Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 33.951
1.
Biol Pharm Bull ; 47(6): 1123-1127, 2024.
Article En | MEDLINE | ID: mdl-38839364

This study aimed to validate the In vitro Dissolution Absorption System 2 (IDAS2) containing a biological barrier of Caco-2 or Madin-Darby canine kidney (MDCK) cell monolayer through dose sensitivity studies. Metoprolol and propranolol were selected as Biopharmaceutics Classification System (BCS) Class I model drugs, and atenolol as a Class III model drug. The IDAS2 is comprised of a dissolution vessel (500 mL) and two permeation chambers (2 × 8.0 mL) mounted with Caco-2 or MDCK cell monolayer. One or two immediate-release tablet(s) of the model drug were added to the dissolution vessel, and the time profiles of dissolution and permeation were observed. Greater than 85% of metoprolol and propranolol (tested at two dosing concentrations) were dissolved by 15 min, and all drugs were fully dissolved by 30 min. All three drugs were more permeable across Caco-2 cells than MDCK cells with a linear increase in permeation across both cells at both dose concentrations. Thus, the dose sensitivity of the IDAS2 was demonstrated using both cell barriers. These results indicate a successful qualification of IDAS2 for the development/optimization of oral formulations and that MDCK cells can be utilized as a surrogate for Caco-2 cells.


Atenolol , Metoprolol , Propranolol , Solubility , Dogs , Caco-2 Cells , Humans , Animals , Madin Darby Canine Kidney Cells , Propranolol/pharmacokinetics , Metoprolol/pharmacokinetics , Metoprolol/administration & dosage , Atenolol/pharmacokinetics , Atenolol/administration & dosage , Dose-Response Relationship, Drug , Biopharmaceutics/methods , Permeability , Intestinal Absorption
2.
Thromb Res ; 238: 208-221, 2024 Jun.
Article En | MEDLINE | ID: mdl-38733693

BACKGROUND & AIMS: Nonselective ß blockers (NSBBs) facilitate the development of portal vein thrombosis (PVT) in liver cirrhosis. Considering the potential effect of NSBBs on neutrophils and neutrophil extracellular traps (NETs), we speculated that NSBBs might promote the development of PVT by stimulating neutrophils to release NETs. MATERIALS AND METHODS: Serum NETs biomarkers were measured, use of NSBBs was recorded, and PVT was evaluated in cirrhotic patients. Carbon tetrachloride and ferric chloride (FeCl3) were used to induce liver fibrosis and PVT in mice, respectively. After treatment with propranolol and DNase I, neutrophils in peripheral blood, colocalization and expression of NETs in PVT specimens, and NETs biomarkers in serum were measured. Ex vivo clots lysis analysis was performed and portal vein velocity and coagulation parameters were tested. RESULTS: Serum MPO-DNA level was significantly higher in cirrhotic patients treated with NSBBs, and serum H3Cit and MPO-DNA levels were significantly higher in those with PVT. In fibrotic mice, following treatment with propranolol, DNase I significantly shortened the time of FeCl3-induced PVT formation, lowered the peripheral blood neutrophils labelled by CD11b/Ly6G, inhibited the positive staining of H3Cit and the expression of H3Cit and MPO proteins in PVT tissues, and reduced serum nucleosome level. Furthermore, the addition of DNase I to tissue plasminogen activator (tPA) significantly accelerated clots lysis as compared with tPA alone. Propranolol reduced portal vein velocity in fibrotic mice, but did not influence coagulation parameters. CONCLUSION: Our study provides a clue to the potential impact of NETs formation on the association of NSBBs with the development of PVT.


Extracellular Traps , Portal Vein , Propranolol , Venous Thrombosis , Extracellular Traps/metabolism , Extracellular Traps/drug effects , Propranolol/pharmacology , Propranolol/therapeutic use , Humans , Animals , Portal Vein/pathology , Portal Vein/metabolism , Venous Thrombosis/metabolism , Venous Thrombosis/pathology , Venous Thrombosis/drug therapy , Venous Thrombosis/blood , Male , Mice , Female , Middle Aged , Neutrophils/metabolism , Neutrophils/drug effects , Liver Cirrhosis/pathology , Liver Cirrhosis/metabolism , Mice, Inbred C57BL , Adult , Aged
3.
BMC Pediatr ; 24(1): 368, 2024 May 28.
Article En | MEDLINE | ID: mdl-38807073

BACKGROUND: Lip infantile hemangiomas tend to show less volumetric regression and are more susceptible to visible sequelae in the involuted stage. Some of them still require surgical management after propranolol therapy. This study aimed to evaluate the efficacy and safety of the Stepwise, Multi-Incisional, and Single-Stage (SMISS) approach applied to lip reduction for those with involuted lip hemangiomas. METHODS: A retrospective review was performed to evaluate patients with lip hemangioma who received previous propranolol treatment and underwent the aforementioned procedure. Demographic characteristics, lesion morphology, and medical history were reviewed. The Visual Analog Scale was applied to assess the postoperative appearance. Complications within 12 months postoperatively were recorded. RESULTS: A total of 18 patients with lip hemangioma were eligible. All patients received oral propranolol therapy before surgery, with treatment duration ranging from 6.0 to 23.0 months. Their age at surgery ranged from 2.5 to 9.0 years. The median Visual Analog Scale scores were 8.0, ranging from 4.0 to 10.0. No severe complications were reported. CONCLUSIONS: This modified technique based on the SMISS approach has proven reliable and effective in improving the aesthetic outcome for involuted lip infantile hemangiomas. Practical surgical techniques still play an important part in the propranolol era.


Hemangioma , Lip Neoplasms , Propranolol , Humans , Retrospective Studies , Male , Female , Hemangioma/surgery , Lip Neoplasms/surgery , Propranolol/therapeutic use , Child, Preschool , Child , Infant , Lip/surgery , Treatment Outcome , Lipoma/surgery
4.
Ecotoxicol Environ Saf ; 279: 116510, 2024 Jul 01.
Article En | MEDLINE | ID: mdl-38810284

Microplastics and organic micropollutants are two emerging contaminants that interact with each other in environmental and engineered systems. Sorption of organic micropollutants, such as pharmaceuticals, pesticides and industrial compounds, to microplastics can modify their bioavailability and biodegradation. The present study investigated the capacity of ultra-high density polyethylene particles (125 µm in diameter), before and after aging, to sorb 21 organic micropollutants at different environmentally relevant concentration. Furthermore, the biodegradation of these organic micropollutants by a biofilm microbial community growing on the microplastic surface was compared with the biodegradation by a microbial community originating from activated sludge. Among all tested organic micropollutants, propranolol (70%), trimethoprim (25%) and sotalol (15%) were sorbed in the presence of polyethylene particles. Growth of a biofilm on the polyethylene particles had a beneficial effect on the sorption of bromoxynil, caffeine and chloridazon and on the biodegradation of irbesartan, atenolol and benzotriazole. On the other hand, the biofilm limited the sorption of trimethoprim, propranolol, sotalol and benzotriazole and the biodegradation of 2,4-D. These results showed that ultra-high density polyethylene particles can affect both in a positive and negative way for the abiotic and biotic removal of organic micropollutants in wastewater. This project highlights the need for further investigation regarding the interaction between microplastics and organic micropollutants in the aquatic environment.


Biodegradation, Environmental , Biofilms , Microplastics , Polyethylene , Propranolol , Water Pollutants, Chemical , Water Pollutants, Chemical/analysis , Polyethylene/chemistry , Adsorption , Trimethoprim , Atenolol , Triazoles/chemistry , Sewage/chemistry , Sewage/microbiology
5.
J Anxiety Disord ; 104: 102870, 2024 Jun.
Article En | MEDLINE | ID: mdl-38733644

Exposure therapy is an evidence-based treatment option for anxiety-related disorders. Many patients also take medication that could, in principle, affect exposure therapy efficacy. Clinical and laboratory evidence indeed suggests that benzodiazepines may have detrimental effects. Large clinical trials with propranolol, a common beta-blocker, are currently lacking, but several preclinical studies do indicate impaired establishment of safety memories. Here, we investigated the effects of propranolol given prior to extinction training in 9 rat studies (N = 215) and one human study (N = 72). A Bayesian meta-analysis of our rat studies provided strong evidence against propranolol-induced extinction memory impairment during a drug-free test, and the human study found no significant difference with placebo. Two of the rat studies actually suggested a small beneficial effect of propranolol. Lastly, two rat studies with a benzodiazepine (midazolam) group provided some evidence for a harmful effect on extinction memory, i.e., impaired extinction retention. In conclusion, our midazolam findings are in line with prior literature (i.e., an extinction retention impairment), but this is not the case for the 10 studies with propranolol. Our data thus support caution regarding the use of benzodiazepines during exposure therapy, but argue against a harmful effect of propranolol on extinction learning.


Adrenergic beta-Antagonists , Extinction, Psychological , Fear , Memory , Midazolam , Propranolol , Propranolol/pharmacology , Propranolol/administration & dosage , Animals , Fear/drug effects , Extinction, Psychological/drug effects , Rats , Humans , Adrenergic beta-Antagonists/pharmacology , Adrenergic beta-Antagonists/administration & dosage , Male , Memory/drug effects , Midazolam/pharmacology , Midazolam/administration & dosage , Midazolam/adverse effects , Adult , Bayes Theorem , Female , Conditioning, Classical/drug effects , Young Adult
6.
Chemosphere ; 357: 141985, 2024 Jun.
Article En | MEDLINE | ID: mdl-38614404

Carbonate radical (CO3•-) has been proved to be an important secondary radical in advanced oxidation processes due to various radical reactions involved HCO3-/CO32-. However, the roles and contributions of CO3•- in organic micropollutant degradation have not been explored systematically. Here, we quantified the impact of CO3•- on the degradation kinetics of propranolol, a representative pollutant in the UV/peroxymonosulfate (PMS) system, by constructing a steady-state radical model. Substantially, the measured values were coincident with the predictive values, and the contributions of CO3•- on propranolol degradation were the water matrix-dependent. Propranolol degradation increased by 130% in UV/PMS system containing 10 mM HCO3-, and the contribution of CO3•- was as high as 58%. Relatively high pH values are beneficial for propranolol degradation in pure water containing HCO3-, and the contributions of CO3•- also enhanced, while an inverse phenomenon was shown for the effects of propranolol concentrations. Dissolved organic matter exhibited significant scavenging effects on HO•, SO4•-, and CO3•-, substantially retarding the elimination process. The developed model successfully predicted oxidation degradation kinetics of propranolol in actual sewage, and CO3•- contribution was up to 93%, which in indicative of the important role of CO3•- in organic micropollutant removal via AOPs treatment.


Carbonates , Oxidation-Reduction , Peroxides , Propranolol , Ultraviolet Rays , Water Pollutants, Chemical , Propranolol/chemistry , Water Pollutants, Chemical/chemistry , Carbonates/chemistry , Kinetics , Peroxides/chemistry , Hydrogen-Ion Concentration
7.
Am J Physiol Cell Physiol ; 326(5): C1334-C1344, 2024 May 01.
Article En | MEDLINE | ID: mdl-38557356

Cardiac maturation represents the last phase of heart development and is characterized by morphofunctional alterations that optimize the heart for efficient pumping. Its understanding provides important insights into cardiac regeneration therapies. Recent evidence implies that adrenergic signals are involved in the regulation of cardiac maturation, but the mechanistic underpinnings involved in this process are poorly understood. Herein, we explored the role of ß-adrenergic receptor (ß-AR) activation in determining structural and functional components of cardiomyocyte maturation. Temporal characterization of tyrosine hydroxylase and norepinephrine levels in the mouse heart revealed that sympathetic innervation develops during the first 3 wk of life, concurrent with the rise in ß-AR expression. To assess the impact of adrenergic inhibition on maturation, we treated mice with propranolol, isolated cardiomyocytes, and evaluated morphofunctional parameters. Propranolol treatment reduced heart weight, cardiomyocyte size, and cellular shortening, while it increased the pool of mononucleated myocytes, resulting in impaired maturation. No changes in t-tubules were observed in cells from propranolol mice. To establish a causal link between ß-AR signaling and cardiomyocyte maturation, mice were subjected to sympathectomy, followed or not by restoration with isoproterenol treatment. Cardiomyocytes from sympathectomyzed mice recapitulated the salient immaturity features of propranolol-treated mice, with the additional loss of t-tubules. Isoproterenol rescued the maturation deficits induced by sympathectomy, except for the t-tubule alterations. Our study identifies the ß-AR stimuli as a maturation promoting signal and implies that this pathway can be modulated to improve cardiac regeneration therapies.NEW & NOTEWORTHY Maturation involves a series of morphofunctional alterations vital to heart development. Its regulatory mechanisms are only now being unveiled. Evidence implies that adrenergic signaling regulates cardiac maturation, but the mechanisms are poorly understood. To address this point, we blocked ß-ARs or performed sympathectomy followed by rescue experiments with isoproterenol in neonatal mice. Our study identifies the ß-AR stimuli as a maturation signal for cardiomyocytes and highlights the importance of this pathway in cardiac regeneration therapies.


Myocytes, Cardiac , Propranolol , Signal Transduction , Animals , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/drug effects , Mice , Propranolol/pharmacology , Receptors, Adrenergic, beta/metabolism , Mice, Inbred C57BL , Isoproterenol/pharmacology , Male , Heart/drug effects , Cells, Cultured , Adrenergic beta-Agonists/pharmacology , Norepinephrine/metabolism , Norepinephrine/pharmacology , Adrenergic beta-Antagonists/pharmacology
8.
J Mater Chem B ; 12(17): 4217-4231, 2024 May 01.
Article En | MEDLINE | ID: mdl-38596904

Numerous studies have shown that there are multiple neural activities involved in the process of bone resorption and bone regeneration, and promoting osteogenesis by promoting neural network reconstruction is an effective strategy for repairing critical size bone defects. However, traumatic bone defects often cause activation of the sympathetic nervous system (SNS) in the damaged area, releasing excess catecholamines (CAs), resulting in a decrease in the rate of bone formation. Herein, a 3D-printed scaffold loaded with propranolol (PRN) is proposed to reduce CA concentrations in bone defect areas and promote bone regeneration through drug release. For this purpose, PRN-loaded methacrylated gelatin (GelMA) microspheres were mixed with low-concentration GelMA and perfused into a 3D-printed porous hydroxyapatite (HAp) scaffold. By releasing PRN, which can block ß-adrenergic receptors, it hinders the activation of sympathetic nerves and inhibits the release of excess CA by the SNS. At the same time, the composite scaffold recruits bone marrow mesenchymal stem cells (BMSCs) and promotes the differentiation of BMSCs in the direction of osteoblasts, which effectively promotes bone regeneration in the rabbit femoral condyle defect model. The results of the study showed that the release of PRN from the composite scaffold could effectively hinder the activation of sympathetic nerves and promote bone regeneration, providing a new strategy for the treatment of bone defects.


Bone Regeneration , Mesenchymal Stem Cells , Printing, Three-Dimensional , Sympathetic Nervous System , Tissue Scaffolds , Bone Regeneration/drug effects , Animals , Rabbits , Sympathetic Nervous System/drug effects , Mesenchymal Stem Cells/drug effects , Tissue Scaffolds/chemistry , Propranolol/pharmacology , Propranolol/chemistry , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Gelatin/chemistry , Osteogenesis/drug effects , Durapatite/chemistry , Durapatite/pharmacology
9.
Physiol Behav ; 280: 114564, 2024 Jun 01.
Article En | MEDLINE | ID: mdl-38657747

Although salivation is essential during eating behavior, little is known about the brainstem centers that directly control the salivary glands. With regard to the inferior salivatory nucleus (ISN), the site of origin of the parasympathetic preganglionic cell bodies that innervate the parotid glands, previous anatomical studies have located it within the rostrodorsal medullary reticular formation. However, to date there is no functional data that shows the secretory nature of the somas grouped in this region. To activate only the somas and rule out the activation of the efferent fibers from and the afferent fibers to the ISN, in exp. 1, NMDA neurotoxin was administered to the rostrodorsal medullary region and the secretion of saliva was recorded during the following hour. Results showed an increased secretion of parotid saliva but a total absence of submandibular-sublingual secretion. In exp. 2, results showed that the hypersecretion of parotid saliva after NMDA microinjection was completely blocked by the administration of atropine (a cholinergic blocker) but not after administration of dihydroergotamine plus propranolol (α and ß-adrenergic blockers, respectively). These findings suggest that the somata of the rostrodorsal medulla are secretory in nature, controlling parotid secretion via a cholinergic pathway. The data thus functionally supports the idea that these cells constitute the ISN.


N-Methylaspartate , Parotid Gland , Receptors, N-Methyl-D-Aspartate , Salivation , Animals , Male , Rats , Adrenergic beta-Antagonists/pharmacology , Atropine/pharmacology , Excitatory Amino Acid Agonists/pharmacology , Medulla Oblongata/metabolism , Medulla Oblongata/drug effects , Microinjections , N-Methylaspartate/pharmacology , N-Methylaspartate/metabolism , Parotid Gland/metabolism , Parotid Gland/drug effects , Propranolol/pharmacology , Rats, Wistar , Receptors, N-Methyl-D-Aspartate/metabolism , Saliva/metabolism , Salivation/drug effects , Salivation/physiology , Sialorrhea
10.
Pediatr Blood Cancer ; 71(7): e30989, 2024 Jul.
Article En | MEDLINE | ID: mdl-38602300

BACKGROUND: Infantile hemangiomas (IHs) are common benign vascular tumors in infants. Apelin, an endogenous cytokine, is implicated in the angiogenesis of neoplastic diseases. We aimed to explore the association between apelin and IHs, providing a foundation for clinical applications. METHODS: We identified differential expression of apelin in proliferative IHs compared to healthy controls (HCs) through bioinformatics analysis of publicly available databases and verified by Immunofluorescence. Enzyme-linked immunosorbent assay was used to quantify the serum levels of apelin and vascular endothelial growth factor (VEGF) in a cohort of 116 cases of proliferative IHs, 65 cases of capillary malformations (CMs), and 70 HCs. RESULTS: Apelin and APJ (APLNR, apelin receptor) were identified as the significantly upregulated differentially expressed genes (DEGs) in proliferative IHs. Immunofluorescence staining indicated high expression of apelin in proliferative IHs, while minimal expression in non-IH lesions. Apelin in IHs was reduced following 6 months of propranolol treatment. Serum apelin levels were significantly higher in the IH group compared to both the CM and HC groups. Moreover, apelin exhibited excellent discriminatory ability in distinguishing IHs from HCs, with an area under the curve (AUC) exceeding 0.90. A positive correlation was observed between the levels of apelin and the size of superficial IHs. The expression profiles of VEGF and apelin in IHs were found to be consistent. CONCLUSIONS: Apelin shows promise as a potential biomarker for IHs. The association between apelin and IH size, as well as its responsiveness to propranolol treatment, indicates its possible utility as a valuable indicator for the therapeutic evaluation of IHs.


Apelin , Biomarkers, Tumor , Humans , Apelin/blood , Infant , Male , Female , Biomarkers, Tumor/blood , Hemangioma/blood , Hemangioma/pathology , Apelin Receptors/blood , Apelin Receptors/metabolism , Vascular Endothelial Growth Factor A/blood , Case-Control Studies , Propranolol/therapeutic use , Prognosis , Infant, Newborn
11.
BMJ Case Rep ; 17(4)2024 Apr 02.
Article En | MEDLINE | ID: mdl-38569730

An outborn male term neonate presented with a complaint of respiratory distress since birth on day 9 of life. On examination, baby was having tachypnoea, tachycardia and hepatomegaly. The baby was delivered at term gestation and cried immediately after birth. The chest X-ray showed cardiomegaly. The abdomen ultrasound showed a complex cystic vascular lesion suggestive of hepatic haemangioma. The echocardiography showed an atrial septal defect. The baby was initially treated conservatively along with specific treatment (steroids and propranolol) for haemangioma for 6 weeks. However, the symptoms persisted and there was non-resolution, therefore, particle embolisation of the right hepatic artery was performed. Subsequently, it resulted in complete resolution of the lesion.


Hemangioma , Liver Neoplasms , Infant, Newborn , Infant , Humans , Male , Hemangioma/diagnostic imaging , Hemangioma/therapy , Liver Neoplasms/diagnostic imaging , Liver Neoplasms/therapy , Propranolol/therapeutic use , Hepatic Artery , Ultrasonography
12.
J Clin Invest ; 134(8)2024 Apr 15.
Article En | MEDLINE | ID: mdl-38618963

Infantile hemangioma (IH) is a benign vascular tumor that occurs in 5% of newborns. The tumor follows a life cycle of rapid proliferation in infancy, followed by slow involution in childhood. This unique life cycle has attracted the interest of basic and clinical scientists alike as a paradigm for vasculogenesis, angiogenesis, and vascular regression. Unanswered questions persist about the genetic and molecular drivers of the proliferating and involuting phases. The beta blocker propranolol usually accelerates regression of problematic IHs, yet its mechanism of action on vascular proliferation and differentiation is unclear. Some IHs fail to respond to beta blockers and regrow after discontinuation. Side effects occur and long-term sequelae of propranolol treatment are unknown. This poses clinical challenges and raises novel questions about the mechanisms of vascular overgrowth in IH.


Hemangioma , Physicians , Vascular Neoplasms , Infant, Newborn , Humans , Propranolol/therapeutic use , Disease Progression , Hemangioma/drug therapy
13.
Int J Mol Sci ; 25(7)2024 Mar 26.
Article En | MEDLINE | ID: mdl-38612493

Adrenergic pathways represent the main channel of communication between the nervous system and the immune system. During inflammation, blood monocytes migrate within tissue and differentiate into macrophages, which polarize to M1 or M2 macrophages with tissue-damaging or -reparative properties, respectively. This study investigates whether the ß-adrenergic receptor (ß-AR)-blocking drug propranolol modulates the monocyte-to-macrophage differentiation process and further influences macrophages in their polarization toward M1- and M2-like phenotypes. Six-day-human monocytes were cultured with M-CSF in the presence or absence of propranolol and then activated toward an M1 pro-inflammatory state or an M2 anti-inflammatory state. The chronic exposure of monocytes to propranolol during their differentiation into macrophages promoted the increase in the M1 marker CD16 and in the M2 markers CD206 and CD163 and peroxisome proliferator-activated receptor É£ expression. It also increased endocytosis and the release of IL-10, whereas it reduced physiological reactive oxygen species. Exposure to the pro-inflammatory conditions of propranolol-differentiated macrophages resulted in an anti-inflammatory promoting effect. At the molecular level, propranolol upregulated the expression of the oxidative stress regulators NRF2, heme oxygenase-1 and NQO1. By contributing to regulating macrophage activities, propranolol may represent a novel anti-inflammatory and immunomodulating compound with relevant therapeutic potential in several inflammatory diseases.


Monocytes , Propranolol , Humans , Propranolol/pharmacology , Antioxidants/pharmacology , NF-E2-Related Factor 2 , Macrophages , Anti-Inflammatory Agents/pharmacology
14.
J Environ Sci (China) ; 142: 57-68, 2024 Aug.
Article En | MEDLINE | ID: mdl-38527896

Non-radical activation of persulfate (PS) by photocatalysts is an effective approach for removing organic pollutants from aqueous environments. In this study, a novel Bi2O3/BiO1.3I0.4 heterojunction was synthesized using a facile solvothermal approach and used for the first time for non-radical activation of PS to degrade propranolol (PRO) in the presence of visible light. The findings found that the degradation rate of PRO in the Bi2O3/BiO1.3I0.4/PS system was significantly increased from 19% to more than 90% within 90 min compared to the Bi2O3/BiO1.3I0.4 system. This indicated that the composite system exerted an excellent synergistic effect between the photocatalyst and the persulfate-based oxygenation. Quenching tests and electron paramagnetic resonance demonstrated that the non-radical pathway with singlet oxygen as the active species played a major role in the photocatalytic process. The existence of photo-generated holes during the reaction could also be directly involved in the oxidation of pollutants. Meanwhile, a possible PRO degradation pathway was also proposed. Furthermore, the impacts of pH, humic acid and common anions on the PRO degradation by the Bi2O3/BiO1.3I0.4/PS were explored, and the system's stability and reusability were also studied. This study exhibits a highly productive catalyst for PS activation via a non-radical pathway and provides a new idea for the degradation of PRO.


Environmental Pollutants , Propranolol , Singlet Oxygen , Oxidation-Reduction , Light
15.
Eur J Cancer ; 202: 113974, 2024 May.
Article En | MEDLINE | ID: mdl-38452721

BACKGROUND: Angiosarcoma is a rare and aggressive cancer of the endothelial cells. Propranolol, a non-selective ß-blocker, was able to initiate apoptosis in angiosarcoma cell lines and its anti-tumor activity has been described in several case reports. The aim of this trial was to prospectively evaluate the anti-tumor activity of propranolol monotherapy in patients with angiosarcoma before proceeding to standard of care treatment. METHODS: Propranolol was dosed 80 mg to 240 mg/day for 3 to 6 weeks according to a dose titration schedule. The primary endpoint was clinical response (response according to RECIST 1.1 or stable disease with improvement of cutaneous lesions) in at least three patients. Exploratory objectives included histologic response (>30% decrease in Ki-67), FDG PET response, and ß-receptor expression levels. RESULTS: Fourteen patients were enrolled. The median duration of treatment was 26 days (range 21-42 days). The median highest propranolol dose was 160 mg/day (range 80 - 240 mg). Two patients showed clinical response (14%, 95% CI 3-100%). One of these patients showed a partial metabolic response on PET-CT. None of the tumors showed histologic response. The most common adverse event was grade 1/2 bradycardia (86%). There were no grade ≥ 3 adverse events. ADRB2 was overexpressed in 16 out of 18 tumors, in both responders and non-responders. None of the tumors showed ADRB1 overexpression. CONCLUSIONS: This window-of-opportunity trial did not show clinical efficacy of propranolol monotherapy. However, two out of 14 patients did show clinical benefit. ADRB1/2 expression did not correlate with clinical response.


Hemangiosarcoma , Propranolol , Humans , Propranolol/therapeutic use , Hemangiosarcoma/drug therapy , Positron Emission Tomography Computed Tomography , Endothelial Cells , Adrenergic beta-Antagonists/therapeutic use
16.
Toxicol Appl Pharmacol ; 484: 116881, 2024 Mar.
Article En | MEDLINE | ID: mdl-38437958

Pain has a negative impact on public health, reducing quality of life. Unfortunately, current treatments are not fully effective and have adverse effects. Therefore, there is a need to develop new analgesic compounds. Due to promising results regarding the antinociceptive effect of N-(3-(phenylselanyl)prop-2-in-1-yl)benzamide (SePB), this study aimed to evaluate the participation of the dopaminergic and noradrenergic systems in this effect in mice, as well as its toxicity. To this, the antagonists sulpiride (D2/D3 receptor antagonist, 5 mg/kg), SCH-23390 (D1 receptor antagonist, 0.05 mg/kg), prazosin (α1 adrenergic receptor antagonist, 0.15 mg/kg), yohimbine (α2-adrenergic receptors, 0.15 mg/kg) and propranolol (non-selective ß-adrenergic antagonist, 10 mg/kg) were administered intraperitoneally to mice 15 min before SePB (10 mg/kg, intragastrically), except for propranolol (20 min). After 26 min of SePB administration, the open field test was performed for 4 min to assess locomotor activity, followed by the tail immersion test to measure the nociceptive response. For the toxicity test, animals received a high dose of 300 mg/kg of SePB. SePB showed an increase in the latency for nociceptive response in the tail immersion test, and this effect was prevented by SCH-23390, yohimbine and propranolol, indicating the involvement of D1, α2 and ß-adrenergic receptors in the antinociceptive mechanism of the SePB effect. No changes were observed in the open field test, and the toxicity assessment suggested that SePB has low potential to induce toxicity. These findings contribute to understanding SePB's mechanism of action, with a focus on the development of new alternatives for pain treatment.


Propranolol , Quality of Life , Mice , Animals , Propranolol/pharmacology , Propranolol/therapeutic use , Analgesics/toxicity , Pain/drug therapy , Norepinephrine , Yohimbine/toxicity , Yohimbine/therapeutic use , Adrenergic alpha-1 Receptor Antagonists/therapeutic use , Dopamine , Sulpiride , Receptors, Adrenergic, alpha-2
17.
Eur J Clin Pharmacol ; 80(6): 901-910, 2024 Jun.
Article En | MEDLINE | ID: mdl-38436704

PURPOSE: To systematically review the impact of propranolol combined with oxytocin on the process and outcomes of labor. METHODS: A comprehensive literature search was performed across multiple databases, including China National Knowledge Infrastructure (CNKI), VIP, Wanfang, China Biomedical Literature Database, PubMed, Embase, and the Cochrane Library. All publicly published randomized controlled trials (RCTs) of propranolol combined with oxytocin compared to the use of oxytocin alone in labor were collected. After screening the literature and extracting data, the Cochrane Handbook for Systematic Reviews of Interventions 5.1.0 recommended bias risk assessment tool was used to assess the quality of the included studies. A meta-analysis was conducted using RevMan 5.3 software, and the Grading of Recommendations Assessment, Development and Evaluation (GRADE) system was used to rate the quality of evidence for outcome measures. RESULTS: Meta-analysis results showed that the group receiving propranolol combined with oxytocin was more capable of reducing the cesarean section rate (eight studies, 815 women, RR = 0.67, 95% CI (0.53, 0.86), P = 0.001) and shortening the duration of the latent phase (two studies, 206 women, MD = - 1.20, 95% CI (- 1.97, - 0.43), P = 0.002) and the duration of the active phase on day 1 (two studies, 296 women, MD = - 0.69, 95% CI (- 0.83, - 0.54), P < 0.00001), compared to the oxytocin monotherapy group. No significant difference was found between the two groups in terms of the 5-min Apgar score (five studies, 609 women, MD = - 0.05, 95% CI (- 0.14, 0.04), P = 0.32) and the rate of admissions to the Neonatal Intensive Care Unit (NICU) (three studies, 359 women, RR = 0.82, 95% CI (0.38, 1.79), P = 0.62). CONCLUSION: The combined use of propranolol and oxytocin can significantly reduce the cesarean section rate, shorten the duration of the latent phase and the duration of the active phase on day 1, and is safe. However, due to the limitations, the conclusions of this article still need to be verified by large-sample, multicenter, rigorously designed high-quality clinical RCTs. TRIAL REGISTRATION: Registration number is INPLASY202390107.


Cesarean Section , Drug Therapy, Combination , Oxytocin , Propranolol , Randomized Controlled Trials as Topic , Humans , Propranolol/administration & dosage , Oxytocin/administration & dosage , Pregnancy , Female , Labor, Obstetric/drug effects , Oxytocics/administration & dosage , Oxytocics/therapeutic use
19.
Sci Total Environ ; 925: 171769, 2024 May 15.
Article En | MEDLINE | ID: mdl-38499104

Aquatic ecosystems continue to be threatened by chemical pollution. To what extent organisms are able to cope with chemical exposure depends on their ability to display mechanisms of defense across different organs. Among these mechanisms, biotransformation processes represent key physiological responses that facilitate detoxification and reduce the bioaccumulation potential of chemicals. Biotransformation does not only depend on the ability of different organs to display biotransformation enzymes but also on the affinity of chemicals towards these enzymes. In the present study, we explored the ability of different organs and of two freshwater fish to support biotransformation processes through the determination of in vitro phase I and II biotransformation enzyme activity, and their role in supporting intrinsic clearance and the formation of biotransformation products. Three environmentally relevant pollutants were evaluated: the polycyclic aromatic hydrocarbon (PAH) pyrene (as recommended by the OECD 319b test guideline), the fungicide azoxystrobin, and the pharmaceutical propranolol. Comparative studies using S9 sub-cellular fractions derived from the liver, intestine, gills, and brain of brown trout (Salmo trutta) and rainbow trout (Oncorhynchus mykiss) revealed significant phase I and II enzyme activity in all organs. However, organ- and species-specific differences were found. In brown trout, significant extrahepatic biotransformation was observed for pyrene but not for azoxystrobin and propranolol. In rainbow trout, the brain appeared to biotransform azoxystrobin. In this same species, propranolol appeared to be biotransformed by the intestine and gills. Biotransformation products could be detected only from hepatic biotransformation, and their profiles and formation rates displayed species-specific patterns and occurred at different magnitudes. Altogether, our findings further contribute to the current understanding of organ-specific biotransformation capacity, beyond the expression and activity of enzymes, and its dependence on specific enzyme-chemical interactions to support mechanisms of defense against exposure.


Ecosystem , Oncorhynchus mykiss , Pyrimidines , Strobilurins , Animals , Propranolol , Liver/metabolism , Oncorhynchus mykiss/metabolism , Pyrenes/metabolism , Biotransformation
20.
Bone Res ; 12(1): 18, 2024 Mar 21.
Article En | MEDLINE | ID: mdl-38514644

The autonomic nervous system plays a crucial role in regulating bone metabolism, with sympathetic activation stimulating bone resorption and inhibiting bone formation. We found that fractures lead to increased sympathetic tone, enhanced osteoclast resorption, decreased osteoblast formation, and thus hastened systemic bone loss in ovariectomized (OVX) mice. However, the combined administration of parathyroid hormone (PTH) and the ß-receptor blocker propranolol dramatically promoted systemic bone formation and osteoporotic fracture healing in OVX mice. The effect of this treatment is superior to that of treatment with PTH or propranolol alone. In vitro, the sympathetic neurotransmitter norepinephrine (NE) suppressed PTH-induced osteoblast differentiation and mineralization, which was rescued by propranolol. Moreover, NE decreased the PTH-induced expression of Runx2 but enhanced the expression of Rankl and the effect of PTH-stimulated osteoblasts on osteoclastic differentiation, whereas these effects were reversed by propranolol. Furthermore, PTH increased the expression of the circadian clock gene Bmal1, which was inhibited by NE-ßAR signaling. Bmal1 knockdown blocked the rescue effect of propranolol on the NE-induced decrease in PTH-stimulated osteoblast differentiation. Taken together, these results suggest that propranolol enhances the anabolic effect of PTH in preventing systemic bone loss following osteoporotic fracture by blocking the negative effects of sympathetic signaling on PTH anabolism.


Anabolic Agents , Bone Resorption , Osteoporotic Fractures , Mice , Animals , Parathyroid Hormone/pharmacology , Anabolic Agents/pharmacology , Osteoporotic Fractures/drug therapy , Propranolol/pharmacology , ARNTL Transcription Factors , Bone Resorption/drug therapy , Adrenergic beta-Antagonists/pharmacology
...