebivolol were validated in an independent assay in all the cell line models. The effects of nebivolol were independent of its activation of ß3 receptors and nitric oxide production. Nebivolol reduced invasion and migration potentials which also suggests its inhibitory role in metastasis. Analysis of the Surveillance, Epidemiology and End Results (SEER)-Medicare dataset found numerically but not statistically significant reduced risk of all-cause mortality in the nebivolol group. In-depth future analyses, including detailed in vivo studies and real-world data analysis with more patients, are needed to further investigate the potential of nebivolol as a repurposed therapy for BC.

13.
Behav Pharmacol ; 22(5-6): 385-9, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21712710

ABSTRACT

The acute and chronic effects of certain drugs can often be opposite. For example, in congestive heart failure acute administration of ß-adrenoceptor agonists results in beneficial improvement in symptoms of the disease, but their chronic use increases mortality. Conversely, certain ß-adrenoceptor antagonists/inverse agonists (ß-blockers) initially cause a detrimental response by decreasing cardiac contractility in congestive heart failure, whereas chronic treatment with the same ß-blockers improves contractility and survival. Furthermore, this time-dependent reversal of outcomes occurs in nonpharmacological interventions, such as exercise, and can even be observed in the response of plants to pruning or other stressors, with the results being a different short-term versus long-term effect. Here, we review some of these phenomena with a special emphasis on the temporal dissociation of pharmacological effects. Although Francis Colpaert used this knowledge to lead a drug discovery project for an analgesic compound that initially produced hyperalgesia, we focused on examples outside the central nervous system.


Subject(s)
Adrenergic beta-Antagonists/pharmacology , Asthma/drug therapy , Heart Failure/drug therapy , Adrenergic beta-Agonists/administration & dosage , Adrenergic beta-Agonists/adverse effects , Adrenergic beta-Agonists/pharmacology , Adrenergic beta-Antagonists/administration & dosage , Adrenergic beta-Antagonists/adverse effects , Analgesics/administration & dosage , Analgesics/adverse effects , Analgesics/pharmacology , Asthma/physiopathology , Drug Inverse Agonism , Heart Failure/physiopathology , Humans , Hyperalgesia/chemically induced , Pain/drug therapy , Time Factors
14.
Front Cardiovasc Med ; 8: 709509, 2021.
Article in English | MEDLINE | ID: mdl-34447792

ABSTRACT

Cardiovascular (CV) diseases are the major cause of death in industrialized countries. The main function of the CV system is to deliver nutrients and oxygen to all tissues. During most CV pathologies, oxygen and nutrient delivery is decreased or completely halted. Several mechanisms, including increased oxygen transport and delivery, as well as increased blood flow are triggered to compensate for the hypoxic state. If the compensatory mechanisms fail to sufficiently correct the hypoxia, irreversible damage can occur. Thus, hypoxia plays a central role in the pathogenesis and pathophysiology of CV diseases. Hypoxia inducible factors (HIFs) orchestrate the gene transcription for hundreds of proteins involved in erythropoiesis, glucose transport, angiogenesis, glycolytic metabolism, reactive oxygen species (ROS) handling, cell proliferation and survival, among others. The overall regulation of the expression of HIF-dependent genes depends on the severity, duration, and location of hypoxia. In the present review, common CV diseases were selected to illustrate that HIFs, and proteins derived directly or indirectly from their stabilization and activation, are related to the development and perpetuation of hypoxia in these pathologies. We further classify CV diseases into acute and chronic hypoxic states to better understand the temporal relevance of HIFs in the pathogenesis, disease progression and clinical outcomes of these diseases. We conclude that HIFs and their derived factors are fundamental in the genesis and progression of CV diseases. Understanding these mechanisms will lead to more effective treatment strategies leading to reduced morbidity and mortality.

15.
Curr Opin Pulm Med ; 16(1): 1-5, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19887938

ABSTRACT

PURPOSE OF REVIEW: beta2-Adrenoceptor (AR) agonists are the most effective bronchodilators known, and play important roles in every step of asthma therapy. Intrinsic efficacy is an important pharmacological property that differentiates the clinical effects and safety profile of beta2-AR agonists. We review the role of beta2-AR agonist intrinsic efficacy in asthma treatment focusing on recent literature. RECENT FINDINGS: In acute asthma, a full agonist (high intrinsic efficacy) offers a clinical advantage over a partial agonist (low intrinsic efficacy) but with the potential of inducing dose-dependent adverse effects. The chronic use of beta2-AR agonists may be associated with several adverse outcomes including loss of asthma control and even increased mortality. Recently, the role of beta-AR inverse agonists (beta-blockers) which have a negative intrinsic efficacy was studied. Whereas contraindicated in acute asthma, preliminary data suggest that the chronic use of these agents may be associated with attenuation of airway hyper-responsiveness in patients with mild asthma. Studies in a murine model of asthma suggest that such effects may be related to decreased airway inflammation and mucous metaplasia. SUMMARY: Rational choice among beta2-AR agonists in acute and chronic asthma should be influenced by differences in intrinsic efficacy among these agents. In acute severe asthma, a full agonist offers a clinical advantage over a partial agonist. Whereas the use of inverse agonists in the treatment of asthma is still experimental and needs further exploration in future trials, preliminary studies suggest that their chronic use is well tolerated and is associated with decreased airway hyper-responsiveness.


Subject(s)
Adrenergic beta-Agonists/therapeutic use , Asthma/drug therapy , Bronchodilator Agents/therapeutic use , Adrenergic beta-Agonists/adverse effects , Asthma/physiopathology , Bronchodilator Agents/adverse effects , Dose-Response Relationship, Drug , Humans , Respiratory Hypersensitivity/physiopathology , Treatment Outcome
17.
Neurosci Lett ; 725: 134892, 2020 04 23.
Article in English | MEDLINE | ID: mdl-32165259

ABSTRACT

Benzodiazepines and SSRIs are considered as standard treatment options for anxiety and depression, hallmarks of Post-Traumatic Stress Disorder (PTSD), although their use is often limited by adverse effects. While promising evidence emerged with ß-adrenergic receptor (ß-AR) antagonists (or 'ß-blockers') and PTSD relief, efficacy issues dampened the excitement. However, we believe it is premature to completely eliminate a beneficial role of ß-blockers. Our previous work has suggested that social defeat (SD) results in anxiety-like and depression-like behaviors in rats. Here, using the SD paradigm, we examined the effect of several ß-adrenergic receptor antagonists (propranolol, nadolol, bisoprolol) on these behaviors in rats. Following acclimatization, Sprague-Dawley rats received no treatment (for control groups) or treated with ; propranolol (50 mg/kg/day in water), or nadolol (18 mg/kg/day in rats' chow), or bisoprolol (15 mg/kg/day in water). The treatment lasted for 36 days, following which rats were subjected to SD/control exposures (1 week). Later, anxiety-like and depression-like behaviors, social interaction and learning-memory function tests were conducted. SD rats exhibited anxiety- and depression-like behavior as well as learning-memory impairment. Propranolol and nadolol protected SD rats from exhibiting anxiety-or depression-like behaviors. Bisoprolol treatment did not mitigate SD-induced behavioral impairments in rats. Nadolol, propranolol or bisoprolol have no effect in attenuating SD-induced memory function tests. These results suggest that certain 'ß-blockers' have the potential to mitigate the negative psychological effects of traumatic events.


Subject(s)
Adrenergic beta-Antagonists/therapeutic use , Nadolol/therapeutic use , Neuroprotective Agents/therapeutic use , Propranolol/therapeutic use , Social Defeat , Stress, Psychological/drug therapy , Adrenergic beta-Antagonists/pharmacology , Animals , Male , Maze Learning/drug effects , Maze Learning/physiology , Nadolol/pharmacology , Neuroprotective Agents/pharmacology , Propranolol/pharmacology , Random Allocation , Rats , Rats, Long-Evans , Rats, Sprague-Dawley , Social Interaction/drug effects , Stress, Psychological/psychology
18.
J Tissue Eng Regen Med ; 14(2): 306-318, 2020 02.
Article in English | MEDLINE | ID: mdl-31821703

ABSTRACT

Clinical trials using human adipogenic mesenchymal stem cells (hAdMSCs) for the treatment of cardiac diseases have shown improvement in cardiac function and were proven safe. However, hAdMSCs do not convert efficiently into cardiomyocytes (CMs) or vasculature. Thus, reprogramming hAdMSCs into myocyte progenitors may fare better in future investigations. To reprogramme hAdMSCs into electrically conductive cardiac progenitor cells, we pioneered a three-step reprogramming strategy that uses proven MESP1/ETS2 transcription factors, ß-adrenergic and hypoxic signalling induced in three-dimensional (3D) cardiospheres. In Stage 1, ETS2 and MESP1 activated NNKX2.5, TBX5, MEF2C, dHAND, and GATA4 during the conversion of hAdMSCs into cardiac progenitor cells. Next, in Stage 2, ß2AR activation repositioned cardiac progenitors into de novo immature conductive cardiac cells, along with the appearance of RYR2, CAV2.1, CAV3.1, NAV1.5, SERCA2, and CX45 gene transcripts and displayed action potentials. In Stage 3, electrical conduction that was fostered by 3D cardiospheres formed in a Synthecon®, Inc. rotating bioreactor induced the appearance of hypoxic genes: HIF-1α/ß, PCG 1α/ß, and NOS2, which coincided with the robust activation of adult contractile genes including MLC2v, TNNT2, and TNNI3, ion channel genes, and the appearance of hyperpolarization-activated and cyclic nucleotide-gated channels (HCN1-4). Conduction velocities doubled to ~200 mm/s after hypoxia and doubled yet again after dissociation of the 3D cell clusters to ~400 mm/s. By comparison, normal conduction velocities within working ventricular myocytes in the whole heart range from 0.5 to 1 m/s. Epinephrine stimulation of stage 3 cardiac cells in patches resulted in an increase in amplitude of the electrical wave, indicative of conductive cardiac cells. Our efficient protocol that converted hAdMSCs into highly conductive cardiac progenitors demonstrated the potential utilization of stage 3 cells for tissue engineering applications for cardiac repair.


Subject(s)
Cell Culture Techniques/methods , Mesenchymal Stem Cells/cytology , Receptors, Adrenergic, beta/metabolism , Adipogenesis , Adrenergic Agents , Bioreactors , Cell Differentiation/physiology , Cell Proliferation , Electric Conductivity , Epinephrine/pharmacology , Humans , Hypoxia , Kinetics , Myocytes, Cardiac/cytology , Signal Transduction , Tissue Engineering/instrumentation , Tissue Engineering/methods , Tissue Scaffolds , Transcription Factors/metabolism
19.
Cell Stress Chaperones ; 25(6): 993-1012, 2020 11.
Article in English | MEDLINE | ID: mdl-32779001

ABSTRACT

The adoptive transfer of donor-derived virus-specific T cells (VSTs) is an effective treatment for infections following allogeneic hematopoietic cell transplantation. Acute exercise mobilizes effector lymphocytes and VSTs to the circulation and augments the ex vivo manufacture of VSTs. This study determined if ß2 adrenergic receptor (AR) signaling precipitated the VST response to acute exercise. Healthy participants (n = 12) completed 30 min of steady-state cycling exercise after ingesting a placebo, a ß1 + 2 AR antagonist (nadolol) or a ß1 AR antagonist (bisoprolol). Circulating VSTs to cytomegalovirus (CMV), Epstein-Barr virus (EBV), and adenovirus (AdV) antigens were enumerated before and after exercise, and peripheral blood mononuclear cells were cultured with viral peptides for 8 days to expand multi-VSTs. Compared with placebo, nadolol blunted the exercise-induced mobilization of CMV-VSTs (Δ VSTs/100,000 CD3+ T cells = 93 ± 104 vs. 22 ± 91 for placebo and nadolol, respectively; p = 0.036), while bisoprolol did not, despite both drugs evoking similar reductions in exercising heart rate and blood pressure. Circulating AdV and EBV VSTs (VSTs/mL blood) only increased after exercise with placebo. Although not significant, nadolol partially mitigated exercise-induced increases in multi-VST expansion, particularly in participants that demonstrated an exercise-induced increase in VST expansion. We conclude that exercise-induced enhancements in VST mobilization and expansion are at least partially ß2 AR mediated, thus highlighting a role for the ß2 AR in targeted therapy for the augmentation of VST immune cell therapeutics in the allogeneic adoptive transfer setting. Moreover, long-term regular exercise may provide additional viral protection in the host through frequent ß2 AR-dependent mobilization and redistribution of VSTs cumulated with each bout of exercise.


Subject(s)
Adrenergic beta-1 Receptor Antagonists/pharmacology , Adrenergic beta-2 Receptor Antagonists/pharmacology , Antiviral Agents/pharmacology , Cell- and Tissue-Based Therapy , Exercise , T-Lymphocytes/immunology , Viruses/immunology , Adult , Blood Pressure/drug effects , Catecholamines/blood , Cell Proliferation/drug effects , Cytokines/metabolism , Female , Heart Rate/drug effects , Humans , Hydrocortisone/blood , Lactic Acid/blood , Male , Peptides/pharmacology , Phenotype , Receptors, Adrenergic, beta/metabolism , Species Specificity , T-Lymphocytes/drug effects , Young Adult
20.
Br J Pharmacol ; 176(14): 2339-2342, 2019 07.
Article in English | MEDLINE | ID: mdl-31240712

ABSTRACT

LINKED ARTICLES: This article is part of a themed section on Adrenoceptors-New Roles for Old Players. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v176.14/issuetoc.


Subject(s)
Receptors, Adrenergic/metabolism , Animals , Humans
SELECTION OF CITATIONS
SEARCH DETAIL