Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 13.997
Filter
1.
Respir Res ; 25(1): 347, 2024 Sep 28.
Article in English | MEDLINE | ID: mdl-39342317

ABSTRACT

BACKGROUND: Obstructive sleep apnea (OSA), characterized by chronic intermittent hypoxia (CIH), is a prevalent condition that has been associated with various forms of cancer. Although some clinical studies suggest a potential link between OSA and lung cancer, this association remains uncertain, and the underlying mechanisms are not fully understood. This study investigated the role of the catecholamine-ß-adrenergic receptor (ßAR) and the NLRP3 inflammasome in mediating the effects of CIH on lung cancer progression in mice. METHODS: Male C57BL/6 N mice were subjected to CIH for four weeks, with Lewis lung carcinoma cells seeded subcutaneously. Propranolol (a ßAR blocker) or nepicastat (an inhibitor of catecholamine production) was administered during this period. Tumor volume and tail artery blood pressure were monitored. Immunohistochemical staining and immunofluorescence staining were employed to assess protein expression of Ki-67, CD31, VEGFR2, PD-1, PD-L1, and ASC specks in tumor tissues. ELISA was used to detect catecholamine and various cytokines, while western blot assessed the expression of cyclin D1, caspase-1, and IL-1ß. In vitro tube formation assay investigated angiogenesis. NLRP3 knockout mice were used to determine the mechanism of NLRP3 in CIH. RESULTS: CIH led to an increase in catecholamine. Catecholamine-ßAR inhibitor drugs prevented the increase in blood pressure caused by CIH. Notably, the drugs inhibited CIH-induced murine lung tumor growth, and the expression of Ki-67, cyclin D1, CD31, VEGFR2, PD-1 and PD-L1 in tumor decreased. In vitro, propranolol inhibits tube formation induced by CIH mouse serum. Moreover, CIH led to an increase in TNF-α, IL-6, IL-1ß, IFN-γ and sPD-L1 levels and a decrease in IL-10 in peripheral blood, accompanied by activation of NLRP3 inflammasomes in tumor, but these effects were also stopped by drugs. In NLRP3-knockout mice, CIH-induced upregulation of PD-1/PD-L1 in tumor was inhibited. CONCLUSIONS: Our study underscores the significant contribution of ß-adrenergic signaling and the NLRP3 inflammasome to CIH-induced lung cancer progression. These pathways represent potential therapeutic targets for mitigating the impact of OSA on lung cancer.


Subject(s)
Disease Progression , Hypoxia , Inflammasomes , Lung Neoplasms , Mice, Inbred C57BL , NLR Family, Pyrin Domain-Containing 3 Protein , Receptors, Adrenergic, beta , Signal Transduction , Animals , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Male , Mice , Inflammasomes/metabolism , Signal Transduction/physiology , Hypoxia/metabolism , Receptors, Adrenergic, beta/metabolism , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Carcinoma, Lewis Lung/metabolism , Carcinoma, Lewis Lung/pathology , Adrenergic beta-Antagonists/pharmacology , Chronic Disease , Propranolol/pharmacology , Mice, Knockout , Indenes , Furans , Sulfonamides
2.
Proc Natl Acad Sci U S A ; 121(40): e2404243121, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39331410

ABSTRACT

Gprotein-coupled receptors (GPCRs) regulate several physiological and pathological processes and represent the target of approximately 30% of Food and Drug Administration-approved drugs. GPCR-mediated signaling was thought to occur exclusively at the plasma membrane. However, recent studies have unveiled their presence and function at subcellular membrane compartments. There is a growing interest in studying compartmentalized signaling of GPCRs. This requires development of tools to separate GPCR signaling at the plasma membrane from the ones initiated at intracellular compartments. We leveraged the structural and pharmacological information available for ß-adrenergic receptors (ßARs) and focused on ß1AR as exemplary GPCR that functions at subcellular compartments, and rationally designed spatially restricted antagonists. We generated a cell-impermeable ßAR antagonist by conjugating a suitable pharmacophore to a sulfonate-containing fluorophore. This cell-impermeable antagonist only inhibited ß1AR on the plasma membrane. In contrast, a cell-permeable ßAR antagonist containing a nonsulfonated fluorophore efficiently inhibited both the plasma membrane and Golgi pools of ß1ARs. Furthermore, the cell-impermeable antagonist selectively inhibited the phosphorylation of PKA downstream effectors near the plasma membrane, which regulate sarcoplasmic reticulum (SR) Ca2+ release in adult cardiomyocytes, while the ß1AR Golgi pool remained active. Our tools offer promising avenues for investigating compartmentalized ßAR signaling in various contexts, potentially advancing our understanding of ßAR-mediated cellular responses in health and disease. They also offer a general strategy to study compartmentalized signaling for other GPCRs in various biological systems.


Subject(s)
Cell Membrane , Receptors, Adrenergic, beta-1 , Humans , Animals , Cell Membrane/metabolism , Cell Membrane/drug effects , Receptors, Adrenergic, beta-1/metabolism , Signal Transduction/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/drug effects , HEK293 Cells , Adrenergic beta-Antagonists/pharmacology , Receptors, Adrenergic, beta/metabolism , Calcium/metabolism , Golgi Apparatus/metabolism , Golgi Apparatus/drug effects , Rats
3.
Hypertension ; 81(10): 2027-2037, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39136127

ABSTRACT

More than 1.5 billion people worldwide have arterial hypertension. Hypertension increases the risks of death and cardiovascular disease, such as atrial fibrillation and heart failure. The autonomic nervous system plays an essential role in hypertension development and disease progression. While lifestyle factors, such as obesity and obstructive sleep apnea, predispose to hypertension by increasing sympathetic activity, hypertension itself maintains the autonomic nervous imbalance, providing the substrate for atrial fibrillation and heart failure. Therefore, autonomic nervous system modulation either by direct targeting or indirect treatment of comorbidities has the potential to treat both hypertension and related atrial and ventricular end-organ damage. We discuss interventions for the modulation of the autonomic nervous system for hypertension and related cardiac end-organ damage, including pharmacological adrenergic beta-receptor blockade, renal denervation, carotid baroreceptor stimulation, low-level vagal stimulation, and ablation of ganglionated plexuses. In summary, the literature suggests that targeting the autonomic nervous system potentially represents a therapeutic approach to prevent atrial and ventricular end-organ damage in patients with hypertension. However, clinical trials specifically designed to test the effect of autonomic modulation on hypertension-mediated cardiac end-organ damage are scarce.


Subject(s)
Autonomic Nervous System , Hypertension , Humans , Hypertension/physiopathology , Hypertension/therapy , Autonomic Nervous System/physiopathology , Adrenergic beta-Antagonists/therapeutic use , Adrenergic beta-Antagonists/pharmacology , Heart Failure/physiopathology , Heart Failure/therapy
4.
J Mol Med (Berl) ; 102(10): 1245-1254, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39145814

ABSTRACT

Severe burn injuries are defined by a prolonged hypermetabolic response characterized by increases in resting energy expenditure, systemic catabolism, and multi-organ dysfunction. The sustained elevation of catecholamines following a burn injury is thought to significantly contribute to this hypermetabolic response, leading to changes in adipose tissue such as increased lipolysis and the browning of subcutaneous white adipose tissue (WAT). Failure to mitigate these adverse changes within the adipose tissue has been shown to exacerbate the post-burn hypermetabolic response and lead to negative outcomes. Propranolol, a non-selective ß-blocker, has been clinically administered to improve outcomes of pediatric and adult burn patients, but there is inadequate knowledge of its effects on the distinct adipose tissue depots. In this study, we investigated the adipose depot-specific alterations that occur in response to burn injury. Moreover, we explored the therapeutic effects of ß-adrenoceptor blockade via the drug propranolol in attenuating these burn-induced pathophysiological changes within the different fat depots. Using a murine model of thermal injury, we show that burn injury induces endoplasmic reticulum (ER) stress in the epididymal (eWAT) but not in the inguinal (iWAT) WAT depot. Conversely, burn injury induces the activation of key lipolytic pathways in both eWAT and iWAT depots. Treatment of burn mice with propranolol effectively mitigated adverse burn-induced alterations in the adipose by alleviating ER stress in the eWAT and reducing lipolysis in both depots. Furthermore, propranolol treatment in post-burn mice attenuated UCP1-mediated subcutaneous WAT browning following injury. Overall, our findings suggest that propranolol serves as an effective therapeutic intervention to mitigate the adverse changes induced by burn injury, including ER stress, lipotoxicity, and WAT browning, in both adipose tissue depots. KEY MESSAGES: Burn injury adversely affects adipose tissue metabolism via distinct changes in both visceral and subcutaneous adipose depots. Propranolol, a non-selective ß-adrenergic blocker, attenuates many of the adverse adipose tissue changes mediated by burn injury.


Subject(s)
Adipose Tissue , Adrenergic beta-Antagonists , Burns , Endoplasmic Reticulum Stress , Propranolol , Animals , Burns/drug therapy , Burns/metabolism , Adrenergic beta-Antagonists/pharmacology , Adrenergic beta-Antagonists/therapeutic use , Male , Mice , Endoplasmic Reticulum Stress/drug effects , Propranolol/pharmacology , Propranolol/therapeutic use , Adipose Tissue/metabolism , Adipose Tissue/drug effects , Lipolysis/drug effects , Mice, Inbred C57BL , Adipose Tissue, White/metabolism , Adipose Tissue, White/drug effects , Disease Models, Animal , Energy Metabolism/drug effects
5.
Int J Mol Sci ; 25(15)2024 Jul 24.
Article in English | MEDLINE | ID: mdl-39125627

ABSTRACT

The autonomic nervous system plays a key role in maintaining body hemostasis through both the sympathetic and parasympathetic nervous systems. Sympathetic overstimulation as a reflex to multiple pathologies, such as septic shock, brain injury, cardiogenic shock, and cardiac arrest, could be harmful and lead to autonomic and immunologic dysfunction. The continuous stimulation of the beta receptors on immune cells has an inhibitory effect on these cells and may lead to immunologic dysfunction through enhancing the production of anti-inflammatory cytokines, such as interleukin-10 (IL-10), and inhibiting the production of pro-inflammatory factors, such as interleukin-1B IL-1B and tissue necrotizing factor-alpha (TNF-alpha). Sympathetic overstimulation-induced autonomic dysfunction may also happen due to adrenergic receptor insensitivity or downregulation. Administering anti-adrenergic medication, such as beta-blockers, is a promising treatment to compensate against the undesired effects of adrenergic surge. Despite many misconceptions about beta-blockers, beta-blockers have shown a promising effect in decreasing mortality in patients with critical illness. In this review, we summarize the recently published articles that have discussed using beta-blockers as a promising treatment to decrease mortality in critically ill patients, such as patients with septic shock, traumatic brain injury, cardiogenic shock, acute decompensated heart failure, and electrical storm. We also discuss the potential pathophysiology of beta-blockers in various types of critical illness. More clinical trials are encouraged to evaluate the safety and effectiveness of beta-blockers in improving mortality among critically ill patients.


Subject(s)
Adrenergic beta-Antagonists , Autonomic Nervous System , Critical Illness , Humans , Adrenergic beta-Antagonists/therapeutic use , Adrenergic beta-Antagonists/pharmacology , Autonomic Nervous System/drug effects , Animals , Shock, Septic/drug therapy , Shock, Septic/immunology
6.
J Hazard Mater ; 477: 135403, 2024 Sep 15.
Article in English | MEDLINE | ID: mdl-39096644

ABSTRACT

Drug resistance poses a high risk to human health. Extensive use of non-antibiotic drugs contributes to antibiotic resistance genes (ARGs) transfer. However, how they affect the spread of broad-host plasmids in complex biological systems remains unknown. This study investigated the effect of metoprolol on the transfer frequency and host range of ARGs in both intrageneric and intergeneric pure culture systems, as well as in anammox microbiome. The results showed that environmental concentrations of metoprolol significantly promoted the intrageneric and intergeneric conjugative transfer. Initially, metoprolol induced excessive oxidative stress, resulting in high cell membrane permeability and bacterial SOS response. Meanwhile, more pili formation increased the adhesion and contact between bacteria, and the abundance of conjugation-related genes also increased significantly. Activation of the electron transport chain provided more ATP for this energy-consuming process. The underlying mechanism was further verified in the complex anammox conjugative system. Metoprolol induced the enrichment of ARGs and mobile genetic elements. The enhanced bacterial interaction and energy generation facilitated the high conjugative transfer frequency of ARGs. In addition, plasmid-borne ARGs tended to transfer to opportunistic pathogens. This work raises public concerns about the health and ecological risks of non-antibiotic drugs.


Subject(s)
Conjugation, Genetic , Metoprolol , Plasmids , Plasmids/genetics , Conjugation, Genetic/drug effects , Drug Resistance, Multiple, Bacterial/genetics , Drug Resistance, Multiple, Bacterial/drug effects , Adrenergic beta-Antagonists/pharmacology , Gene Transfer, Horizontal , Bacteria/genetics , Bacteria/drug effects , Bacteria/metabolism , Anti-Bacterial Agents/pharmacology , Genes, MDR/genetics , Microbiota/drug effects
7.
J Clin Psychopharmacol ; 44(5): 462-467, 2024.
Article in English | MEDLINE | ID: mdl-39174017

ABSTRACT

BACKGROUND: Despite the use of behavioral interventions and psychotropic medications, many individuals with autism spectrum disorder (ASD) who engage in severe aggression remain refractory to conventional treatment. Propranolol, a beta-blocker, has accumulated much anecdotal evidence as a promising option. However, well-designed studies are rare, and the apprehension about cardiovascular side effects from large doses continues to exist. PURPOSE: The aims of this study were (1) to demonstrate the feasibility of treating aggression with high-dose propranolol using telehealth study visits and (2) to document cardiac safety. METHODS: This study utilized a randomized, double-blind, placebo-controlled, crossover design. Dosing was titrated up in a flexible but stepwise fashion until therapeutic response was obtained or up to 200 mg tid. Following washout, those who were assigned propranolol were crossed over to placebo and vice versa. Six participants between the ages 12-19 participated. The primary outcome measures were the final Clinical Global Impression Improvement Scale (CGI-I) and the Aberrant Behavior Checklist-Community Irritability (ABC-C/I) scores at 200 mg tid. RESULTS: The CGI-I indicated a 50% reduction in symptoms in the propranolol phase, while the ABC-I indicated a 37% reduction in comparison to placebo. The effect sizes ( r ) for the CGI-I and the ABC-C/I were large, -0.74 and -0.64, respectively. The average blood pressure was 122/68 during the placebo phase and 109/72 during the propranolol phase. All Holter monitor exams were unremarkable. CONCLUSION: These results suggest that propranolol is an effective option in decreasing aggression in individuals with ASD. As this was a small study, a larger clinical trial is needed.


Subject(s)
Adrenergic beta-Antagonists , Aggression , Autism Spectrum Disorder , Cross-Over Studies , Propranolol , Humans , Autism Spectrum Disorder/drug therapy , Double-Blind Method , Propranolol/administration & dosage , Propranolol/adverse effects , Aggression/drug effects , Male , Adolescent , Child , Adrenergic beta-Antagonists/administration & dosage , Adrenergic beta-Antagonists/adverse effects , Adrenergic beta-Antagonists/pharmacology , Pilot Projects , Young Adult , Female , Treatment Outcome , Adult
8.
J Pharmacol Sci ; 156(2): 134-141, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39179332

ABSTRACT

Risperidone is a second-generation antipsychotic for treating schizophrenia and bipolar disorder. It can potently inhibit IKr, but is classified into conditional risk for torsade de pointes (TdP) by CredibleMeds®. Our previous studies using chronic atrioventricular block dogs showed that risperidone alone did not induce TdP, and that dl-sotalol (ß-adrenoceptor blockade plus IKr inhibition) induced TdP three times more frequently than d-sotalol (IKr inhibition alone). Since risperidone can block α1-adrenoceptor and decrease blood pressure, the resulting reflex-mediated increase of sympathetic tone on ß-adrenoceptor might protect the heart from its IKr inhibition-associated TdP. To validate this hypothesis, risperidone was administered to chronic atrioventricular block dogs after ß-adrenoceptor blocker atenolol infusion with monitoring J-Tpeak and Tpeak-Tend, which are proarrhythmic surrogate markers of "substrate" and "trigger" toward TdP, respectively. Atenolol alone induced TdP in 1 out of 5 dogs; moreover, an additional infusion of risperidone induced TdP in 3 out of 4 dogs. Risperidone prolonged QT interval, J-Tpeak and Tpeak-Tend in animals that induced TdP. These findings indicate that ß-adrenoceptor blockade can diminish repolarization reserve to augment risperidone's torsadogenic potential, thus advising caution when using ß-adrenoceptor blockers in patients with IKr inhibition-linked labile repolarization.


Subject(s)
Adrenergic beta-Antagonists , Antipsychotic Agents , Atenolol , Risperidone , Torsades de Pointes , Risperidone/pharmacology , Animals , Dogs , Atenolol/pharmacology , Antipsychotic Agents/pharmacology , Antipsychotic Agents/adverse effects , Adrenergic beta-Antagonists/pharmacology , Torsades de Pointes/chemically induced , Atrioventricular Block/chemically induced , Male
9.
Am J Physiol Endocrinol Metab ; 327(3): E290-E301, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-38984949

ABSTRACT

Type 1 diabetes recipients of intrahepatic islet transplantation exhibit glucose-dependent suppression of insulin and activation of glucagon secretion in response to insulin-induced hypoglycemia associated with clinical protection from hypoglycemia. Whether sympathetic activation of adrenergic receptors on transplanted islets is required for these responses in defense against hypoglycemia is not known. To evaluate the adrenergic contribution to posttransplant glucose counterregulation, we performed a randomized, double-blind crossover study of responses during a hyperinsulinemic euglycemic-hypoglycemic clamp under phentolamine (α-adrenergic blockage), propranolol (ß-adrenergic blockage), or placebo infusion. Characteristics of participants (5 females/4 males) were as follows: median (range) age 53 (34-63) yr, diabetes duration 29 (18-56) yr, posttransplant 7.0 (1.9-8.4) yr, HbA1c 5.8 (4.5-6.8)%, insulin in-/dependent 5/4, all on tacrolimus-based immunosuppression. During the clamp, blood pressure was lower with phentolamine and heart rate was lower with propranolol versus placebo (P < 0.05). There was no difference in the suppression of endogenous insulin secretion (derived from C-peptide measurements) during the euglycemic or hypoglycemic phases, and although levels of glucagon were similar with phentolamine or propranolol vs. placebo, the increase in glucagon from eu- to hypoglycemia was greater with propranolol vs. placebo (P < 0.05). Pancreatic polypeptide was greater with phentolamine versus placebo during the euglycemic phase (P < 0.05), and free fatty acids were lower and the glucose infusion rate was higher with propranolol versus placebo during the hypoglycemic phase (P < 0.05 for both). These results indicate that neither physiological α- nor ß-adrenergic blockade attenuates transplanted islet responses to hypoglycemia, suggesting sympathetic reinnervation of the islet graft is not necessary for posttransplant glucose counterregulation.NEW & NOTEWORTHY Whether adrenergic input to islets is necessary for glucose homeostasis in humans is debated. Here, the adrenergic contribution to intrahepatically transplanted islet cell responses to hypoglycemia in individuals with type 1 diabetes was investigated through α- or ß-adrenergic receptor blockade during hyperinsulinemic euglycemic-hypoglycemic clamps. Neither α- nor ß-adrenergic blockage affected the suppression of endogenous insulin or activation of glucagon secretion, suggesting that sympathetic reinnervation of islet grafts is not required for posttransplant defense against hypoglycemia.


Subject(s)
Adrenergic beta-Antagonists , Cross-Over Studies , Diabetes Mellitus, Type 1 , Glucose Clamp Technique , Hypoglycemia , Islets of Langerhans Transplantation , Phentolamine , Propranolol , Humans , Female , Male , Diabetes Mellitus, Type 1/metabolism , Middle Aged , Adult , Islets of Langerhans Transplantation/adverse effects , Hypoglycemia/chemically induced , Hypoglycemia/metabolism , Double-Blind Method , Adrenergic beta-Antagonists/pharmacology , Phentolamine/pharmacology , Propranolol/pharmacology , Blood Glucose/metabolism , Blood Glucose/drug effects , Adrenergic alpha-Antagonists/pharmacology , Insulin/metabolism , Glucagon/metabolism , Glucagon/blood , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism
11.
J Comp Physiol B ; 194(4): 519-535, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38972930

ABSTRACT

Ultradian rhythms of metabolism, body temperature and activity are attenuated or disappear completely during torpor in Djungarian hamsters, for all three ultradian periodicities (URsmall, URmedium and URlarge). URsmall and URmedium disappear during entrance into torpor, whereas URlarge disappear later or continue with a low amplitude. This suggests a tight functional link between torpor and the expression of ultradian rhythms, i.e. torpor is achieved by suppression of metabolic rate as well as silencing of ultradian rhythms. Spontaneous torpor is often initiated after an ultradian burst of activity and metabolic rate, beginning with a period of motionless rest and accompanied by a decrease of metabolic rate and body temperature. To extend previous findings on the potential role of the adrenergic system on torpor induction we analysed the influence of the ß3-adrenergic agonist Mirabegron on torpor in Djungarian hamsters, as compared to the influence of the ß-adrenergic antagonist Propranolol. Hamsters were implanted with 10 day release pellets of Mirabegron (0.06 mg day-1) or Propranolol (0.3 mg day-1). Mirabegron transiently supressed and accelerated ultradian rhythms but had no effect on torpor behaviour. Propranolol did not affect torpor behaviour nor the expression of ultradian rhythms with the dosage applied during this study.


Subject(s)
Phodopus , Torpor , Ultradian Rhythm , Animals , Torpor/physiology , Phodopus/physiology , Cricetinae , Male , Ultradian Rhythm/physiology , Propranolol/pharmacology , Adrenergic beta-Antagonists/pharmacology , Body Temperature , Motor Activity , Basal Metabolism
12.
Int Immunopharmacol ; 139: 112670, 2024 Sep 30.
Article in English | MEDLINE | ID: mdl-39018694

ABSTRACT

Acute Respiratory Distress Syndrome (ARDS) manifests as an acute inflammatory lung injury characterized by persistent hypoxemia, featuring a swift onset, high mortality, and predominantly supportive care as the current therapeutic approach, while effective treatments remain an area of active investigation. Adrenergic receptors (AR) play a pivotal role as stress hormone receptors, extensively participating in various inflammatory processes by initiating downstream signaling pathways. Advancements in molecular biology and pharmacology continually unveil the physiological significance of distinct AR subtypes. Interventions targeting these subtypes have the potential to induce specific alterations in cellular and organismal functions, presenting a promising avenue as a therapeutic target for managing ARDS. This article elucidates the pathogenesis of ARDS and the basic structure and function of AR. It also explores the relationship between AR and ARDS from the perspective of different AR subtypes, aiming to provide new insights for the improvement of ARDS.


Subject(s)
Adrenergic alpha-2 Receptor Agonists , Adrenergic beta-Antagonists , Respiratory Distress Syndrome , Sympathetic Nervous System , Humans , Respiratory Distress Syndrome/drug therapy , Animals , Adrenergic beta-Antagonists/therapeutic use , Adrenergic beta-Antagonists/pharmacology , Adrenergic alpha-2 Receptor Agonists/therapeutic use , Adrenergic alpha-2 Receptor Agonists/pharmacology , Sympathetic Nervous System/drug effects , Sympathetic Nervous System/physiopathology , Receptors, Adrenergic/metabolism , Signal Transduction/drug effects
13.
Nat Med ; 30(7): 2030-2036, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39009776

ABSTRACT

Consumer-grade wearable technology has the potential to support clinical research and patient management. Here, we report results from the RATE-AF trial wearables study, which was designed to compare heart rate in older, multimorbid patients with permanent atrial fibrillation and heart failure who were randomized to treatment with either digoxin or beta-blockers. Heart rate (n = 143,379,796) and physical activity (n = 23,704,307) intervals were obtained from 53 participants (mean age 75.6 years (s.d. 8.4), 40% women) using a wrist-worn wearable linked to a smartphone for 20 weeks. Heart rates in participants treated with digoxin versus beta-blockers were not significantly different (regression coefficient 1.22 (95% confidence interval (CI) -2.82 to 5.27; P = 0.55); adjusted 0.66 (95% CI -3.45 to 4.77; P = 0.75)). No difference in heart rate was observed between the two groups of patients after accounting for physical activity (P = 0.74) or patients with high activity levels (≥30,000 steps per week; P = 0.97). Using a convolutional neural network designed to account for missing data, we found that wearable device data could predict New York Heart Association functional class 5 months after baseline assessment similarly to standard clinical measures of electrocardiographic heart rate and 6-minute walk test (F1 score 0.56 (95% CI 0.41 to 0.70) versus 0.55 (95% CI 0.41 to 0.68); P = 0.88 for comparison). The results of this study indicate that digoxin and beta-blockers have equivalent effects on heart rate in atrial fibrillation at rest and on exertion, and suggest that dynamic monitoring of individuals with arrhythmia using wearable technology could be an alternative to in-person assessment. ClinicalTrials.gov identifier: NCT02391337 .


Subject(s)
Adrenergic beta-Antagonists , Atrial Fibrillation , Digoxin , Heart Rate , Wearable Electronic Devices , Humans , Digoxin/therapeutic use , Digoxin/pharmacology , Heart Rate/drug effects , Female , Male , Aged , Atrial Fibrillation/drug therapy , Atrial Fibrillation/physiopathology , Adrenergic beta-Antagonists/therapeutic use , Adrenergic beta-Antagonists/pharmacology , Aged, 80 and over , Heart Failure/drug therapy , Heart Failure/physiopathology , Exercise , Smartphone
14.
FASEB J ; 38(13): e23813, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38976162

ABSTRACT

Beta-blockers are commonly used medications that antagonize ß-adrenoceptors, reducing sympathetic nervous system activity. Emerging evidence suggests that beta-blockers may also have anticancer effects and help overcome drug resistance in cancer treatment. This review summarizes the contribution of different isoforms of beta-adrenoceptors in cancer progression, the current preclinical and clinical data on associations between beta-blockers use and cancer outcomes, as well as their ability to enhance responses to chemotherapy and other standard therapies. We discuss proposed mechanisms, including effects on angiogenesis, metastasis, cancer stem cells, and apoptotic pathways. Overall, results from epidemiological studies and small clinical trials largely indicate the beneficial effects of beta-blockers on cancer progression and drug resistance. However, larger randomized controlled trials are needed to firmly establish their clinical efficacy and optimal utilization as adjuvant agents in cancer therapy.


Subject(s)
Adrenergic beta-Antagonists , Drug Resistance, Neoplasm , Neoplasms , Humans , Adrenergic beta-Antagonists/therapeutic use , Adrenergic beta-Antagonists/pharmacology , Neoplasms/drug therapy , Neoplasms/pathology , Animals , Cardiovascular Diseases/drug therapy , Disease Progression , Receptors, Adrenergic, beta/metabolism , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/pharmacology
15.
J Cardiovasc Pharmacol ; 84(1): 110-117, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38922579

ABSTRACT

ABSTRACT: Hypercatecholaminergic conditions are known to cause heart failure and cardiac fibrosis when severe. Although previous investigations have studied the effects of beta-blockade in experimental models of catecholaminergic states, the detailed benefits of beta-blockade in more realistic models of hyper-adrenergic states were less clear. In this study, we examined acute cardiac changes in rats with hyperacute catecholamine-induced heart failure with and without propranolol treatment. Male Sprague-Dawley rats (n = 12) underwent a 6-hour infusion of epinephrine and norepinephrine alone, with an additional propranolol bolus (1 mg/kg) at hour 1 (n = 6). Cardiac tissues were examined after 6 hours. Cardiac immunohistochemistry revealed significantly decreased expression of phosphorylated p-38 (left ventricle, P = 0.021; right ventricle, P = 0.021), with upregulation of reactive oxidative species and other profibrosis proteins, after catecholamine infusion alone. After 1 propranolol 1 mg/kg bolus, the levels of phosphorylated-p38 returned to levels comparable with sham (left ventricle, P = 0.021; right ventricle, P = 0.043), with additional findings including downregulation of the apoptotic pathway and profibrotic proteins. We conclude that catecholamine-induced heart failure exerts damage through the p-38 mitogen-activated protein kinase pathway and demonstrates profibrotic changes mediated by matrix metalloproteinase 9, alpha-smooth muscle actin, and fibroblast growth factor 23. Changes in these pathways attenuated acute catecholamine-induced heart failure after propranolol bolus 1 mg/kg. We conclude that propranolol bolus at 1 mg/kg is able to mediate the effects of catecholamine excess through the p-38 mitogen-activated protein kinase pathway, profibrosis, and extrinsic apoptosis pathway.


Subject(s)
Adrenergic beta-Antagonists , Fibrosis , Heart Failure , Norepinephrine , Propranolol , Rats, Sprague-Dawley , p38 Mitogen-Activated Protein Kinases , Animals , Male , Propranolol/pharmacology , p38 Mitogen-Activated Protein Kinases/metabolism , Rats , Adrenergic beta-Antagonists/pharmacology , Adrenergic beta-Antagonists/administration & dosage , Heart Failure/drug therapy , Heart Failure/metabolism , Heart Failure/pathology , Heart Failure/physiopathology , Heart Failure/chemically induced , Norepinephrine/metabolism , Epinephrine/toxicity , Epinephrine/administration & dosage , Phosphorylation , Apoptosis/drug effects , Disease Models, Animal , Myocardium/pathology , Myocardium/metabolism , Myocardium/enzymology , Catecholamines/metabolism , Reactive Oxygen Species/metabolism
16.
Biochem Pharmacol ; 226: 116396, 2024 08.
Article in English | MEDLINE | ID: mdl-38942089

ABSTRACT

This study introduces (S)-Opto-prop-2, a second-generation photoswitchable ligand designed for precise modulation of ß2-adrenoceptor (ß2AR). Synthesised by incorporating an azobenzene moiety with propranolol, (S)-Opto-prop-2 exhibited a high PSScis (photostationary state for cis isomer) percentage (∼90 %) and a favourable half-life (>10 days), facilitating diverse bioassay measurements. In vitro, the cis-isomer displayed substantially higher ß2AR binding affinity than the trans-isomer (1000-fold), making (S)-Opto-prop-2 one of the best photoswitchable GPCR (G protein-coupled receptor) ligands reported so far. Molecular docking of (S)-Opto-prop-2 in the X-ray structure of propranolol-bound ß2AR followed by site-directed mutagenesis studies, identified D1133.32, N3127.39 and F2896.51 as crucial residues that contribute to ligand-receptor interactions at the molecular level. In vivo efficacy was assessed using a rabbit ocular hypertension model, revealing that the cis isomer mimicked propranolol's effects in reducing intraocular pressure, while the trans isomer was inactive. Dynamic optical modulation of ß2AR by (S)-Opto-prop-2 was demonstrated in two different cAMP bioassays and using live-cell confocal imaging, indicating reversible and dynamic control of ß2AR activity using the new photopharmacology tool. In conclusion, (S)-Opto-prop-2 emerges as a promising photoswitchable ligand for precise and reversible ß2AR modulation with light. The new tool shows superior cis-on binding affinity, one of the largest reported differences in affinity (1000-fold) between its two configurations, in vivo efficacy, and dynamic modulation. This study contributes valuable insights into the evolving field of photopharmacology, offering a potential avenue for targeted therapy in ß2AR-associated pathologies.


Subject(s)
Adrenergic beta-2 Receptor Antagonists , Receptors, Adrenergic, beta-2 , Animals , Humans , Male , Rabbits , Adrenergic beta-2 Receptor Antagonists/pharmacology , Adrenergic beta-2 Receptor Antagonists/chemistry , Adrenergic beta-Antagonists/pharmacology , Adrenergic beta-Antagonists/chemistry , Azo Compounds/chemistry , Azo Compounds/pharmacology , CHO Cells , Cricetulus , HEK293 Cells , Molecular Docking Simulation/methods , Photochemical Processes , Propranolol/pharmacology , Propranolol/chemistry , Receptors, Adrenergic, beta-2/metabolism , Receptors, Adrenergic, beta-2/chemistry
17.
Int Immunopharmacol ; 138: 112568, 2024 Sep 10.
Article in English | MEDLINE | ID: mdl-38936055

ABSTRACT

Chronic stress negatively affects the immune system and promotes tumor progression. Tumor-associated macrophage (TAM) is an important component of the tumor immune microenvironment. However, the influence of chronic stress on M1-M2 polarization of TAM is unclear. We used flow cytometry to measure the M1-M2 polarization of TAM in chronic stress hepatocellular carcinoma (HCC) bearing mice. We also measured the level of norepinephrine and blocked ß-adrenergic signaling to explore the role of ß-adrenergic receptor in the effect of chronic stress on M1-M2 polarization of TAM. We found that chronic stress disrupts the M1-M2 polarization in tumor tissues, increased the level of CD11b+Ly6C+CCR2+ monocyte and interleukin-1beta in blood and promoted the growth of HCC. Furthermore, chronic stress upregulated the level of CCL2 in tumor tissues. Finally, we found chronic stress increased norepinephrine level in serum and propranolol, a blocker of ß-adrenergic signaling, inhibited HCC growth, recovered the M1-M2 polarization balance of TAM in tumor tissues, blocked the increase of CD11b+Ly6C+CCR2+ monocytes in blood, and blocked the increase of CCL2 in tumor tissues induced by chronic stress. Our study indicated that chronic stress disrupts the M1-M2 polarization balance of TAMs through ß-adrenergic signaling, thereby promoting the growth of HCC.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Norepinephrine , Receptors, Adrenergic, beta , Signal Transduction , Animals , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Mice , Liver Neoplasms/immunology , Liver Neoplasms/metabolism , Norepinephrine/metabolism , Norepinephrine/blood , Male , Receptors, Adrenergic, beta/metabolism , Propranolol/pharmacology , Mice, Inbred C57BL , Tumor-Associated Macrophages/immunology , Tumor-Associated Macrophages/metabolism , Cell Line, Tumor , Interleukin-1beta/metabolism , Chemokine CCL2/metabolism , Tumor Microenvironment/immunology , Humans , Adrenergic beta-Antagonists/pharmacology , Macrophages/immunology , Macrophages/metabolism , Stress, Physiological , Monocytes/immunology , Monocytes/metabolism
18.
Handb Exp Pharmacol ; 285: 247-295, 2024.
Article in English | MEDLINE | ID: mdl-38844580

ABSTRACT

ß-Adrenoceptors (ß-ARs) provide an important therapeutic target for the treatment of cardiovascular disease. Three ß-ARs, ß1-AR, ß2-AR, ß3-AR are localized to the human heart. Activation of ß1-AR and ß2-ARs increases heart rate, force of contraction (inotropy) and consequently cardiac output to meet physiological demand. However, in disease, chronic over-activation of ß1-AR is responsible for the progression of disease (e.g. heart failure) mediated by pathological hypertrophy, adverse remodelling and premature cell death. Furthermore, activation of ß1-AR is critical in the pathogenesis of cardiac arrhythmias while activation of ß2-AR directly influences blood pressure haemostasis. There is an increasing awareness of the contribution of ß2-AR in cardiovascular disease, particularly arrhythmia generation. All ß-blockers used therapeutically to treat cardiovascular disease block ß1-AR with variable blockade of ß2-AR depending on relative affinity for ß1-AR vs ß2-AR. Since the introduction of ß-blockers into clinical practice in 1965, ß-blockers with different properties have been trialled, used and evaluated, leading to better understanding of their therapeutic effects and tolerability in various cardiovascular conditions. ß-Blockers with the property of intrinsic sympathomimetic activity (ISA), i.e. ß-blockers that also activate the receptor, were used in the past for post-treatment of myocardial infarction and had limited use in heart failure. The ß-blocker carvedilol continues to intrigue due to numerous properties that differentiate it from other ß-blockers and is used successfully in the treatment of heart failure. The discovery of ß3-AR in human heart created interest in the role of ß3-AR in heart failure but has not resulted in therapeutics at this stage.


Subject(s)
Adrenergic beta-Antagonists , Heart Failure , Receptors, Adrenergic, beta , Humans , Heart Failure/drug therapy , Heart Failure/physiopathology , Heart Failure/metabolism , Receptors, Adrenergic, beta/metabolism , Receptors, Adrenergic, beta/drug effects , Adrenergic beta-Antagonists/therapeutic use , Adrenergic beta-Antagonists/pharmacology , Cardiovascular Diseases/drug therapy , Cardiovascular Diseases/physiopathology , Tachycardia/drug therapy , Tachycardia/physiopathology , Animals
19.
Cancer Biol Ther ; 25(1): 2366451, 2024 Dec 31.
Article in English | MEDLINE | ID: mdl-38857055

ABSTRACT

BACKGROUND: Chronic stress can induce stress-related hormones; norepinephrine (NE) is considered to have the highest potential in cancer. NE can stimulate the expression of hypoxia-inducible factor-1α (HIF-1α), which is associated with vascular endothelial growth factor (VEGF) secretion and tumor angiogenesis. However, the underlying mechanisms are poorly understood. METHODS: Tumor-bearing mice were subjected to chronic restraint stress and treated with normal saline, human monoclonal VEGF-A neutralizing antibody bevacizumab, or ß-adrenergic receptor (ß-AR) antagonist (propranolol). Tumor growth and vessel density were also evaluated. Human colorectal adenocarcinoma cells were treated with NE, propranolol, or the inhibitor of transforming growth factor-ß (TGF-ß) receptor Type I kinase (Ly2157299) in vitro. TGF-ß1 in mouse serum and cell culture supernatants was quantified using ELISA. The expression of HIF-1α was measured using Real time-PCR and western blotting. Cell migration and invasion were tested. RESULTS: Chronic restraint stress attenuated the efficacy of bevacizumab and promoted tumor growth and angiogenesis in a colorectal tumor model. Propranolol blocked this effect and inhibited TGF-ß1 elevation caused by chronic restraint stress or NE. NE upregulated HIF-1α expression, which was reversed by propranolol or Ly2157299. Propranolol and Ly2157199 blocked NE-stimulated cancer cell migration and invasion. CONCLUSIONS: Our results demonstrate the effect of NE on tumor angiogenesis and the critical role of TGF-ß1 signaling during this process. In addition, ß-AR/TGF-ß1 signaling/HIF-1α/VEGF is a potential signaling pathway. This study also indicates that psychosocial stress might be a risk factor which weakens the efficacy of anti-angiogenic therapy.


Subject(s)
Bevacizumab , Colorectal Neoplasms , Hypoxia-Inducible Factor 1, alpha Subunit , Neovascularization, Pathologic , Signal Transduction , Transforming Growth Factor beta1 , Animals , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Colorectal Neoplasms/drug therapy , Humans , Neovascularization, Pathologic/metabolism , Mice , Transforming Growth Factor beta1/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Bevacizumab/pharmacology , Bevacizumab/therapeutic use , Propranolol/pharmacology , Cell Line, Tumor , Vascular Endothelial Growth Factor A/metabolism , Male , Cell Movement , Norepinephrine/pharmacology , Norepinephrine/metabolism , Stress, Psychological/complications , Stress, Psychological/metabolism , Adrenergic beta-Antagonists/pharmacology , Adrenergic beta-Antagonists/therapeutic use , Angiogenesis , Pyrazoles , Quinolines
20.
Ann Noninvasive Electrocardiol ; 29(4): e13128, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38872457

ABSTRACT

BACKGROUND: The aim was to evaluate the effect of beta-blockers (BB) on the response of heart rate (HR) to 6-min walk test (6MWT) in atrial fibrillation (AF) and whether the AF patients treated with BB have a similar HR response to 6MWT as the AF and sinus rhythm (SR) patients without BB treatment at the same resting HR level. METHODS: The before-after study involving 74 AF patients was to evaluate the effect of BB treatment (pre-BB and with BB). The comparison study included 74 BB-treated AF patients (with BB), 74 matched AF patients without BB (no BB), and 74 SR patients. The percentage increase amplitude of HR (HR-PIA) in 6MWT was calculated: [(the exercise HR - the resting HR)/(the resting HR)] × 100%. RESULTS: The before-after study showed that BB treatment decreased the resting and mean exercise HR (98.6 ± 15.2 vs. 85.5 ± 11.2 bpm and 121.3 ± 17.3 vs. 109.0 ± 16.7 bpm) during 6MWT. The comparison study demonstrated that against the SR, the AF with BB and no BB groups have higher mean exercise HR-PIA (28.2 ± 17.1% and 22.0 ± 9.6%, vs. 6.9 ± 3.7%) when their resting HR is similar. Moreover, the mean exercise HR-PIA was also significantly higher in the with BB group than in the no BB group. CONCLUSION: In AF patients with relatively higher resting HR, BB treatment could decrease the resting and exercise HR during 6MWT. However, BB treatment could not effectively attenuate the exercise HR rise as compared with AF without BB treatment, even with similar resting HR levels.


Subject(s)
Adrenergic beta-Antagonists , Atrial Fibrillation , Exercise Test , Heart Rate , Humans , Atrial Fibrillation/drug therapy , Atrial Fibrillation/physiopathology , Heart Rate/drug effects , Heart Rate/physiology , Male , Female , Adrenergic beta-Antagonists/therapeutic use , Adrenergic beta-Antagonists/pharmacology , Aged , Middle Aged , Exercise Test/methods , Exercise Test/drug effects , Walk Test/methods , Walking/physiology , Treatment Outcome , Electrocardiography/methods , Electrocardiography/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL