Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 635
Filter
1.
Am J Physiol Cell Physiol ; 321(5): C884-C896, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34613841

ABSTRACT

Moderate elevations of extracellular K+ concentration ([K+]o) occur during exercise and have been shown to potentiate force during contractions elicited with subtetanic frequencies. Here, we investigated whether lactic acid (reduced chloride conductance), ß2-adrenoceptor activation, and increased temperature would influence the potentiating effect of potassium in slow- and fast-twitch muscles. Isometric contractions were elicited by electrical stimulation at various frequencies in isolated rat soleus and extensor digitorum longus (EDL) muscles incubated at normal (4 mM) or elevated K+, in combination with salbutamol (5 µM), lactic acid (18.1 mM), 9-anthracene-carboxylic acid (9-AC; 25 µM), or increased temperature (30-35°C). Elevating [K+]o from 4 mM to 7 mM (soleus) and 10 mM (EDL) potentiated isometric twitch and subtetanic force while slightly reducing tetanic force. In EDL, salbutamol further augmented twitch force (+27 ± 3%, P < 0.001) and subtetanic force (+22 ± 4%, P < 0.001). In contrast, salbutamol reduced subtetanic force (-28 ± 6%, P < 0.001) in soleus muscles. Lactic acid and 9-AC had no significant effects on isometric force of muscles already exposed to moderate elevations of [K+]o. The potentiating effect of elevated [K+]o was still well maintained at 35°C. Addition of salbutamol exerts a further force-potentiating effect in fast-twitch but not in slow-twitch muscles already potentiated by moderately elevated [K+]o, whereas lactic acid, 9-AC, or increased temperature does not exert any further augmentation. However, the potentiating effect of elevated [K+]o was still maintained in the presence of these, thus emphasizing the positive influence of moderately elevated [K+]o for contractile performance during exercise.


Subject(s)
Adrenergic beta-2 Receptor Agonists/pharmacology , Albuterol/pharmacology , Lactic Acid/pharmacology , Muscle Contraction/drug effects , Muscle Strength/drug effects , Muscle, Skeletal/drug effects , Potassium/metabolism , Receptors, Adrenergic, beta-2/drug effects , Temperature , Animals , Anthracenes/pharmacology , Electric Stimulation , Female , In Vitro Techniques , Male , Muscle, Skeletal/physiology , Rats, Wistar , Receptors, Adrenergic, beta-2/metabolism
2.
Biomed Pharmacother ; 144: 112331, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34673421

ABSTRACT

Chemotherapy-induced neuropathic pain is a debilitating and common side effect of cancer treatment and so far no effective drug is available for treatment of the serious side effect. Previous studies have demonstrated ß2-adrenoreceptor (ADRB2) agonists can attenuate neuropathic pain. However, the role of ADRB2 in paclitaxel -induced neuropathic pain (PINP) remains unclear. In this study, we investigated the effect of formoterol, a long-acting ADRB2 agonist, and related mechanisms in PINP. A rat model of PINP was established by intraperitoneal injection of paclitaxel (2 mg/kg) every other day with a final cumulative dose of 8 mg/kg. Hind paw withdrawal thresholds (PWTs) in response to von Frey filament stimuli were used to evaluate mechanical allodynia. Western blot was used to examine the expression of ADRB2, peroxisome proliferator-activated receptor coactivator-1α (PGC-1α), nuclear respiratory factors 1 (NRF1) and mitochondrial transcription factor A (TFAM) and the immunofluorescence was to detect the cellular localization of ADRB2 and PGC-1α in the spinal cord. Moreover, we measured mitochondrial DNA (mtDNA) copy number by qPCR. In our study, formoterol attenuated established PINP and delayed the onset of PINP. Formoterol restored ADRB2 expression as well as mtDNA copy number and PGC-1α, NRF1, and TFAM protein expression, which are major genes involved in mitochondrial biogenesis, in the spinal cord of PINP rats. Moreover, we found the analgesic effect of formoterol against PINP was partially abolished by PGC-1α inhibitor SR-18292. Collectively, these results demonstrated the activation of ADRB2 with formoterol ameliorates PINP at least partially through induction of mitochondrial biogenesis.


Subject(s)
Adrenergic beta-2 Receptor Agonists/pharmacology , Analgesics/pharmacology , Formoterol Fumarate/pharmacology , Mitochondria/drug effects , Neuralgia/drug therapy , Organelle Biogenesis , Pain Threshold/drug effects , Receptors, Adrenergic, beta-2/drug effects , Spinal Cord/drug effects , Animals , DNA, Mitochondrial/genetics , DNA, Mitochondrial/metabolism , Disease Models, Animal , Male , Mitochondria/genetics , Mitochondria/metabolism , Neuralgia/genetics , Neuralgia/metabolism , Neuralgia/physiopathology , Paclitaxel , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Rats, Sprague-Dawley , Receptors, Adrenergic, beta-2/genetics , Receptors, Adrenergic, beta-2/metabolism , Spinal Cord/metabolism , Spinal Cord/physiopathology
3.
Toxicol In Vitro ; 77: 105251, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34601065

ABSTRACT

ß2-adrenoceptor (ß2AR) agonists can stimulate skeletal muscle growth. Their illegal use in food-producing animals, human athletes and bodybuilders causes adverse health effects. In the present study, we developed 3D-QSAR models for predicting the activity of chemicals which can stimulate skeletal muscle growth through ß2AR. The activity of 25 ß2AR agonists was measured by ß2AR-cAMP response element (CRE) -luciferase (Luc) reporter assay. The 3D-QSAR models were built using comparative molecular field analysis (CoMFA) and comparative molecular similarity indices analysis (CoMSIA). The CoMFA and CoMSIA models displayed high external predictability (R2 0.996 and 0.992, respectively) and good statistical robustness, and revealed that electrostatic effects were the most prominent forces influencing the activity of ß2AR agonists. The CoMFA and CoMSIA contour plots provided clues regarding the main chemical features responsible for the activity variations and also resulted in predictions which correlate very well with the observed activity. In vitro study with differentiated myotubes showed that the potency orders of ß2AR agonists in activating the ß2AR-CRE-Luc reporter and in upregulating CREB target genes related to muscle growth were consistent. These 3D-QSAR models provide tools for predicting the activity of chemicals which might be illegally used in livestock or humans to stimulate skeletal muscle growth.


Subject(s)
Adrenergic beta-2 Receptor Agonists/pharmacology , Models, Molecular , Muscle Development/drug effects , Quantitative Structure-Activity Relationship , Receptors, Adrenergic, beta-2/drug effects , Animals , HEK293 Cells/drug effects , Humans , Mice , Real-Time Polymerase Chain Reaction
4.
Am J Respir Crit Care Med ; 204(10): 1143-1152, 2021 11 15.
Article in English | MEDLINE | ID: mdl-34464242

ABSTRACT

Rationale: Inhaled corticosteroids (ICS) are commonly prescribed with long-acting ß2-agonists (LABA) in chronic obstructive pulmonary disease (COPD). To date, the effects of ICS therapy on the airway microbiome in COPD are unknown. Objectives: To determine the effects of ICS/LABA on the airway microbiome of patients with COPD. Methods: Clinically stable patients with COPD were enrolled into a 4-week run-in period during which ICS was discontinued and all participants were placed on formoterol (Form) 12 µg twice daily (BID). The participants were then randomized to budesonide/formoterol (Bud + Form; 400/12 µg BID), fluticasone/salmeterol (Flu + Salm; 250/50 µg BID), or formoterol only (12 µg BID) for 12 weeks. Participants underwent bronchoscopy before and after the 12-week treatment period. The primary endpoint was the comparison of changes in the airway microbiome over the trial period between the ICS/LABA and LABA-only groups. Measurements and Main Results: Sixty-three participants underwent randomization: Bud + Form (n = 20), Flu + Salm (n = 22), and Form (n = 21) groups; 56 subjects completed all visits. After the treatment period, changes in α-diversity were significantly different across groups, especially between Flu + Salm and Form groups (Δrichness: P = 0.02; ΔShannon index: P = 0.03). Longitudinal differential abundance analyses revealed more pronounced microbial shifts from baseline in the fluticasone (vs. budesonide or formoterol only) group. Conclusions: Fluticasone-based ICS/LABA therapy modifies the airway microbiome in COPD, leading to a relative reduction in α-diversity and a greater number of bacterial taxa changes. These data may have implications in patients who develop pneumonia on ICS. Clinical trial registered with www.clinicaltrials.gov (NCT02833480).


Subject(s)
Adrenal Cortex Hormones/therapeutic use , Adrenergic beta-2 Receptor Agonists/therapeutic use , Drug Combinations , Microbiota/drug effects , Pulmonary Disease, Chronic Obstructive/drug therapy , Receptors, Adrenergic, beta-2/drug effects , Receptors, Adrenergic, beta-2/therapeutic use , Administration, Inhalation , Adult , Aged , Aged, 80 and over , Female , Humans , Male , Middle Aged , Treatment Outcome
5.
Front Immunol ; 12: 628065, 2021.
Article in English | MEDLINE | ID: mdl-34220796

ABSTRACT

Objective: Hypersympathetic activity is prominent in rheumatoid arthritis, and major life stressors precede onset in ~80% of patients. These findings and others support a link between stress, the sympathetic nervous system and disease onset and progression. Here, we extend previous research by evaluating how selective peripherally acting α/ß2-adrenergic drugs affect joint destruction in adjuvant-induced arthritis. Methods: Complete Freund's adjuvant induced inflammatory arthritis in male Lewis rats. Controls received no treatment. Arthritic rats then received vehicle or twice-daily treatment with the α-adrenergic antagonist, phentolamine (0.5 mg/day) and the ß2-adrenergic agonist, terbutaline (1200 µg/day, collectively named SH1293) from day (D) of disease onset (D12) through acute (D21) and severe disease (D28). Disease progression was assessed in the hind limbs using dorsoplantar widths, X-ray analysis, micro-computed tomography, and routine histology on D14, D21, and D28 post-immunization. Results: On D21, SH1293 significantly attenuated arthritis in the hind limbs, based on reduced lymphocytic infiltration, preservation of cartilage, and bone volume. Pannus formation and sympathetic nerve loss were not affected by SH1293. Bone area and osteoclast number revealed high- and low-treatment-responding groups. In high-responding rats, treatment with SH1293 significantly preserved bone area and decreased osteoclast number, data that correlated with drug-mediated joint preservation. SH1293 suppressed abnormal bone formation based on reduced production of osteophytes. On D28, the arthritic sparing effects of SH1293 on lymphocytic infiltration, cartilage and bone sparing were maintained at the expense of bone marrow adipocity. However, sympathetic nerves were retracted from the talocrural joint. Conclusion and Significance: Our findings support a significant delay in early arthritis progression by treatment with SH1293. Targeting sympathetic neurotransmission may provide a strategy to slow disease progression.


Subject(s)
Adrenergic alpha-Antagonists/pharmacology , Adrenergic beta-2 Receptor Agonists/pharmacology , Arthritis, Experimental/prevention & control , Joints/drug effects , Phentolamine/pharmacology , Receptors, Adrenergic, alpha/drug effects , Receptors, Adrenergic, beta-2/drug effects , Terbutaline/pharmacology , Animals , Arthritis, Experimental/chemically induced , Arthritis, Experimental/metabolism , Arthritis, Experimental/pathology , Drug Combinations , Freund's Adjuvant , Joints/diagnostic imaging , Joints/metabolism , Joints/pathology , Male , Rats, Inbred Lew , Receptors, Adrenergic, alpha/metabolism , Receptors, Adrenergic, beta-2/metabolism , Signal Transduction
6.
Am J Physiol Regul Integr Comp Physiol ; 320(5): R575-R587, 2021 05 01.
Article in English | MEDLINE | ID: mdl-33565369

ABSTRACT

Approximately 14% of the general population suffer from chronic kidney disease that can lead to acute kidney injury (AKI), a condition with up to 50% mortality for which there is no effective treatment. Hypertension, diabetes, and cardiovascular disease are the main comorbidities, and more than 660,000 Americans have kidney failure. ß2-Adrenergic receptors (ß2ARs) have been extensively studied in association with lung and cardiovascular disease, but with limited scope in kidney and renal diseases. ß2ARs are expressed in multiple parts of the kidney including proximal and distal convoluted tubules, glomeruli, and podocytes. Classical and noncanonical ß2AR signaling pathways interface with other intracellular mechanisms in the kidney to regulate important cellular functions including renal blood flow, electrolyte balance and salt handling, and tubular function that in turn exert control over critical physiology and pathology such as blood pressure and inflammatory responses. Nephroprotection through activation of ß2ARs has surfaced as a promising field of investigation; however, there is limited data on the pharmacology and potential side effects of renal ß2AR modulation. Here, we provide updates on some of the major areas of preclinical kidney research involving ß2AR signaling that have advanced to describe molecular pathways and identify potential drug targets some of which are currently under clinical development for the treatment of kidney-related diseases.


Subject(s)
Adrenergic beta-2 Receptor Agonists/therapeutic use , Kidney Diseases/drug therapy , Kidney/drug effects , Receptors, Adrenergic, beta-2/drug effects , Water-Electrolyte Balance/drug effects , Animals , Humans , Kidney/metabolism , Kidney/physiopathology , Kidney Diseases/metabolism , Kidney Diseases/physiopathology , Receptors, Adrenergic, beta-2/metabolism , Signal Transduction
7.
Biochim Biophys Acta Proteins Proteom ; 1869(4): 140605, 2021 04.
Article in English | MEDLINE | ID: mdl-33453412

ABSTRACT

The ß2-adrenergic receptor (ß2AR) is a member of the G protein-coupled receptor (GPCR) family that is an important drug target for asthma and COPD. Clinical studies coupled with biochemical data have identified a critical receptor variant, Thr164Ile, to have a reduced response to agonist-based therapy, although the molecular mechanism underlying this seemingly "non-deleterious" substitution is not clear. Here, we couple molecular dynamics simulations with network analysis and free-energy calculations to identify the molecular determinants underlying the differential drug response. We are able to identify hydration sites in the transmembrane domain that are essential to maintain the integrity of the binding site but are absent in the variant. The loss of these hydration sites in the variant correlates with perturbations in the intra-protein interaction network and rearrangements in the orthosteric ligand binding site. In conjunction, we observe an altered binding and reduced free energy of a series of agonists, in line with experimental trends. Our work identifies a functional allosteric pathway connected by specific hydration sites in ß2AR that has not been reported before and provides insight into water-mediated networks in GPCRs in general. Overall, the work is one of the first step towards developing variant-specific potent and selective agonists.


Subject(s)
Adrenergic beta-Agonists/pharmacology , Receptors, Adrenergic, beta-2/drug effects , Water/chemistry , Humans , Protein Binding , Protein Conformation , Receptors, Adrenergic, beta-2/chemistry
8.
Life Sci ; 265: 118864, 2021 Jan 15.
Article in English | MEDLINE | ID: mdl-33301808

ABSTRACT

Beta-2 adrenergic receptors (ß2-ARs) have important roles in the pathogenesis and treatment of chronic obstructive pulmonary disease (COPD). In recent years, progress has been made in the study of ß2-ARs. Here, we introduce the basic concepts of ß2-ARs, related pathways, as well as application of blockers/agonists of ß2-ARs, and ß2-AR autoantibodies in COPD. Drugs targeting the ß2-AR are being developed rapidly, and we expect them to improve the symptoms and prognosis of COPD patients in the future.


Subject(s)
Pulmonary Disease, Chronic Obstructive/physiopathology , Receptors, Adrenergic, beta-2/metabolism , Adrenergic beta-2 Receptor Agonists/pharmacology , Adrenergic beta-2 Receptor Antagonists/pharmacology , Autoantibodies/immunology , Drug Development , Humans , Prognosis , Pulmonary Disease, Chronic Obstructive/drug therapy , Pulmonary Disease, Chronic Obstructive/immunology , Receptors, Adrenergic, beta-2/drug effects , Receptors, Adrenergic, beta-2/immunology
9.
Toxicol Appl Pharmacol ; 411: 115366, 2021 01 15.
Article in English | MEDLINE | ID: mdl-33316273

ABSTRACT

Mitochondrial dysfunction is a well-characterized consequence of spinal cord injury (SCI). We previously reported that treatment with the FDA-approved ß2-adrenergic receptor agonist formoterol beginning 8 h post-SCI induces mitochondrial biogenesis (MB) and improves body composition and locomotor recovery in female mice. To determine the time-to-treatment window of formoterol, female mice were subjected to 80 kdyn contusion SCI and daily administration of vehicle or formoterol (0.3 mg/kg) beginning 24 h after injury. This delayed treatment paradigm improved body composition in female mice by 21 DPI, returning body weight to pre-surgery weight and restoring gastrocnemius mass to sham levels; however, there was no effect on locomotor recovery, as measured by the Basso-Mouse Scale (BMS), or lesion volume. To assess the cross-sex potential of formoterol, injured male mice were treated with vehicle or formoterol (0.3 or 1.0 mg/kg) beginning 8 h after SCI. Formoterol also improved body composition post-SCI in male mice, restoring body weight and muscle mass regardless of dose. Interestingly, however, improved BMS scores and decreased lesion volume was observed only in male mice treated with 0.3 mg/kg. Additionally, 0.3 mg/kg formoterol induced MB in the gastrocnemius and injured spinal cord, as evidenced by increased MB protein expression and mitochondrial number. These data indicate that formoterol treatment improves recovery post-SCI in both male and female mice in a dose- and initiation time-dependent manner. Furthermore, formoterol-induced functional recovery post-SCI is not directly associated with peripheral effects, such as muscle mass and body weight.


Subject(s)
Adrenergic beta-2 Receptor Agonists/administration & dosage , Formoterol Fumarate/administration & dosage , Mitochondria, Muscle/drug effects , Muscle, Skeletal/drug effects , Organelle Biogenesis , Receptors, Adrenergic, beta-2/drug effects , Spinal Cord Injuries/drug therapy , Spinal Cord/drug effects , Time-to-Treatment , Animals , Body Composition/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Administration Schedule , Female , Male , Mice, Inbred C57BL , Mitochondria, Muscle/metabolism , Mitochondria, Muscle/ultrastructure , Muscle, Skeletal/metabolism , Muscle, Skeletal/ultrastructure , Receptors, Adrenergic, beta-2/metabolism , Recovery of Function , Sex Factors , Spinal Cord/metabolism , Spinal Cord/ultrastructure , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/pathology , Time Factors
10.
Eur J Pharmacol ; 892: 173733, 2021 Feb 05.
Article in English | MEDLINE | ID: mdl-33220278

ABSTRACT

Over the past decade, dexmedetomidine (DEX) has been found to possess an anti-inflammatory effect. However, the local anti-inflammatory mechanism of DEX has not been fully clarified. Some intracellular inflammatory pathways lead to negative feedback during the inflammatory process. The cyclooxygenase (COX) cascade synthesizes prostaglandins (PGs) and plays a key role in inflammation, but is known to also have anti-inflammatory properties through an alternative route of a PGD2 metabolite, 15-deoxy-delta-12,14-prostaglandin J2 (15d-PGJ2), and its receptor, peroxisome proliferator-activated receptor gamma (PPARγ). Therefore, we hypothesized that DEX inhibits LPS-induced inflammatory responses through 15d-PGJ2 and/or PPARγ activation, and evaluated the effects of DEX on these responses. The RAW264.7 mouse macrophage-like cells were pre-incubated with DEX, followed by the addition of LPS to induce inflammatory responses. Concentrations of TNFα, IL-6, PGE2, and 15d-PGJ2 in the supernatants of the cells were measured, and gene expressions of PPARγ and COX-2 were evaluated in the cells. Furthermore, we evaluated whether a selective α2 adrenoceptor antagonist, yohimbine or a selective PPARγ antagonist, T0070907, reversed the effects of DEX on the LPS-induced inflammatory responses. DEX inhibited LPS-induced TNFα, IL-6, and PGE2 productions and COX-2 mRNA expression, and the effects of DEX were reversed by yohimbine. On the other hand, DEX significantly increased 15d-PGJ2 production and PPARγ mRNA expression, and yohimbine reversed these DEX's effects. Furthermore, T0070907 reversed the anti-inflammatory effects of DEX on TNFα and IL-6 productions in the cells. These results suggest that DEX inhibits LPS-induced inflammatory responses through PPARγ activation following binding to α2 adrenoceptors.


Subject(s)
Adrenergic alpha-2 Receptor Agonists/pharmacology , Anti-Inflammatory Agents/pharmacology , Dexmedetomidine/pharmacology , Inflammation/prevention & control , Macrophages/drug effects , PPAR gamma/agonists , Receptors, Adrenergic, beta-2/drug effects , Adrenergic alpha-2 Receptor Agonists/metabolism , Animals , Anti-Inflammatory Agents/metabolism , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Dexmedetomidine/metabolism , Dinoprostone/metabolism , Inflammation/chemically induced , Inflammation/metabolism , Interleukin-6/metabolism , Lipopolysaccharides/toxicity , Macrophages/metabolism , Macrophages/pathology , Mice , PPAR gamma/genetics , PPAR gamma/metabolism , Prostaglandin D2/analogs & derivatives , Prostaglandin D2/metabolism , Protein Binding , RAW 264.7 Cells , Receptors, Adrenergic, beta-2/metabolism , Signal Transduction , Tumor Necrosis Factor-alpha/metabolism
11.
Front Immunol ; 11: 588724, 2020.
Article in English | MEDLINE | ID: mdl-33117402

ABSTRACT

SARS-CoV-2 infection is a new threat to global public health in the 21st century (2020), which has now rapidly spread around the globe causing severe pneumonia often linked to Acute Respiratory Distress Syndrome (ARDS) and hyperinflammatory syndrome. SARS-CoV-2 is highly contagious through saliva droplets. The structural analysis suggests that the virus enters human cells through the ligation of the spike protein to angiotensin-converting enzyme 2 (ACE2). The progression of Covid-19 has been divided into three main stages: stage I-viral response, stage II-pulmonary phase, and stage III-hyperinflammation phase. Once the patients enter stage III, it will likely need ventilation and it becomes difficult to manage. Thus, it will be of paramount importance to find therapies to prevent or slow down the progression of the disease toward stage III. The key event leading to hyperinflammation seems to be the activation of Th-17 immunity response and Cytokine storm. B2-adrenergic receptors (B2ARs) are expressed on airways and on all the immune cells such as macrophages, dendritic cells, B and T lymphocytes. Blocking (B2AR) has been proven, also in clinical settings, to reduce Th-17 response and negatively modulate inflammatory cytokines including IL-6 while increasing IFNγ. Non-selective beta-blockers are currently used to treat several diseases and have been proven to reduce stress-induced inflammation and reduce anxiety. For these reasons, we speculate that targeting B2AR in the early phase of Covid-19 might be beneficial to prevent hyperinflammation.


Subject(s)
Adrenergic beta-2 Receptor Antagonists/therapeutic use , Coronavirus Infections/drug therapy , Coronavirus Infections/pathology , Cytokine Release Syndrome/drug therapy , Pneumonia, Viral/drug therapy , Pneumonia, Viral/pathology , Receptors, Adrenergic, beta-2/drug effects , Respiratory Distress Syndrome/drug therapy , Betacoronavirus/drug effects , COVID-19 , Cytokine Release Syndrome/pathology , Humans , Inflammation/drug therapy , Inflammation/immunology , Inflammation/pathology , Neoplasms/drug therapy , Neoplasms/pathology , Pandemics , Respiratory Distress Syndrome/pathology , Respiratory Distress Syndrome/virology , SARS-CoV-2 , Th17 Cells/immunology
12.
Aging (Albany NY) ; 12(14): 14300-14313, 2020 07 21.
Article in English | MEDLINE | ID: mdl-32692723

ABSTRACT

Diabetes mellitus (DM) might increase the incidence and mortality of cardiac failure after acute myocardial infarction (AMI) in patients. We attempted to investigate whether Caveolin-3 showed beneficial effects in DM patient post-MI injury through the cAMP/PKA and BDNF/TrkB signaling pathways. The activity of ADRB2 and cAMP/PKA signaling were impaired in nondiabetic ischemia-reperfusion (I/R) group compared with the sham and DM groups and were more impaired in diabetic I/R group than in the I/R group. In H9C2 cells, high-glucose (HG) stimulation further enhanced H/R injury by promoting cell apoptosis, inhibiting cell viability, and suppressing TrkB and Akt signaling; in contrast, the ADRB2 agonist isoprenaline (ISO) significantly attenuated the above-described effects of HG stimulation. Caveolin-3 overexpression promoted the localization of ADRB2 on the membrane of the HG-stimulated H9C2 cells, subsequently inhibiting apoptosis and promoting cell viability. Under HG stimulation, Caveolin-3 overexpression enhanced the activity of the cAMP/PKA and BDNF/TrkB signaling pathways, whereas ADRB2 silencing reversed the effects of Caveolin-3 overexpression. In conclusion, ADRB2 agonist promoted the activity of the BDNF/TrkB and cAMP/PKA signaling pathways, mitigating the HG-aggravated H/R injuries in H9C2 cells. Caveolin-3 exerts a protective effect on diabetic hearts against I/R damage through the ß2AR, cAMP/PKA, and BDNF/TrkB signaling pathways.


Subject(s)
Cardiotonic Agents/therapeutic use , Caveolin 3/therapeutic use , Diabetic Cardiomyopathies/prevention & control , Myocardial Infarction/prevention & control , Myocardial Reperfusion Injury/prevention & control , Signal Transduction/drug effects , Animals , Apoptosis/drug effects , Brain-Derived Neurotrophic Factor , Cell Line , Cyclic AMP , Cyclic AMP-Dependent Protein Kinases , Electrocardiography , Glucose/pharmacology , Male , Rats , Rats, Sprague-Dawley , Receptor, trkB , Receptors, Adrenergic, beta-2/drug effects
13.
Naunyn Schmiedebergs Arch Pharmacol ; 393(11): 2043-2052, 2020 11.
Article in English | MEDLINE | ID: mdl-32500188

ABSTRACT

Circulating catecholamines contribute to the regulation of retinal vascular tone. Our previous studies have demonstrated that the activation of large-conductance Ca2+-activated K+ (BKCa) channels is involved in the ß2-adrenoceptor-mediated dilation of retinal arterioles in rats. The present study aimed to examine the role of Gi protein in the ß2-adrenoceptor-mediated activation of BKCa channels in the retinal arterioles. Images of in vivo rat ocular fundi were captured, and the diameters of retinal arterioles were measured. Systemic blood pressure and heart rate were recorded continuously. Intravenous infusion of formoterol (0.01-0.3 µg/kg/min), a ß2-adrenoceptor agonist, increased the diameter of retinal arterioles but decreased mean arterial pressure in a dose-dependent manner. Intravitreal injection of iberiotoxin (20 pmol/eye), an inhibitor of BKCa channels, significantly attenuated the formoterol-induced dilation of retinal arterioles. Similar results were obtained when salbutamol (0.03-3 µg/kg/min), another ß2-adrenoceptor agonist, was used instead of formoterol. However, iberiotoxin had no significant effect on retinal vasodilator responses to intravenous infusion of denopamine (1-30 µg/kg/min; a ß1-adrenoceptor agonist), CL316243 (0.3-10 µg/kg/min; a ß3-adrenoceptor agonist), prostaglandin I2 (0.03-10 µg/kg/min; a prostanoid IP receptor agonist), and forskolin (1-10 µg/kg/min; an adenylyl cyclase activator). Intravitreal injection of pertussis toxin (66 ng/eye; a Gi protein inhibitor) significantly attenuated the dilation of retinal arterioles induced by formoterol but not by denopamine and CL316243. In the presence of pertussis toxin, iberiotoxin had no inhibitory effect on formoterol-induced dilation of retinal arterioles. These results suggest that stimulation of ß2-adrenoceptors dilates retinal arterioles through pertussis toxin-sensitive Gi protein-dependent activation of BKCa channels in rats in vivo.


Subject(s)
Adrenergic beta-2 Receptor Agonists/pharmacology , Arterioles/drug effects , Formoterol Fumarate/pharmacology , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/metabolism , Receptors, Adrenergic, beta-2/drug effects , Retinal Vessels/drug effects , Vasodilation/drug effects , Albuterol/pharmacology , Animals , Arterioles/metabolism , Calcium Signaling , Male , Rats, Wistar , Receptors, Adrenergic, beta-2/metabolism , Retinal Vessels/metabolism
14.
Biomed Res Int ; 2020: 1693602, 2020.
Article in English | MEDLINE | ID: mdl-32104676

ABSTRACT

Salbutamol (SBAL), a kind of short-acting beta 2-adrenergic agonist, has been mainly used to treat bronchial asthma and other allergic airway diseases clinically. In this study, the interaction mechanism between salbutamol and human serum albumin was researched by the multispectral method and molecular docking. The fluorescence intensity of HSA could be regularly enhanced with the increase of SBAL concentration. Both the results of the multispectral method and molecular docking showed that SBAL could bind HSA with van der Waals force and hydrogen bonds. The binding mechanism was further analysed by UV-Vis and synchronous fluorescence spectra. The contents of the secondary structure of free HSA and SBAL-HSA complex were evaluated using CD spectra.


Subject(s)
Adrenergic beta-2 Receptor Agonists/chemistry , Albuterol/chemistry , Molecular Docking Simulation , Serum Albumin, Human/chemistry , Adrenergic beta-2 Receptor Agonists/therapeutic use , Albuterol/therapeutic use , Asthma/drug therapy , Humans , Receptors, Adrenergic, beta-2/chemistry , Receptors, Adrenergic, beta-2/drug effects
15.
Annu Rev Pharmacol Toxicol ; 60: 155-174, 2020 01 06.
Article in English | MEDLINE | ID: mdl-31561738

ABSTRACT

Formation of signaling complexes is crucial for the orchestration of fast, efficient, and specific signal transduction. Pharmacological disruption of defined signaling complexes has the potential for specific intervention in selected regulatory pathways without affecting organism-wide disruption of parallel pathways. Signaling by epinephrine and norepinephrine through α and ß adrenergic receptors acts on many signaling pathways in many cell types. Here, we initially provide an overview of the signaling complexes formed between the paradigmatic ß2 adrenergic receptor and two of its most important targets, the L-type Ca2+ channel CaV1.2 and the AMPA-type glutamate receptor. Importantly, both complexes contain the trimeric Gs protein, adenylyl cyclase, and the cAMP-dependent protein kinase, PKA. We then discuss the functional implications of the formation of these complexes, how those complexes can be specifically disrupted, and how such disruption could be utilized in the pharmacological treatment of disease.


Subject(s)
Calcium Channels, L-Type/metabolism , Receptors, AMPA/metabolism , Receptors, Adrenergic, beta-2/metabolism , Animals , Calcium Channels, L-Type/drug effects , Cyclic AMP-Dependent Protein Kinases/metabolism , Epinephrine/metabolism , Humans , Norepinephrine/metabolism , Receptors, AMPA/drug effects , Receptors, Adrenergic, beta-2/drug effects , Signal Transduction/drug effects
16.
Br J Pharmacol ; 177(2): 282-297, 2020 01.
Article in English | MEDLINE | ID: mdl-31506926

ABSTRACT

BACKGROUND AND PURPOSE: Chronic inflammation may play a role in the pathogenesis of Parkinson's disease (PD). Noradrenaline is an endogenous neurotransmitter with anti-inflammatory properties. In the present investigation, we assessed the immunomodulatory and neuroprotective efficacy of pharmacologically targeting the CNS noradrenergic system in a rat model of PD. EXPERIMENTAL APPROACH: The impact of treatment with the ß2 -adrenoceptor agonists clenbuterol and formoterol was assessed in the intranigral LPS rat model of PD. The immunomodulatory potential of formoterol to influence the CNS response to systemic inflammation was also assessed. KEY RESULTS: LPS-induced deficits in motor function (akinesia and forelimb-use asymmetry) and nigrostriatal dopamine loss were rescued by both agents. Treatment with the noradrenaline reuptake inhibitor atomoxetine reduced striatal dopamine loss and motor deficits following intranigral LPS injection. Co-treatment with the ß2 -adrenoceptor antagonist ICI 118,551 attenuated the protective effects of atomoxetine. Systemic LPS challenge exacerbated reactive microgliosis, IL-1ß production, dopamine cell loss in the substantia nigra, nerve terminal degeneration in the striatum, and associated motor impairments in animals that previously received intranigral LPS. This exacerbation was attenuated by formoterol treatment. CONCLUSION AND IMPLICATIONS: The results indicate that pharmacologically targeting ß2 -adrenoceptors has the propensity to regulate the neuroinflammatory phenotype in vivo and may be a potential neuroprotective strategy where inflammation contributes to the progression of dopaminergic neurodegeneration. In accordance with this, clinical agents such as ß2 -adrenoceptor agonists may prove useful as immunomodulatory agents in the treatment of neurodegenerative conditions associated with brain inflammation.


Subject(s)
Adrenergic Uptake Inhibitors/pharmacology , Adrenergic beta-2 Receptor Agonists/pharmacology , Anti-Inflammatory Agents/pharmacology , Brain/drug effects , Neurons/drug effects , Neuroprotective Agents/pharmacology , Parkinsonian Disorders/prevention & control , Receptors, Adrenergic, beta-2/drug effects , Animals , Atomoxetine Hydrochloride/pharmacology , Brain/metabolism , Brain/pathology , Brain/physiopathology , Clenbuterol/pharmacology , Disease Models, Animal , Formoterol Fumarate/pharmacology , Lipopolysaccharides , Male , Microglia/drug effects , Microglia/metabolism , Microglia/pathology , Motor Activity/drug effects , Nerve Degeneration , Neurons/metabolism , Neurons/pathology , Parkinsonian Disorders/chemically induced , Parkinsonian Disorders/metabolism , Parkinsonian Disorders/pathology , Rats, Wistar , Receptors, Adrenergic, beta-2/metabolism
17.
Respir Physiol Neurobiol ; 273: 103334, 2020 02.
Article in English | MEDLINE | ID: mdl-31689533

ABSTRACT

BACKGROUND: Hypoxia reportedly increases free radical generation in the body, causing oxidative stress and inhibiting ß2-AR signaling. The present study correlates the prophylactic potential of quercetin and salbutamol in ameliorating fluid clearing capacity of lungs by re-sensitizing ß2-AR signaling under hypoxia. METHODS: Male SD rats supplemented orally with quercetin (50 mg/Kg BW), and salbutamol (2 mg/Kg BW) were exposed to hypobaric hypoxia at 7620 m for 6 h. Western blotting and ELISA quantitated NFĸB and related genes and GPCR pathway proteins. The binding affinities of drugs with receptor were determined by SPR spectroscopy and further confirmed insilico. RESULTS: Quercetin and salbutamol pre-treatment significantly up-regulated the expressions of ß2-AR, GPR-1, GPR-10, GCSα, cAMP content, and down-regulated GRK-2, ß-arrestin, ROS, NFκB (p < 0.001), thus, enhancing alveolar fluid clearance (AFC). SPR and insilico findings revealed a higher binding affinity of ß2-AR with quercetin over salbutamol. CONCLUSION: Results indicated quercetin to be a better prophylactic that augmented AFC in rats exposed to hypoxia by attenuating inflammation and stimulating ß2-AR.


Subject(s)
Adrenergic beta-2 Receptor Agonists/pharmacology , Albuterol/pharmacology , Antioxidants/pharmacology , Cyclic AMP-Dependent Protein Kinases/drug effects , Gene Expression/drug effects , Hypoxia/metabolism , Inflammation/drug therapy , Pulmonary Alveoli/drug effects , Quercetin/pharmacology , Receptors, Adrenergic, beta-2/metabolism , Signal Transduction/drug effects , Adrenergic beta-2 Receptor Agonists/administration & dosage , Albuterol/administration & dosage , Animals , Antioxidants/administration & dosage , Body Fluids/drug effects , Disease Models, Animal , Hypoxia/drug therapy , Male , Quercetin/administration & dosage , Rats , Rats, Sprague-Dawley , Receptors, Adrenergic, beta-2/drug effects , Up-Regulation
18.
J Neuroimmunol ; 338: 577082, 2020 01 15.
Article in English | MEDLINE | ID: mdl-31707103

ABSTRACT

ß2-adrenoceptors are G-protein coupled receptors expressed on both astrocytes and microglia that play a key role in mediating the anti-inflammatory actions of noradrenaline in the CNS. Here the effect of an inflammatory stimulus (LPS + IFN-γ) was examined on glial ß2-adrenoceptor expression and function. Exposure of glia to LPS + IFN-γ decreased ß2-adrenoceptor mRNA and agonist-stimulated production of the intracellular second messenger cAMP. Pre-treatment with the synthetic glucocorticoid and potent anti-inflammatory agent dexamethasone prevented the LPS + IFN-γ-induced suppression of ß2-adrenoceptor mRNA expression. These results raise the possibility that inflammation-mediated ß2-adrenoceptor downregulation in glia may dampen the innate anti-inflammatory properties of noradrenaline in the CNS.


Subject(s)
Dexamethasone/pharmacology , Inflammation/metabolism , Neuroglia/drug effects , Receptors, Adrenergic, beta-2/drug effects , Animals , Cells, Cultured , Cyclic AMP/biosynthesis , Interferon-beta/pharmacology , Interferon-gamma/pharmacology , Lipopolysaccharides/pharmacology , Neuroglia/metabolism , Rats , Rats, Wistar , Receptors, Adrenergic, beta-2/genetics , Receptors, Adrenergic, beta-2/physiology , Tumor Necrosis Factor-alpha/biosynthesis
19.
J Am Heart Assoc ; 8(15): e012273, 2019 08 06.
Article in English | MEDLINE | ID: mdl-31311394

ABSTRACT

Background In murine heart failure models and in humans with diabetic-related heart hypertrophy, inhibition of phosphodiesterase 5 (PDE5) by sildenafil improves cardiac outcomes. However, the mechanism by which sildenafil improves cardiac function is unclear. We have observed a relationship between PDE5 and ß2 adrenergic receptor (ß2AR), which is characterized here as a novel mechanistic axis by which sildenafil improves symptoms of diabetic cardiomyopathy. Methods and Results Wild-type and ß2AR knockout mice fed a high fat diet (HFD) were treated with sildenafil, and echocardiogram analysis was performed. Cardiomyocytes were isolated for excitation-contraction (E-C) coupling, fluorescence resonant energy transfer, and proximity ligation assays; while heart tissues were implemented for biochemical and histological analyses. PDE5 selectively associates with ß2AR, but not ß1 adrenergic receptor, and inhibition of PDE5 with sildenafil restores the impaired response to adrenergic stimulation in HFD mice and isolated ventriculomyocytes. Sildenafil enhances ß adrenergic receptor (ßAR)-stimulated cGMP and cAMP signals in HFD myocytes. Consequently, inhibition of PDE5 leads to protein kinase G-, and to a lesser extent, calcium/calmodulin-dependent kinase II-dependent improvements in adrenergically stimulated E-C coupling. Deletion of ß2AR abolishes sildenafil's effect. Although the PDE5-ß2AR association is not altered in HFD, phosphodiesterase 3 displays an increased association with the ß2AR-PDE5 complex in HFD myocytes. Conclusions This study elucidates mechanisms by which the ß2AR-PDE5 axis can be targeted for treating diabetic cardiomyopathy. Inhibition of PDE5 enhances ß2AR stimulation of cGMP and cAMP signals, as well as protein kinase G-dependent E-C coupling in HFD myocytes.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 5/physiology , Diabetes Mellitus, Type 2/physiopathology , Diabetic Cardiomyopathies/drug therapy , Diabetic Cardiomyopathies/physiopathology , Heart/physiopathology , Phosphodiesterase 5 Inhibitors/pharmacology , Phosphodiesterase 5 Inhibitors/therapeutic use , Receptors, Adrenergic, beta-2/drug effects , Receptors, Adrenergic, beta-2/physiology , Sildenafil Citrate/pharmacology , Sildenafil Citrate/therapeutic use , Animals , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
20.
Am J Physiol Cell Physiol ; 317(4): C674-C686, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31268780

ABSTRACT

G protein-coupled receptor kinase 2 (GRK2) is an important protein involved in ß-adrenergic receptor desensitization. In addition, studies have shown GRK2 can modulate different metabolic processes in the cell. For instance, GRK2 has been recently shown to promote mitochondrial biogenesis and increase ATP production. However, the role of GRK2 in skeletal muscle and the signaling mechanisms that regulate GRK2 remain poorly understood. Myostatin is a well-known myokine that has been shown to impair mitochondria function. Here, we have assessed the role of myostatin in regulating GRK2 and the subsequent downstream effect of myostatin regulation of GRK2 on mitochondrial respiration in skeletal muscle. Myostatin treatment promoted the loss of GRK2 protein in myoblasts and myotubes in a time- and dose-dependent manner, which we suggest was through enhanced ubiquitin-mediated protein loss, as treatment with proteasome inhibitors partially rescued myostatin-mediated loss of GRK2 protein. To evaluate the effects of GRK2 on mitochondrial respiration, we generated stable myoblast lines that overexpress GRK2. Stable overexpression of GRK2 resulted in increased mitochondrial content and enhanced mitochondrial/oxidative respiration. Interestingly, although overexpression of GRK2 was unable to prevent myostatin-mediated impairment of mitochondrial respiratory function, elevated levels of GRK2 blocked the increased autophagic flux observed following treatment with myostatin. Overall, our data suggest a novel role for GRK2 in regulating mitochondria mass and mitochondrial respiration in skeletal muscle.


Subject(s)
Autophagy/drug effects , G-Protein-Coupled Receptor Kinase 2/drug effects , Mitochondria/drug effects , Myoblasts/drug effects , Myostatin/pharmacology , Animals , G-Protein-Coupled Receptor Kinase 2/metabolism , Mice , Mitochondria/metabolism , Muscle Cells/metabolism , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Myoblasts/metabolism , Myostatin/metabolism , Receptors, Adrenergic, beta/drug effects , Receptors, Adrenergic, beta/metabolism , Receptors, Adrenergic, beta-2/drug effects , Receptors, Adrenergic, beta-2/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...