Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 4.040
Filtrar
Mais filtros











Intervalo de ano de publicação
1.
Int J Mol Sci ; 25(7)2024 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-38612493

RESUMO

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.


Assuntos
Monócitos , Propranolol , Humanos , Propranolol/farmacologia , Antioxidantes/farmacologia , Fator 2 Relacionado a NF-E2 , Macrófagos , Anti-Inflamatórios/farmacologia
2.
J Mater Chem B ; 12(17): 4217-4231, 2024 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-38596904

RESUMO

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.


Assuntos
Regeneração Óssea , Células-Tronco Mesenquimais , Impressão Tridimensional , Sistema Nervoso Simpático , Alicerces Teciduais , Regeneração Óssea/efeitos dos fármacos , Animais , Coelhos , Sistema Nervoso Simpático/efeitos dos fármacos , Células-Tronco Mesenquimais/efeitos dos fármacos , Alicerces Teciduais/química , Propranolol/farmacologia , Propranolol/química , Materiais Biocompatíveis/química , Materiais Biocompatíveis/farmacologia , Gelatina/química , Osteogênese/efeitos dos fármacos , Durapatita/química , Durapatita/farmacologia
3.
Cells ; 13(3)2024 Jan 29.
Artigo em Inglês | MEDLINE | ID: mdl-38334645

RESUMO

We previously published that in patients with infantile hemangioma (IH) at the onset (T0) colony forming unit-fibroblasts (CFU-Fs) are present in in vitro cultures from PB. Herein, we characterize these CFU-Fs and investigate their potential role in IH pathogenesis, before and after propranolol therapy. The CFU-F phenotype (by flow cytometry), their differentiation capacity and ability to support angiogenesis (by in vitro cultures) and their gene expression (by RT-PCR) were evaluated. We found that CFU-Fs are actual circulating MSCs (cMSCs). In patients at T0, cMSCs had reduced adipogenic potential, supported the formation of tube-like structures in vitro and showed either inflammatory (IL1ß and ESM1) or angiogenic (F3) gene expression higher than that of cMSCs from CTRLs. In patients receiving one-year propranolol therapy, the cMSC differentiation in adipocytes improved, while their support in in vitro tube-like formation was lost; no difference was found between patient and CTRL cMSC gene expressions. In conclusion, in patients with IH at T0 the cMSC reduced adipogenic potential, their support in angiogenic activity and the inflammatory/angiogenic gene expression may fuel the tumor growth. One-year propranolol therapy modifies this picture, suggesting cMSCs as one of the drug targets.


Assuntos
Hemangioma , Células-Tronco Mesenquimais , Humanos , Propranolol/farmacologia , Propranolol/uso terapêutico , Propranolol/metabolismo , Transcriptoma , Células-Tronco Mesenquimais/metabolismo , Adipogenia/genética , Hemangioma/genética , Hemangioma/tratamento farmacológico , Hemangioma/metabolismo
4.
J Cancer Res Clin Oncol ; 150(2): 59, 2024 Jan 31.
Artigo em Inglês | MEDLINE | ID: mdl-38294713

RESUMO

PURPOSE: This study will focus on 4T1 cells, a murine mammary adenocarcinoma cell line, as the primary research subject. We aim to investigate the inhibitory effects and mechanisms of propranolol on epithelial-mesenchymal transition (EMT) in breast cancer cells, aiming to elucidate this phenomenon at the miRNA level. METHODS: In this study, the EMT inhibitory effect of propranolol was observed through in vitro and animal experiments. For the screening of potential target miRNAs and downstream target genes, second-generation sequencing (SGS) and bioinformatics analysis were conducted. Following the screening process, the identified target miRNAs and their respective target genes were confirmed using various experimental methods. To confirm the target miRNAs and target genes, Western Blot (WB), reverse transcription polymerase chain reaction (RT-PCR), and immunofluorescence experiments were performed. RESULTS: In this study, we found that propranolol significantly reduced lung metastasis in 4T1 murine breast cancer cells (p < 0.05). In vitro and in vivo experiments demonstrated that propranolol inhibited the epithelial-mesenchymal transition (EMT) as evidenced by Western Blot analysis (p < 0.05). Through next-generation sequencing (SGS), subsequent bioinformatics analysis, and PCR validation, we identified a marked downregulation of miR-499-5p (p < 0.05), suggesting its potential involvement in mediating the suppressive effects of propranolol on EMT. Overexpression of miR-499-5p promoted EMT, migration, and invasion of 4T1 cells, and these effects were not reversed or attenuated by propranolol (Validated via Western Blot, wound healing assay, transwell migration, and invasion assays, p < 0.05). Sox6 was identified as a functional target of miR-499-5p, with its downregulation correlating with the observed EMT changes (p < 0.05). Silencing Sox6 or overexpressing miR-499-5p inhibited Sox6 expression, further promoting the processes of EMT, invasion, and migration in 4T1 cells. Notably, these effects were not alleviated by propranolol (validated via Western Blot, wound healing assay, transwell migration, and invasion assays, p < 0.05). The direct interaction between miR-499-5p and Sox6 mRNA was confirmed by dual-luciferase reporter gene assay. CONCLUSION: These results suggest that propranolol may have potential as a therapeutic agent for breast cancer treatment by targeting EMT and its regulatory mechanisms.


Assuntos
Neoplasias da Mama , Transição Epitelial-Mesenquimal , MicroRNAs , Propranolol , Animais , Camundongos , Western Blotting , Linhagem Celular , Transição Epitelial-Mesenquimal/efeitos dos fármacos , MicroRNAs/genética , Propranolol/farmacologia , Fatores de Transcrição SOXD , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/genética
5.
Sci Rep ; 13(1): 21944, 2023 12 11.
Artigo em Inglês | MEDLINE | ID: mdl-38081864

RESUMO

Activated brown fat (aBAT) is known to affect the evaluation of 18F-FDG PET scans, especially in young patients. The aim of this study was to determine factors influencing the occurrence of aBAT, and to investigate the effectiveness of the two preventive measures, warming and beta-blocker (propranolol) administration. Five-hundred-twenty-eight 18F-FDG-PET scans of 241 EuroNet-PHL-C2 trial patients from 41 nuclear medicine departments in Germany and Czech Republic were screened for aBAT. The occurrence of aBAT was analyzed with patient characteristics (age, sex, body mass index, predisposition to aBAT), weather data at the day of 18F-FDG PET scanning as well as the preventive measures taken. Potentially important factors from univariate analyses were included into a logistic regression model. Warming as a preventive measure was used in 243 18F-FDG-PET scans, propranolol was administered in 36, warming and propranolol were combined in 84, and no preventive measures were taken in 165 scans. Whereas age, sex and body mass index had no clear impact, there was an individual predisposition to aBAT. Logistic regression model revealed that the frequency of aBAT mainly depends on the outside temperature (p = 0.005) and can be effectively reduced by warming (p = 0.004), the administration of unselective beta-blocker or the combination of both. Warming is a simple, cheap and non-invasive method to reduce the frequency of aBAT. However, the effect of warming decreases with increasing outside temperatures. Administration of propranolol seems to be equally effective and provides advantages whenever the positive effect of warming is compromised. The combination of both preventive measures could have an additive effect.


Assuntos
Fluordesoxiglucose F18 , Linfoma , Humanos , Tecido Adiposo Marrom/diagnóstico por imagem , Antagonistas Adrenérgicos beta/farmacologia , Fluordesoxiglucose F18/farmacologia , Tomografia por Emissão de Pósitrons/métodos , Propranolol/farmacologia , Compostos Radiofarmacêuticos/farmacologia
6.
Biochem Pharmacol ; 218: 115922, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-37956892

RESUMO

Infantile hemangioma (IH) is the most common benign tumor in children. Propranolol is the first-line treatment for IH, but the underlying mechanism of propranolol treatment in IH is not completely understood. Integrated transcriptional and metabolic analyses were performed to investigate the metabolic changes in hemangioma-derived endothelial cells (HemECs) after propranolol treatment. The findings were then further validated through independent cell experiments using a Seahorse XFp analyzer, Western blotting, immunohistochemistry and mitochondrial functional assays. Thirty-four differentially expressed metabolites, including the glycolysis metabolites glucose 6-phosphate, fructose 6-phosphate and fructose 1,6-bisphosphate, were identified by targeted metabolomics. A KEGG pathway enrichment analysis showed that the disturbances in these metabolites were highly related to glucose metabolism-related pathways, including the pentose phosphate pathway, the Warburg effect, glycolysis and the citric acid cycle. Transcriptional analysis revealed that metabolism-related pathways, including glycine, serine and threonine metabolism, tyrosine metabolism, and glutathione metabolism, were highly enriched. Moreover, integration of the metabolomic and transcriptomic data revealed that glucose metabolism-related pathways, particularly glycolysis, were altered after propranolol treatment. Cell experiments demonstrated that HemECs exhibited higher levels of glycolysis than human umbilical vein ECs (HUVECs) and that propranolol suppressed glycolysis in HemECs. In conclusion, propranolol inhibited glucose metabolism in HemECs by suppressing glucose metabolic pathways, particularly glycolysis.


Assuntos
Células Endoteliais , Hemangioma , Criança , Humanos , Células Endoteliais/metabolismo , Propranolol/farmacologia , Propranolol/metabolismo , Transdução de Sinais , Proliferação de Células , Hemangioma/tratamento farmacológico , Hemangioma/metabolismo , Hemangioma/patologia , Glucose/metabolismo , Fosfatos/farmacologia
7.
Medicine (Baltimore) ; 102(32): e34672, 2023 Aug 11.
Artigo em Inglês | MEDLINE | ID: mdl-37565874

RESUMO

BACKGROUND: Propranolol is the first choice for treating infantile hemangioma (IH). How propranolol works in IH remains unclear. Infantile hemangioma endothelial cells (HemECs) express Notch1, Jagged, Hey1, and other molecules in the Notch pathway, suggesting that Notch pathway-related molecules play an important role in affecting vascular endothelial cell proliferation. Whether propranolol can affect the Notch signaling pathway in IH treatment is unclear. METHODS: We performed this study to observe the effect of propranolol on the expression of Notch signaling pathway molecules in human umbilical vein endothelial cells (HUVECs) and to explore the therapeutic mechanism of propranolol on IH. HUVECs cultured in vitro were exposed to 60, 120, 240, 360, or 480 µM propranolol. The morphological changes of the HUVECs were observed under an inverted microscope. HUVECs proliferation was detected with Cell Counting Kit-8 (CCK-8). The effects of propranolol on HUVECs apoptosis were detected by flow cytometry. The role of Notch in propranolol inhibition of HUVEC proliferation was analyzed with real-time polymerase chain reaction (PCR) and western blotting. RESULTS: Propranolol reduced HUVECs numbers and altered their morphology. The inhibitory effect of propranolol on cell proliferation was dependent on the reaction time and drug concentration. Propranolol upregulated Jagged1, Notch1, and Hey1 expression and downregulated delta-like ligand4 (DLL4) expression. CONCLUSIONS: Propranolol may play a role in IH treatment by increasing Jagged1 expression in endothelial cells, activating the Notch pathway and inducing the upregulation of the downstream target gene HEY1.


Assuntos
Hemangioma , Propranolol , Humanos , Células Endoteliais da Veia Umbilical Humana , Propranolol/farmacologia , Propranolol/uso terapêutico , Transdução de Sinais/genética , Hemangioma/tratamento farmacológico , Hemangioma/genética , Biologia , Proliferação de Células
8.
PeerJ ; 11: e15643, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37456875

RESUMO

Hemangioma (HA) is one of the most common benign vascular tumors among children. Propranolol is used as the first-line treatment for hemangioma and is a non-selective blocker of the ß-adrenergic receptor. ß-elemene is a compound extracted from Rhizoma zedoariae and has been approved for the treatment of tumors in clinical practice. However, the combinatorial effects of ß-elemene and propranolol in the treatment of HA remains unclear. This study explored the combinative effects and mechanisms of ß-elemene and propranolol using hemangioma-derived endothelial cells (HemECs). Cytotoxic assays showed that the combinatorial treatment of ß-elemene and propranolol did not increase the cytotoxic effects of HemECs. Furthermore, functional analysis showed that the combinatorial treatment with ß-elemene and propranolol significantly inhibited the proliferation, migration, and tube formation of the HemECs compared to the single treatment regimens. Mechanistic analysis showed that combinative treatment with ß-elemene and propranolol synergistically down-regulated the hypoxia-inducible factor-1 alpha/vascular endothelial growth factor-A (HIF-1-α/VEGFA) signaling pathway. Additionally, in a xenograft tumor model, angiogenesis in the combinatorial treatment group was significantly lower than in the control, propranolol, and ß-elemene treatment alone groups. Our results suggest that ß-elemene combined with propranolol can significantly inhibit the proliferation, migration, and tube formation of HemECs via synergistically down-regulating the HIF-1-α/VEGFA signaling pathway without increasing any cytotoxic side effects.


Assuntos
Hemangioma , Propranolol , Criança , Humanos , Propranolol/farmacologia , Células Endoteliais , Fator A de Crescimento do Endotélio Vascular , Proliferação de Células , Hemangioma/tratamento farmacológico , Penicilinas/metabolismo
9.
Int J Mol Sci ; 24(13)2023 Jul 04.
Artigo em Inglês | MEDLINE | ID: mdl-37446271

RESUMO

Resistance to chemotherapy represents a persisting medical problem, ranking among main causes of chemotherapy failure and cancer mortality. There is a possibility to utilize and repurpose already existing therapeutics which were not primarily intended for oncological treatment. Overactivation of adrenergic receptors and signaling dysregulation promotes tumor progression, metastatic potential, immune system evasion, tumor angiogenesis and drug resistance. The non-selective beta-blocker propranolol, approved in infantile haemangioma treatment, has a high potential for use in cancer therapy. We analyzed the effects of propranolol and 5-fluorouracil combination on sensitive and resistant cells derived from colorectal carcinoma in monolayers, single-component and co-culture spheroids and in vivo mouse models. Our results revealed that propranolol is able to exert its effect not only in chemosensitive colorectal cells, but also in 5-fluorouracil resistant cells. Propranolol disrupts the hypoxic adaptation machinery by inhibiting HIF1α, carbonic anhydrase IX, and activates apoptosis, which may be important in the management of chemo-resistant patients. We showed that propranolol slows down the growth of xenografts formed from colorectal cancer cells, even from cells already adapted to the ß-blocker. We provide clear evidence that blockade of ß-adrenergic receptors affects essential signaling pathways modulating tumor microenvironment and thus the response to anticancer therapy. Our findings indicate that propranolol could be repurposed to serve as chemosensitizer in combined therapy aimed at disrupting homeostasis of tumor microenvironment.


Assuntos
Neoplasias , Propranolol , Humanos , Animais , Camundongos , Anidrase Carbônica IX/metabolismo , Propranolol/farmacologia , Propranolol/uso terapêutico , Microambiente Tumoral , Antígenos de Neoplasias/metabolismo , Neoplasias/patologia , Fluoruracila/farmacologia , Fluoruracila/uso terapêutico , Linhagem Celular Tumoral
10.
Neurourol Urodyn ; 42(6): 1344-1351, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37306331

RESUMO

AIMS: To determine the role of opioid and ß-adrenergic receptors in bladder underactivity induced by prolonged pudendal nerve stimulation (PNS). METHODS: In α-chloralose anesthetized cats, 30-min PNS was applied repeatedly for 3-9 times to induce poststimulation or persistent bladder underactivity. Then, naloxone (opioid receptor antagonist, 1 mg/kg, IV) or propranolol (ß-adrenergic receptor antagonist, 3 mg/kg, IV) was given to reverse the bladder underactivity. After the drug treatment, an additional 30-min PNS was applied to counteract the drug effect. Repeated cystometrograms were performed by slowly (1-2 mL/min) infusing the bladder with saline via a urethral catheter to determine the bladder underactivity and the treatment effects. RESULTS: Prolonged (2-4.5 h) PNS induced bladder underactivity evident as a large bladder capacity (169 ± 49% of control) and a reduced amplitude of bladder contraction (59 ± 17% of control). Naloxone fully reversed the bladder underactivity by reducing bladder capacity to 113 ± 58% and increasing the amplitude of bladder contraction to 104 ± 34%. After administration of naloxone an additional 30-min PNS temporarily increased the bladder capacity to the underactive bladder level (193 ± 74%) without changing the amplitude of the bladder contraction. Propranolol had no effect on bladder underactivity. CONCLUSIONS: A tonic enkephalinergic inhibitory mechanism in the CNS plays a critical role in the bladder underactivity induced by prolonged PNS, while the peripheral ß-adrenergic receptor mechanism in the detrusor is not involved. This study provides basic science evidence consistent with the clinical observation that comorbid opioid usage may contribute to voiding dysfunction in patients with Fowler's syndrome.


Assuntos
Nervo Pudendo , Doenças da Bexiga Urinária , Gatos , Animais , Bexiga Urinária , Analgésicos Opioides/farmacologia , Propranolol/farmacologia , Receptores Adrenérgicos beta , Reflexo/fisiologia , Estimulação Elétrica , Naloxona/farmacologia
11.
Sci Rep ; 13(1): 7923, 2023 05 16.
Artigo em Inglês | MEDLINE | ID: mdl-37193722

RESUMO

Drug repositioning explores the reuse of non-cancer drugs to treat tumors. In this work, we evaluated the effect of the combination of chloroquine and propranolol on colorectal and triple-negative breast cancers. Using as in vitro models the colorectal cancer cell lines HCT116, HT29, and CT26, and as triple-negative breast cancer models the 4T1, M-406, and MDA-MB-231 cell lines, we evaluated the effect of the drugs combination on the viability, apoptosis, clonogenicity, and cellular migratory capacity. To explore the in vivo effects of the combination on tumor growth and metastasis development we employed graft models in BALB/c, nude, and CBi mice. In vitro studies showed that combined treatment decreased cell viability in a dose-dependent manner and increased apoptosis. Also, we demonstrated that these drugs act synergically and that it affects clonogenicity and migration. In vivo studies indicated that this drug combination was effective on colorectal models but only partially on breast cancer. These results contributed to the search for new and safe treatments for colorectal and triple-negative carcinomas.


Assuntos
Neoplasias Colorretais , Neoplasias de Mama Triplo Negativas , Humanos , Animais , Camundongos , Neoplasias de Mama Triplo Negativas/patologia , Propranolol/farmacologia , Cloroquina/farmacologia , Cloroquina/uso terapêutico , Linhagem Celular Tumoral , Ensaios Antitumorais Modelo de Xenoenxerto , Apoptose , Neoplasias Colorretais/tratamento farmacológico , Proliferação de Células
12.
Behav Pharmacol ; 34(4): 206-212, 2023 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-37171459

RESUMO

Propranolol is the treatment of choice for infantile hemangioma. We investigated the effects of long-term propranolol use in early infancy on learning and memory later in life in mice. At three weeks of age, mice were randomly divided into six experimental groups. Groups 1 and 2 (controls) received only saline for 21 days. Groups 3 and 4 received propranolol (2.5 mg/kg) for 21 days. Groups 5 and 6 received propranolol (5 mg/kg) for 21 days. Groups 1, 3 and 5 were tested at the end of 21 days of treatment (week 6). However, groups 2, 4 and 6 received a 2-week break and then (week 8) exposed to tests. In the Morris water maze test, propranolol (2.5 and 5 mg/kg) dose-dependently increased the time spent in the target quadrant in mice at weeks 6 and 8. However, propranolol did not affect the swimming speed in both time periods. There were no significant effects of propranolol on the number of errors evaluated during the radial arm maze tests. In conclusion, long-term use of propranolol in early infancy did not disrupt the learning and memory of mice.


Assuntos
Memória , Propranolol , Camundongos , Animais , Propranolol/farmacologia , Aprendizagem em Labirinto , Natação
13.
Brain Behav Immun ; 111: 247-248, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37127226

RESUMO

In this issue of the BBI, Haldar et al. demonstrate that major surgical stress from laparotomy caused a significant increase in post-operative metastatic burden in a mouse model of cancer. They identified this metastatic outbreak was driven by a novel mechanism of direct, surgery-induced activation of the primary tumour which, if left in situ, released pro-metastatic factors (IL-6, IL-8, and VEGF). Surgical stress induced significant changes in the transcriptional programming of the primary tumor, with marked activation of NF-κB and down-regulation of IRF-1. Pharmaceutical blockade of post-operative ß-adrenergic and prostanoid signalling, by administration of propranolol and etodolac, prevented post-operative activation of the primary tumour and metastatic disease.


Assuntos
Antagonistas Adrenérgicos beta , Inibidores de Ciclo-Oxigenase 2 , Camundongos , Animais , Inibidores de Ciclo-Oxigenase 2/farmacologia , Antagonistas Adrenérgicos beta/farmacologia , Neoplasia Residual , Etodolac , Propranolol/farmacologia , NF-kappa B
14.
Eur J Pharmacol ; 949: 175718, 2023 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-37054937

RESUMO

Colorectal cancer (CRC) stands as the second leading cause of cancer-related deaths worldwide with limited available medicines. While drug repurposing comes as a promising strategy for cancer treatment, we discovered that propranolol (Prop), a non-selective ß1 and ß2 adrenergic receptor blocker, significantly inhibited the development of subcutaneous CT26 CRC and AOM/DSS-induced CRC models. The RNA-seq analysis highlighted the activated immune pathways after Prop treatment, with KEGG analysis enriched in T-cell differentiation. Routine analyses of blood revealed a decrease in neutrophil to lymphocyte ratio, a biomarker of systemic inflammation, and a prognostic indicator in the Prop-treated groups in both CRC models. Analysis of the tumor-infiltrating immune cells exhibited that Prop regressed the exhaustion of CD4+ and CD8+ T cells in the CT26-derived graft models, which was further corroborated in the AOM/DSS-induced models. Furthermore, bioinformatic analysis fitted well with the experimental data, showing that ß2 adrenergic receptor (ADRB2) was positively correlated with T-cell exhaustion signature in various tumors. The in vitro experiment showed no direct effect of Prop on CT26 cell viability, while T cells were activated with significantly-upregulated production of IFN-γ and Granzyme B. Consistently, Prop was unable to restrain CT26 tumor growth in nude mice. At last, the combination of Prop and the chemotherapeutic drug Irinotecan acted out the strongest inhibition in CT26 tumor progress. Collectively, we repurpose Prop as a promising and economical therapeutic drug for CRC treatment and highlight T-cell as its target.


Assuntos
Neoplasias Colorretais , Propranolol , Animais , Camundongos , Propranolol/farmacologia , Propranolol/uso terapêutico , Irinotecano/farmacologia , Irinotecano/uso terapêutico , Linfócitos T CD8-Positivos , Camundongos Nus , Antagonistas Adrenérgicos beta/farmacologia , Antagonistas Adrenérgicos beta/uso terapêutico , Neoplasias Colorretais/patologia , Linhagem Celular Tumoral
15.
Vet Comp Oncol ; 21(2): 159-165, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-36876492

RESUMO

Recent studies have highlighted a key role played by the sympathetic nervous system (SNS) and adrenergic stress in mediating immune suppression associated with chronic inflammation in cancer and other diseases. The connection between chronic SNS activation, adrenergic stress and immune suppression is linked in part to the ability of catecholamines to stimulate the bone marrow release and differentiation of myeloid-derived suppressor cells (MDSC). Rodent model studies have revealed an important role for ß-adrenergic receptor signalling in suppression of cancer immunity in mice subjected to chronic stresses, including thermal stress. Importantly, therapeutic blockade of beta-adrenergic responses by drugs such as propranolol can partially reverse the generation and differentiation of MDSC, and partly restore tumour immunity. Clinical trials in both humans and dogs with cancer have demonstrated that propranolol blockade can improve responses to radiation therapy, cancer vaccines and immune checkpoint inhibitors. Thus, the SNS stress response has become an important new target to relieve immune suppression in cancer and other chronic inflammatory conditions.


Assuntos
Doenças do Cão , Células Supressoras Mieloides , Neoplasias , Humanos , Cães , Camundongos , Animais , Propranolol/farmacologia , Adrenérgicos , Doenças do Cão/terapia , Imunoterapia/veterinária , Neoplasias/terapia , Neoplasias/veterinária
16.
Int J Mol Sci ; 24(5)2023 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-36902020

RESUMO

The SARS-CoV-2 pandemic highlighted the need for broad-spectrum antivirals to increase our preparedness. Patients often require treatment by the time that blocking virus replication is less effective. Therefore, therapy should not only aim to inhibit the virus, but also to suppress pathogenic host responses, e.g., leading to microvascular changes and pulmonary damage. Clinical studies have previously linked SARS-CoV-2 infection to pathogenic intussusceptive angiogenesis in the lungs, involving the upregulation of angiogenic factors such as ANGPTL4. The ß-blocker propranolol is used to suppress aberrant ANGPTL4 expression in the treatment of hemangiomas. Therefore, we investigated the effect of propranolol on SARS-CoV-2 infection and the expression of ANGPTL4. SARS-CoV-2 upregulated ANGPTL4 in endothelial and other cells, which could be suppressed with R-propranolol. The compound also inhibited the replication of SARS-CoV-2 in Vero-E6 cells and reduced the viral load by up to ~2 logs in various cell lines and primary human airway epithelial cultures. R-propranolol was as effective as S-propranolol but lacks the latter's undesired ß-blocker activity. R-propranolol also inhibited SARS-CoV and MERS-CoV. It inhibited a post-entry step of the replication cycle, likely via host factors. The broad-spectrum antiviral effect and suppression of factors involved in pathogenic angiogenesis make R-propranolol an interesting molecule to further explore for the treatment of coronavirus infections.


Assuntos
COVID-19 , Animais , Chlorocebus aethiops , Humanos , Propranolol/farmacologia , SARS-CoV-2 , Células Vero , Linhagem Celular , Antivirais/farmacologia , Replicação Viral
17.
Eye (Lond) ; 37(14): 2909-2914, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-36782058

RESUMO

BACKGROUND/OBJECTIVES: Tremor and expertise are potentially influenced variables in vitreoretinal surgery. We investigated whether surgeon experience impacts the association of microsurgical performance with caffeine and ß-blockers weight-adjusted intake. SUBJECTS/METHODS: Novice and senior surgeons (<2 and >10 practice years, respectively) were recruited in this self-controlled, cross-sectional study. A simulator's task sequence was repeated over 2 days, 30 min after the following exposures: day 1, placebo, 2.5 mg/kg caffeine, 5.0 mg/kg caffeine, and 0.6 mg/kg propranolol; and day 2, placebo, 0.2 mg/kg propranolol, 0.6 mg/kg propranolol, and 5.0 mg/kg caffeine. Outcomes were total score (0-700, worst-best), simulation time (minutes), intraocular trajectory (centimeters), and tremor-specific score (0-100, worst-best). RESULTS: We recruited 15 novices (9 men [60%], 1.33 ± 0.49 practice years) and 11 seniors (8 men [72.7%], 16.00 ± 4.24 practice years). Novices performed worse after 2.5 mg/kg caffeine and improved following 0.2 mg/kg propranolol in total score (557 vs. 617, p = 0.009), trajectory (229.86 vs. 208.07, p = 0.048), time (14.9 vs. 12.7, p = 0.048), and tremor-score (55 vs. 75, p = 0.009). Surgical performance improved with propranolol post-caffeine but remained worse than 0.2 mg/kg propranolol in total score (570 vs. 617, p = 0.014), trajectory (226.59 vs. 208.07, p = 0.033), and tremor-score (50 vs. 75, p = 0.029). Seniors' tremor-score was lower after 2.5 mg/kg caffeine compared to 0.2 mg/kg propranolol (8 vs. 37, p = 0.015). Tremor-score following propranolol post-caffeine remained inferior to 0.6 mg/kg propranolol alone (17 vs. 38, p = 0.012). CONCLUSION: While caffeine and propranolol were associated with performance changes among novices, only tremor was affected in seniors, without dexterity changes. The pharmacologic exposure impact on surgical dexterity seems to be offset by increased experience.


Assuntos
Cafeína , Cirurgiões , Humanos , Masculino , Antagonistas Adrenérgicos beta/farmacologia , Antagonistas Adrenérgicos beta/uso terapêutico , Cafeína/farmacologia , Estudos Transversais , Método Duplo-Cego , Propranolol/farmacologia , Propranolol/uso terapêutico , Retina , Tremor/tratamento farmacológico
18.
Brain Behav Immun ; 110: 1-12, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36796704

RESUMO

The immune system plays an important role in controlling epithelial ovarian cancer (EOC). EOC is considered to be a "cold tumour," a tumour that has not triggered a strong response by the immune system. However, tumour infiltrating lymphocytes (TILs) and the expression of programmed cell death ligand (PD-L1) are used as prognostic indicators in EOC. Immunotherapy such as PD-(L)1 inhibitors have shown limited benefit in EOC. Since the immune system is affected by behavioural stress and the beta-adrenergic signalling pathway, this study aimed to explore the impact of propranolol (PRO), a beta-blocker, on anti-tumour immunity in both in vitro and in vivo EOC models. Noradrenaline (NA), an adrenergic agonist, did not directly regulate PD-L1 expression but PD-L1 was significantly upregulated by IFN-γ in EOC cell lines. IFN-γ also increased PD-L1 on extracellular vesicles (EVs) released by ID8 cells. PRO significantly decreased IFN-γ levels in primary immune cells activated ex vivo and showed increased viability of the CD8+ cell population in an EV-immune cell co-incubation. In addition, PRO reverted PD-L1 upregulation and significantly decreased IL-10 levels in an immune-cancer cell co-culture. Chronic behavioural stress increased metastasis in mice while PRO monotherapy and the combo of PRO and PD-(L)1 inhibitor significantly decreased stress-induced metastasis. The combined therapy also reduced tumour weight compared to the cancer control group and induced anti-tumour T-cell responses with significant CD8 expression in tumour tissues. In conclusion, PRO showed a modulation of the cancer immune response by decreasing IFN-γ production and, in turn, IFN-γ-mediated PD-L1 overexpression. The combined therapy of PRO and PD-(L)1 inhibitor decreased metastasis and improved anti-tumour immunity offering a promising new therapy.


Assuntos
Antígeno B7-H1 , Neoplasias Ovarianas , Propranolol , Animais , Feminino , Humanos , Camundongos , Antígeno B7-H1/imunologia , Antígeno B7-H1/metabolismo , Linfócitos T CD8-Positivos , Terapia de Imunossupressão , Interferon gama/metabolismo , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/imunologia , Neoplasias Ovarianas/metabolismo , Propranolol/farmacologia
19.
Zhongguo Zhong Yao Za Zhi ; 48(1): 220-225, 2023 Jan.
Artigo em Chinês | MEDLINE | ID: mdl-36725274

RESUMO

This paper aimed to investigate the effect of total flavonoids of buckwheat flower and leaf on myocardial cell apoptosis and Wnt/ß-catenin/peroxisome proliferator-activated receptor γ(PPARγ) pathway in arrhythmic rats. SD rats were randomly divided into a control group, a model group, a low-dose(20 mg·kg~(-1)) group of total flavonoids of buckwheat flower and leaf, a medium-dose(40 mg·kg~(-1)) group of total flavonoids of buckwheat flower and leaf, a high-dose(80 mg·kg~(-1)) group of total flavonoids of buckwheat flower and leaf, a propranolol hydrochloride(2 mg·kg~(-1)) group, with 12 rats in each group. Except the control group, rats in other groups were prepared as models of arrhythmia by sublingual injection of 1 mL·kg~(-1) of 0.002% aconitine. After grouping and intervention with drugs, the arrhythmia, myocardial cells apoptosis, myocardial tissue glutathione peroxidase(GSH-Px), catalase(CAT), malondialdehyde(MDA), serum interleukin-6(IL-6), prostaglandin E2(PGE2) levels, myocardial tissue apoptosis, and Wnt/ß-catenin/PPARγ pathway-related protein expression of rats in each group were measured. As compared with the control group, the arrhythmia score, the number of ventricular premature beats, ventricular fibrillation duration, myocardial cell apoptosis rate, MDA levels in myocardial tissues, serum IL-6 and PGE2 levels, Bax in myocardial tissues, and Wnt1 and ß-catenin protein expression levels increased significantly in the model group, whereas the GSH-Px and CAT levels, and Bcl-2 and PPARγ protein expression levels in myocardial tissues reduced significantly. As compared with the model group, the arrhythmia score, the number of ventricular premature beats, ventricular fibrillation duration, myocardial cell apoptosis rate, MDA leve in myocardial tissues, serum IL-6 and PGE2 levels, Bax in myocardial tissues, and Wnt1 and ß-catenin protein expression levels reduced in the drug intervention groups, whereas the GSH-Px and CAT levels and Bcl-2 and PPARγ protein expression levels in myocardial tissues increased. The groups of total flavonoids of buckwheat flower and leaf were in a dose-dependent manner. There was no significant difference in the levels of each index in rats between the propranolol hydrochloride group and the high-dose group of total flavonoids of buckwheat flower and leaf. The total flavonoids of buckwheat flower and leaf inhibit the activation of Wnt/ß-catenin pathway, up-regulate the expression of PPARγ, reduce oxidative stress and inflammatory damage in myocardial tissues of arrhythmic rats, reduce myocardial cell apoptosis, and improve the symptoms of arrhythmia in rats.


Assuntos
Fagopyrum , PPAR gama , Ratos , Animais , PPAR gama/metabolismo , Fagopyrum/genética , Ratos Sprague-Dawley , Proteína X Associada a bcl-2 , beta Catenina/genética , beta Catenina/metabolismo , Interleucina-6 , Flavonoides/farmacologia , Propranolol/farmacologia , Fibrilação Ventricular , Dinoprostona , Via de Sinalização Wnt , Folhas de Planta/metabolismo , Flores/metabolismo , Apoptose , Complexos Cardíacos Prematuros
20.
Bratisl Lek Listy ; 124(4): 292-303, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36598324

RESUMO

OBJECTIVE: Propranolol (PRO) has been recently discovered to possess anti-tumorigenic effects in cancer patients. So, we aimed to enlighten the in vitro effects of propranolol in A549 lung cancer cells and BEAS2B nontumoral lung cells. METHODS: The gene expression levels of apoptotic proteins; caspases 3, 8, and 9 (CASP3, 8, 9), apoptosis inducing factor (AIF), and DNA damage inducible transcript 3 (DDIT3) and cell cycle regulatory proteins; WEE1 G2 checkpoint kinase (WEE1) and cyclin dependent kinase inhibitor 1A (CDKN1A) were analyzed with quantitative reverse-transcription PCR to assess the effect of PRO on A549 tumor and BEAS2B nontumoral cells. The protein levels of caspase 3 and AIF1 were detected with Western blot. RESULTS: PRO exerted its anti-tumorigenic effects against A549 cells by arresting cell cycle via CDNK1A and by inducing apoptosis via caspase-dependent (CASP3) and -independent pathways (AIF, DDIT3). As to nontumoral BEAS2B cells, PRO decreased the cell viability at a lesser extent compared to tumoral cells. In contrast to tumor cells, PRO reduced the protein levels of CASP3 and AIF1. Notably, at 48th hour of PRO treatment, we observed a sustained expression of elevated DDIT3 mRNA levels at 24h in BEAS2B cells unlike in A549 cells. CONCLUSION: We suggest that DDIT3 and CDKN1A play a critical role during cell fate decision after PRO treatment by protecting nontumoral cells against apoptosis and by triggering apoptosis in tumor cells. The selective action mechanism of PRO with less cytotoxicity in nontumoral lung cells puts it forward as a promising adjuvant agent in lung cancer therapy (Tab. 1, Fig. 4, Ref. 50). Text in PDF www.elis.sk Keywords: propranolol, BEAS2B, A549, lung cancer, apoptosis, DDIT3.


Assuntos
Neoplasias Pulmonares , Propranolol , Humanos , Caspase 3/metabolismo , Propranolol/farmacologia , Linhagem Celular Tumoral , Neoplasias Pulmonares/patologia , Apoptose , Proliferação de Células
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA