Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 1.156
Filtrar
Más filtros

Publication year range
1.
Nature ; 623(7989): 992-1000, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37968397

RESUMEN

Cerebral oedema is associated with morbidity and mortality after traumatic brain injury (TBI)1. Noradrenaline levels are increased after TBI2-4, and the amplitude of the increase in noradrenaline predicts both the extent of injury5 and the likelihood of mortality6. Glymphatic impairment is both a feature of and a contributor to brain injury7,8, but its relationship with the injury-associated surge in noradrenaline is unclear. Here we report that acute post-traumatic oedema results from a suppression of glymphatic and lymphatic fluid flow that occurs in response to excessive systemic release of noradrenaline. This post-TBI adrenergic storm was associated with reduced contractility of cervical lymphatic vessels, consistent with diminished return of glymphatic and lymphatic fluid to the systemic circulation. Accordingly, pan-adrenergic receptor inhibition normalized central venous pressure and partly restored glymphatic and cervical lymphatic flow in a mouse model of TBI, and these actions led to substantially reduced brain oedema and improved functional outcomes. Furthermore, post-traumatic inhibition of adrenergic signalling boosted lymphatic export of cellular debris from the traumatic lesion, substantially reducing secondary inflammation and accumulation of phosphorylated tau. These observations suggest that targeting the noradrenergic control of central glymphatic flow may offer a therapeutic approach for treating acute TBI.


Asunto(s)
Edema Encefálico , Lesiones Traumáticas del Encéfalo , Sistema Glinfático , Norepinefrina , Animales , Ratones , Antagonistas Adrenérgicos/farmacología , Antagonistas Adrenérgicos/uso terapéutico , Edema Encefálico/complicaciones , Edema Encefálico/tratamiento farmacológico , Edema Encefálico/metabolismo , Edema Encefálico/prevención & control , Lesiones Traumáticas del Encéfalo/complicaciones , Lesiones Traumáticas del Encéfalo/tratamiento farmacológico , Lesiones Traumáticas del Encéfalo/metabolismo , Modelos Animales de Enfermedad , Sistema Glinfático/efectos de los fármacos , Sistema Glinfático/metabolismo , Inflamación/complicaciones , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Inflamación/prevención & control , Vasos Linfáticos/metabolismo , Norepinefrina/metabolismo , Fosforilación , Receptores Adrenérgicos/metabolismo
2.
Nature ; 578(7795): 449-454, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-32051587

RESUMEN

The solid tumour microenvironment includes nerve fibres that arise from the peripheral nervous system1,2. Recent work indicates that newly formed adrenergic nerve fibres promote tumour growth, but the origin of these nerves and the mechanism of their inception are unknown1,3. Here, by comparing the transcriptomes of cancer-associated trigeminal sensory neurons with those of endogenous neurons in mouse models of oral cancer, we identified an adrenergic differentiation signature. We show that loss of TP53 leads to adrenergic transdifferentiation of tumour-associated sensory nerves through loss of the microRNA miR-34a. Tumour growth was inhibited by sensory denervation or pharmacological blockade of adrenergic receptors, but not by chemical sympathectomy of pre-existing adrenergic nerves. A retrospective analysis of samples from oral cancer revealed that p53 status was associated with nerve density, which was in turn associated with poor clinical outcomes. This crosstalk between cancer cells and neurons represents mechanism by which tumour-associated neurons are reprogrammed towards an adrenergic phenotype that can stimulate tumour progression, and is a potential target for anticancer therapy.


Asunto(s)
Neuronas Adrenérgicas/patología , Transdiferenciación Celular , Reprogramación Celular , Neoplasias de la Boca/patología , Células Receptoras Sensoriales/patología , Proteína p53 Supresora de Tumor/deficiencia , Antagonistas Adrenérgicos/farmacología , Antagonistas Adrenérgicos/uso terapéutico , Animales , División Celular , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , MicroARNs/genética , Neoplasias de la Boca/tratamiento farmacológico , Neoplasias de la Boca/genética , Neoplasias de la Boca/metabolismo , Fibras Nerviosas/patología , Neuritas/patología , Receptores Adrenérgicos/metabolismo , Estudios Retrospectivos , Microambiente Tumoral , Proteína p53 Supresora de Tumor/genética , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Handb Exp Pharmacol ; 285: 617-637, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38177400

RESUMEN

In this chapter, we review how ligands, both agonists and antagonists, for the major classes of adrenoreceptors, are utilized in acute care clinical settings. Adrenergic ligands exert their effects by interacting with the three major classes of adrenoceptors. Adrenoceptor agonists and antagonists have important applications, ranging from treatment of hypotension to asthma, and have proven to be extremely useful in a variety of clinical settings of acute care from the operating room to the critical care environment. Continued research interpreting the mechanisms of adrenoreceptors may help the discovery of new drugs with more desirable clinical profiles.


Asunto(s)
Receptores Adrenérgicos , Humanos , Ligandos , Receptores Adrenérgicos/metabolismo , Receptores Adrenérgicos/efectos de los fármacos , Antagonistas Adrenérgicos/farmacología , Antagonistas Adrenérgicos/uso terapéutico , Animales , Cuidados Críticos , Agonistas Adrenérgicos/farmacología , Agonistas Adrenérgicos/uso terapéutico
4.
FASEB J ; 36(3): e22057, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35133020

RESUMEN

Non-healing wounds are a major medical challenge, affecting over 6.5 million people in the US alone, with associated healthcare costs of about $16 billion annually. They can result in prolonged hospitalizations, work loss, disability, poor quality of life, and in diabetic patients with foot ulcers, amputation of the affected limb in 25% of patients. Though chronic ulcers may arise from different underlying diseases, the unifying feature is chronic infection, driving persistent inflammation that prolongs the healing process. One of the most frequently cultured or genetically identified pathogens in skin wounds is Pseudomonas aeruginosa. This species avidly forms biofilms in the wound that impede bacterial eradication by the host's immune mechanisms and limit efficacy of systemic antibiotics. Thus, non-antibiotic approaches to limit the growth and biofilm formation of this wound pathogen would be an advance in the treatment of chronic wounds. Prior work has demonstrated that the growth of other microbial species can be modulated by catecholamine agonists and antagonists of the adrenergic receptors (ARs). Here, we demonstrate that not only can the growth of this common wound pathogen be modulated by catecholamines, but also that the beta-AR antagonists can significantly decrease their growth, and importantly, limit their ability to form biofilms. These findings suggest that beta adrenergic antagonists may have a therapeutic role in the treatment of chronic skin wounds.


Asunto(s)
Antagonistas Adrenérgicos/farmacología , Biopelículas , Epinefrina/farmacología , Pseudomonas aeruginosa/efectos de los fármacos , Timolol/farmacología , Cicatrización de Heridas , Antagonistas Adrenérgicos/uso terapéutico , Epinefrina/uso terapéutico , Humanos , Infecciones por Pseudomonas/tratamiento farmacológico , Infecciones por Pseudomonas/microbiología , Pseudomonas aeruginosa/patogenicidad , Enfermedades Cutáneas Bacterianas/tratamiento farmacológico , Enfermedades Cutáneas Bacterianas/microbiología , Timolol/uso terapéutico
5.
Int J Neurosci ; 133(5): 496-504, 2023 May.
Artículo en Inglés | MEDLINE | ID: mdl-33998961

RESUMEN

Aim: Low frequency stimulation (LFS) inhibits neuronal hyperexcitability following epileptic activity. However, knowledge about LFS' inhibitory mechanisms is lacking. Here, α1 and α2 adrenergic receptors' roles in mediating LFS inhibitory action on high-K+ induced epileptiform activity (EA) was examined in rat hippocampal slices.Materials and methods: LFS (1 Hz, 900 pulses) was applied to the Schaffer collaterals. Whole-cell, patch clamp recording was used to measure changes in CA1 pyramidal neurons' excitability. By applying high-K+ on hippocampal slices, EA was induced, and neuronal excitability increased.Results: When administered at the beginning of EA, LFS reduced neuronal excitability. In the presence of prazosin (10 µM, an α1 adrenergic receptor antagonist) and yohimbine (5 µM, an α2 adrenergic receptor antagonist), LFS' typically has a restorative impact on EA-induced membrane potential hyperpolarization and spike firing frequency, but this effect was reduced after high-K+ washout; These antagonists did not have a significant effect on LFS' inhibitory action on spike firing during EA.Conclusion: These findings suggest that LFS' anticonvulsant effect, on neuronal hyperexcitability following high-K+ EA, may be mediated partly through α adrenergic receptors in hippocampal slices.


Asunto(s)
Epilepsia , Receptores Adrenérgicos alfa , Ratas , Animales , Ratas Wistar , Hipocampo , Epilepsia/terapia , Receptores Adrenérgicos alfa 2 , Antagonistas Adrenérgicos/farmacología , Estimulación Eléctrica
6.
Neurochem Res ; 47(11): 3440-3453, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-35945306

RESUMEN

Cognitive impairment is a common central nervous system complication that occurs following surgery or organs damage outside the nervous system. Neuroinflammation plays a key role in the molecular mechanisms of cognitive impairment. Dexmedetomidine alleviates neuroinflammation and reduces cognitive dysfunction incidence; however, the mechanism by which dexmedetomidine alleviates cognitive dysfunction remains unclear. This study evaluated the effect of dexmedetomidine on attenuation of early cognitive impairment induced by intestinal ischemia-reperfusion in mice and examined whether the locus coeruleus norepinephrine (LCNE) system participates in the anti-inflammatory effect of dexmedetomidine. The superior mesenteric artery was clamped for 45 min to induce intestinal ischemia reperfusion injury. Dexmedetomidine alone or combined with DSP-4, a selective locus coeruleus noradrenergic neurotoxin, was used for pretreatment. Postoperative cognition was assessed using the Morris water maze. Serum and hippocampal levels of IL-1ß, TNF-α, norepinephrine (NE), and malondialdehyde (MDA) were assessed by enzyme-linked immunosorbent assay. Immunofluorescence, immunohistochemistry, and hematoxylin and eosin staining were used to evaluate the expression of tyrosine hydroxylase (TH) in the locus coeruleus, hippocampal microglia, and intestinal injury. Pretreatment with dexmedetomidine alleviated intestinal injury and decreased the serum and hippocampal levels of NE, IL-1ß, TNF-α, and MDA at 24 h after intestinal ischemia reperfusion, decreased TH-positive neurons in the locus coeruleus, and ameliorated cognitive impairment. Similarly, DSP-4 pre-treatment alleviated neuroinflammation and improved cognitive function. Furthermore, α2-adrenergic receptor antagonist atipamezole or yohimbine administration diminished the neuroprotective effects and improved cognitive function with dexmedetomidine. Therefore, dexmedetomidine attenuated early cognitive dysfunction induced by intestinal ischemia-reperfusion injury in mice, which may be related to its anti-inflammatory effects through the LCNE system.


Asunto(s)
Disfunción Cognitiva , Dexmedetomidina , Fármacos Neuroprotectores , Daño por Reperfusión , Antagonistas Adrenérgicos/uso terapéutico , Animales , Antiinflamatorios/farmacología , Antiinflamatorios/uso terapéutico , Bencilaminas , Disfunción Cognitiva/tratamiento farmacológico , Disfunción Cognitiva/etiología , Dexmedetomidina/farmacología , Dexmedetomidina/uso terapéutico , Eosina Amarillenta-(YS)/uso terapéutico , Hematoxilina/uso terapéutico , Isquemia , Locus Coeruleus/metabolismo , Malondialdehído , Ratones , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/uso terapéutico , Neurotoxinas , Norepinefrina , Reperfusión , Daño por Reperfusión/complicaciones , Daño por Reperfusión/tratamiento farmacológico , Factor de Necrosis Tumoral alfa/metabolismo , Tirosina 3-Monooxigenasa/metabolismo , Yohimbina/uso terapéutico
7.
Mol Biol Rep ; 49(10): 9565-9573, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-35970968

RESUMEN

BACKGROUND: The demand for environmentally friendly and cost-effective plant-based products for the development of cancer therapeutics has been increasing. Yohimbine (α2-adrenergic receptor antagonist) is a stimulant and aphrodisiac used to improve erectile dysfunction. In this study, we aimed to evaluate the anticancer potential of yohimbine in drug-resistant oral cancer KB-ChR-8-5 cells using different biomolecular techniques. METHODS: We estimated the anticancer efficacy of yohimbine using different assays, such as MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) cell cytotoxicity, cell morphology, cell apoptosis, reactive oxygen species (ROS) formation, and modulation in the mitochondrial membrane potential (MMP). RESULTS: Yohimbine showed a dose-dependent increase in cytotoxicity with a 50% inhibitory concentration (IC50) of 44 µM against KB-ChR-8-5 cancer cell lines. Yohimbine treatment at 40 µM and 50 µM resulted in a considerable change in cell morphology, including shrinkage, detachment, membrane blebbing, and deformed shape. Moreover, at the dose of IC50 and above, a significant induction was observed in the generation of ROS and depolarization of MMP. The possible mechanisms of action of yohimbine underlying the dose-dependent increase in cytotoxicity may be due to the induction of apoptosis, ROS generation, and modulation of MMP. CONCLUSION: Overall, yohimbine showed a significant anticancer potential against drug-resistant oral cancer KB-ChR-8-5 cells. Our study suggests that besides being an aphrodisiac, yohimbine can be used as a drug repurposing agent. However, more research is required in different in vitro and in vivo models to confirm the feasibility of yohimbine in clinics.


Asunto(s)
Afrodisíacos , Neoplasias de la Boca , Antagonistas Adrenérgicos/farmacología , Afrodisíacos/farmacología , Apoptosis , Línea Celular Tumoral , Humanos , Potencial de la Membrana Mitocondrial , Neoplasias de la Boca/tratamiento farmacológico , Especies Reactivas de Oxígeno/metabolismo , Yohimbina/farmacología
8.
Proc Natl Acad Sci U S A ; 116(22): 11010-11019, 2019 05 28.
Artículo en Inglés | MEDLINE | ID: mdl-31097598

RESUMEN

Spontaneous waves of cortical spreading depolarization (CSD) are induced in the setting of acute focal ischemia. CSD is linked to a sharp increase of extracellular K+ that induces a long-lasting suppression of neural activity. Furthermore, CSD induces secondary irreversible damage in the ischemic brain, suggesting that K+ homeostasis might constitute a therapeutic strategy in ischemic stroke. Here we report that adrenergic receptor (AdR) antagonism accelerates normalization of extracellular K+, resulting in faster recovery of neural activity after photothrombotic stroke. Remarkably, systemic adrenergic blockade before or after stroke facilitated functional motor recovery and reduced infarct volume, paralleling the preservation of the water channel aquaporin-4 in astrocytes. Our observations suggest that AdR blockers promote cerebrospinal fluid exchange and rapid extracellular K+ clearance, representing a potent potential intervention for acute stroke.


Asunto(s)
Antagonistas Adrenérgicos/farmacología , Isquemia Encefálica/metabolismo , Neuroprotección/efectos de los fármacos , Accidente Cerebrovascular/metabolismo , Animales , Acuaporina 4/metabolismo , Astrocitos/metabolismo , Modelos Animales de Enfermedad , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Potasio/metabolismo
9.
Int J Mol Sci ; 24(1)2022 Dec 24.
Artículo en Inglés | MEDLINE | ID: mdl-36613736

RESUMEN

Cardiovascular diseases remain one of the leading causes of death worldwide. Unfortunately, the available pharmacotherapeutic options have limited effectiveness. Therefore, developing new drug candidates remains very important. We selected six novel arylpiperazine alkyl derivatives of salicylamide to investigate their cardiovascular effects. Having in mind the beneficial role of α1-adrenergic receptors in restoring sinus rhythm and regulating blood pressure, first, using radioligand binding assays, we evaluated the affinity of the tested compounds for α-adrenergic receptors. Our experiments revealed their high to moderate affinity for α1- but not α2-adrenoceptors. Next, we aimed to determine the antiarrhythmic potential of novel derivatives in rat models of arrhythmia induced by adrenaline, calcium chloride, or aconitine. All compounds showed potent prophylactic antiarrhythmic activity in the adrenaline-induced arrhythmia model and no effects in calcium chloride- or aconitine-induced arrhythmias. Moreover, the tested compounds demonstrated therapeutic antiarrhythmic activity, restoring a normal sinus rhythm immediately after the administration of the arrhythmogen adrenaline. Notably, none of the tested derivatives affected the normal electrocardiogram (ECG) parameters in rodents, which excludes their proarrhythmic potential. Finally, all tested compounds decreased blood pressure in normotensive rats and reversed the pressor response to methoxamine, suggesting that their hypotensive mechanism of action is connected with the blockade of α1-adrenoceptors. Our results confirm the antiarrhythmic and hypotensive activities of novel arylpiperazine derivatives and encourage their further investigation as model structures for potential drugs.


Asunto(s)
Aconitina , Antihipertensivos , Animales , Ratas , Aconitina/toxicidad , Antagonistas Adrenérgicos , Antiarrítmicos/uso terapéutico , Antihipertensivos/farmacología , Arritmias Cardíacas/inducido químicamente , Arritmias Cardíacas/tratamiento farmacológico , Arritmias Cardíacas/prevención & control , Cloruro de Calcio , Epinefrina/farmacología , Epinefrina/uso terapéutico , Ratas Wistar , Receptores Adrenérgicos alfa , Receptores Adrenérgicos alfa 1/metabolismo , Salicilamidas
10.
Int J Mol Sci ; 23(18)2022 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-36142287

RESUMEN

Heart rhythm abnormalities are a cause of many deaths worldwide. Unfortunately, the available antiarrhythmic drugs show limited efficacy and proarrhythmic potential. Thus, efforts should be made to search for new, more effective, and safer pharmacotherapies. Several studies suggested that blocking the α1-adrenoceptors could restore normal heart rhythm in arrhythmia. In this study, we aimed to assess the antiarrhythmic potential of S-61 and S-73, two novel pyrrolidin-2-one derivatives with high affinity for α1-adrenergic receptors. First, using radioligand binding studies, we demonstrated that S-61 and S-73 did not bind with ß1-adrenoceptors. Next, we assessed whether S-61 and S-73 could protect rats against arrhythmia in adrenaline-, calcium chloride- and aconitine-induced arrhythmia models. Both compounds showed potent prophylactic antiarrhythmic properties in the adrenaline-induced arrhythmia model, but the effect of S-61 was more pronounced. None of the compounds displayed antiarrhythmic effects in calcium chloride- or aconitine-induced arrhythmia models. Interestingly, both derivatives revealed therapeutic antiarrhythmic activity in the adrenaline-induced arrhythmia, diminishing heart rhythm irregularities. Neither S-61 nor S-73 showed proarrhythmic potential in rats. Finally, the compounds decreased blood pressure in rodents. The hypotensive effects were not observed after coadministration with methoxamine, which suggests the α1-adrenolytic properties of both compounds. Our results confirm that pyrrolidin-2-one derivatives possess potent antiarrhythmic properties. Given the promising results of our experiments, further studies on pyrrolidin-2-one derivatives might result in the development of a new class of antiarrhythmic drugs.


Asunto(s)
Antiarrítmicos , Antihipertensivos , Aconitina/efectos adversos , Antagonistas Adrenérgicos , Animales , Antiarrítmicos/farmacología , Antiarrítmicos/uso terapéutico , Antihipertensivos/farmacología , Arritmias Cardíacas/inducido químicamente , Arritmias Cardíacas/tratamiento farmacológico , Arritmias Cardíacas/prevención & control , Cloruro de Calcio , Epinefrina/farmacología , Epinefrina/uso terapéutico , Metoxamina , Pirrolidinonas/farmacología , Ratas , Ratas Wistar , Receptores Adrenérgicos alfa 1 , Receptores Adrenérgicos beta 1
11.
Int J Mol Sci ; 23(21)2022 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-36361620

RESUMEN

It is well-recognized that cigarette smoking is a primary risk factor in the development of non-small cell lung cancer (NSCLC), known to account for ~80% of all lung cancers with nicotine recognized as the major addictive component. In investigating the effect of nicotine, brain-derived neurotrophic factor (BDNF), and the ß-adrenergic receptor blocker, propranolol, on sensitivity of NSCLC cell lines, A549 and H1299, to cisplatin, we found increased cell viability, and enhanced cisplatin resistance with nicotine and/or BDNF treatment while opposite effects were found upon treatment with propranolol. Cell treatment with epinephrine or nicotine led to EGFR and IGF-1R activation, effects opposite to those found with propranolol. Blocking EGFR and IGF-1R activation increased cell sensitivity to cisplatin in both cell lines. PI3K and AKT activities were upregulated by nicotine or BDNF and downregulated by cell treatment with inhibitors against EGFR and IGF-1R and by propranolol. Apoptosis and cell sensitivity to cisplatin increased upon co-treatment of cells with cisplatin and inhibitors against PI3K or AKT. Our findings shed light on an interplay between nicotine, BDNF, and ß-Adrenergic receptor signaling in regulating survival of lung cancer cells and chemoresistance which can likely expand therapeutic opportunities that target this regulatory network in the future.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Cisplatino/farmacología , Cisplatino/uso terapéutico , Nicotina/farmacología , Factor Neurotrófico Derivado del Encéfalo/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores ErbB/metabolismo , Propranolol/farmacología , Propranolol/uso terapéutico , Antagonistas Adrenérgicos/farmacología , Resistencia a Antineoplásicos , Fosfatidilinositol 3-Quinasas/metabolismo , Receptores Adrenérgicos beta , Línea Celular Tumoral
12.
Ter Arkh ; 94(2): 200-208, 2022 Feb 15.
Artículo en Ruso | MEDLINE | ID: mdl-36286743

RESUMEN

AIM: To study the polymorphic markers CYP2D6*4 (G1846A, rs3892097), CYP2D6*6 (T1707del, rs5030655), CYP2D6*10 (C100T, rs1065852), CYP2D6*41 (G2988A, rs28371725) and CYP2D6*3 (A2549del, rs4986774) role in treatment optimization of portal hypertension with propranolol in patients with liver cirrhosis (LC). MATERIALS AND METHODS: The study included 60 patients with LC who received propranolol therapy at a daily dose of 30 mg for 14 days. The efficacy of treatment was assessed by ultrasonography measuring the linear blood flow velocity of portal vein. Genotyping of CYP2D6*4, CYP2D6*6, CYP2D6*10, CYP2D6*41 and CYP2D6*3 was carried out by real-time polymerase chain reaction. Evaluation of the CYP2D6 activity was carried out by determining the ratio of pinoline and its metabolite concentration in morning urine using high performance liquid chromatography with mass spectrometry. RESULTS: Positive hemodynamics in the form of any increase in the mean linear blood flow velocity of the portal vein compared to baseline was observed in 41 patients. Portal vein mean linear blood flow rate increased from 10.43.9 to 14.74.3 cm/s (p0.001). Of these, 29 patients showed an increase in this indicator by 20% from the initial one with a dynamic of 5.5 cm/s (p0.001). The regression analysis constructed by us revealed the presence of a statistically significant effect of the CYP2D6 gene polymorphic marker G1846A carriage on the propranolol therapeutic effect (p0.05). There was no statistically significant effect of polymorphic markers T1707del, C100T, G2988A, and A2549del of the CYP2D6 gene (p0.05). No convincing reliable dependence of CYP2D6 activity on the severity of LC was revealed (p0.05). CONCLUSION: An association was found between CYP2D6 gene polymorphic marker G1846A carriage and the hemodynamic effect of propranolol in patients with LC of the Russian population. There is a more significant positive dynamics of manifestations of portal hypertension on the background of propranolol therapy in carriers of the homozygous GG CYP2D6*4 genotype, in contrast to patients with the heterozygous GA genotype. Based on the results of the study, an algorithm has been developed for personalizing the treatment of patients with LC with nonselective b-adrenergic blockers using the method of CYP2D6 genotyping. Carriage of polymorphic markers T1707del, C100T, G2988A and A2549del gene CYP2D6 does not affect the effectiveness of propranolol therapy in patients with LC.


Asunto(s)
Hipertensión Portal , Propranolol , Humanos , Antagonistas Adrenérgicos/uso terapéutico , Citocromo P-450 CYP2D6/genética , Citocromo P-450 CYP2D6/uso terapéutico , Hemodinámica , Hipertensión Portal/diagnóstico , Hipertensión Portal/tratamiento farmacológico , Hipertensión Portal/etiología , Cirrosis Hepática/complicaciones , Cirrosis Hepática/diagnóstico , Cirrosis Hepática/tratamiento farmacológico , Propranolol/uso terapéutico , Polimorfismo Genético
13.
Pflugers Arch ; 473(11): 1723-1735, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34510286

RESUMEN

Acute normobaric hypoxia may induce pulmonary injury with edema (PE) and inflammation. Hypoxia is accompanied by sympathetic activation. As both acute hypoxia and high plasma catecholamine levels may elicit PE, we had originally expected that adrenergic blockade may attenuate the severity of hypoxic pulmonary injury. In particular, we investigated whether administration of drugs with reduced fluid load would be beneficial with respect to both cardiocirculatory and pulmonary functions in acute hypoxia. Rats were exposed to normobaric hypoxia (10% O2) over 1.5 or 6 h and received 0.9% NaCl or adrenergic blockers either as infusion (1 ml/h, increased fluid load) or injection (0.5 ml, reduced fluid load). Control animals were kept in normoxia and received infusions or injections of 0.9% NaCl. After 6 h of hypoxia, LV inotropic function was maintained with NaCl injection but decreased significantly with NaCl infusion. Adrenergic blockade induced a similar LV depression when fluid load was low, but did not further deteriorate LV depression after 6 h of infusion. Reduced fluid load also attenuated pulmonary injury after 6 h of hypoxia. This might be due to an effective fluid drainage into the pleural space. Adrenergic blockade could not prevent PE. In general, increased fluid load and impaired LV inotropic function promote the development of PE in acute hypoxia. The main physiologic conclusion from this study is that fluid reduction under hypoxic conditions has a protective effect on cardiopulmonary function. Consequently, appropriate fluid management has particular importance to subjects in hypoxic conditions.


Asunto(s)
Antagonistas Adrenérgicos/farmacología , Ventrículos Cardíacos/efectos de los fármacos , Hipoxia/inducido químicamente , Edema Pulmonar/inducido químicamente , Animales , Femenino , Ventrículos Cardíacos/fisiopatología , Hipoxia/fisiopatología , Pulmón/efectos de los fármacos , Pulmón/fisiopatología , Edema Pulmonar/fisiopatología , Ratas , Ratas Sprague-Dawley
14.
Mol Cell Neurosci ; 108: 103550, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32890729

RESUMEN

The studies of the interaction between the sympathetic and motor nervous systems are extremely relevant due to therapy for many neurodegenerative and cardiovascular disorders involving adrenergic compounds. Evidences indicate close contact between sympathetic varicosities and neuromuscular synapses. This raises questions about the effects of catecholamines on synaptic transmission. The currently available information is contradictory, and the types of adrenoreceptors responsible for modulation of neurotransmitter release have not been identified in mammalian neuromuscular synapses. Our results have shown that the α1A, α1B, α2A, α2B, α2C, and ß1 adrenoreceptor subtypes are expressed in mouse diaphragm muscle containing neuromuscular synapses and sympathetic varicosities. Pharmacological stimulation of adrenoreceptors affects both spontaneous and evoked acetylcholine quantal secretion. Agonists of the α1, α2 and ß1 adrenoreceptors decrease spontaneous release. Activation of the α2 and ß1 adrenoreceptors reduces the number of acetylcholine quanta released in response to a nerve stimulus (quantal content), but an agonist of the ß2 receptors increases quantal content. Activation of α2 and ß2 adrenoreceptors alters the kinetics of acetylcholine quantal release by desynchronizing the neurosecretory process. Specific blockers of these receptors eliminate the effects of the specific agonists. The action of blockers on quantal acetylcholine secretion indicates possible action of endogenous catecholamines on neuromuscular transmission. Elucidating the molecular mechanisms by which clinically utilized adrenomimetics and adrenoblockers regulate synaptic vesicle release at the motor axon terminal will lead to the creation of improved and safer sympathomimetics for the treatment of various neurodegenerative diseases with synaptic defects.


Asunto(s)
Acetilcolina/metabolismo , Unión Neuromuscular/efectos de los fármacos , Receptores Adrenérgicos/metabolismo , Simpatomiméticos/farmacología , Agonistas Adrenérgicos/farmacología , Antagonistas Adrenérgicos/farmacología , Animales , Exocitosis , Femenino , Masculino , Ratones , Ratones Endogámicos BALB C , Potenciales Postsinápticos Miniatura , Unión Neuromuscular/metabolismo , Unión Neuromuscular/fisiología
15.
Int J Mol Sci ; 22(9)2021 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-34063103

RESUMEN

This study was aimed at disclosing the influence of intravesically instilled guanethidine (GUA) on the distribution, relative frequency and chemical coding of both the urinary bladder intramural sympathetic nerve fibers and their parent cell bodies in the caudal mesenteric ganglion (CaMG) in juvenile female pigs. GUA instillation led to a profound decrease in the number of perivascular nerve terminals. Furthermore, the chemical profile of the perivascular innervation within the treated bladder also distinctly changed, as most of axons became somatostatin-immunoreactive (SOM-IR), while in the control animals they were found to be neuropeptide Y (NPY)-positive. Intravesical treatment with GUA led not only to a significant decrease in the number of bladder-projecting tyrosine hydroxylase (TH) CaMG somata (94.3 ± 1.8% vs. 73.3 ± 1.4%; control vs. GUA-treated pigs), but simultaneously resulted in the rearrangement of their co-transmitters repertoire, causing a distinct decrease in the number of TH+/NPY+ (89.6 ± 0.7% vs. 27.8 ± 0.9%) cell bodies and an increase in the number of SOM-(3.6 ± 0.4% vs. 68.7 ± 1.9%), calbindin-(CB; 2.06 ± 0.2% vs. 9.1 ± 1.2%) or galanin-containing (GAL; 1.6 ± 0.3% vs. 28.2 ± 1.3%) somata. The present study provides evidence that GUA significantly modifies the sympathetic innervation of the porcine urinary bladder wall, and thus may be considered a potential tool for studying the plasticity of this subdivision of the bladder innervation.


Asunto(s)
Antagonistas Adrenérgicos/farmacología , Axones/fisiología , Ganglios Simpáticos/fisiología , Guanetidina/farmacología , Vejiga Urinaria/inervación , Animales , Axones/efectos de los fármacos , Dopamina beta-Hidroxilasa/metabolismo , Femenino , Ganglios Simpáticos/efectos de los fármacos , Fibras Nerviosas/efectos de los fármacos , Fibras Nerviosas/metabolismo , Neuropéptido Y/metabolismo , Porcinos , Vejiga Urinaria/efectos de los fármacos
16.
Int J Mol Sci ; 22(24)2021 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-34948196

RESUMEN

Although guanethidine (GUA) was used in the past as a drug to suppress hyperactivity of the sympathetic nerve fibers, there are no available data concerning the possible action of this substance on the sensory component of the peripheral nervous system supplying the urinary bladder. Thus, the present study was aimed at disclosing the influence of intravesically instilled GUA on the distribution, relative frequency, and chemical coding of dorsal root ganglion neurons associated with the porcine urinary bladder. The investigated sensory neurons were visualized with a retrograde tracing method using Fast Blue (FB), while their chemical profile was disclosed with single-labeling immunohistochemistry using antibodies against substance P (SP), calcitonin gene-related peptide (CGRP), pituitary adenylate cyclase activating polypeptide (PACAP), galanin (GAL), neuronal nitric oxide synthase (nNOS), somatostatin (SOM), and calbindin (CB). After GUA treatment, a slight decrease in the number of FB+ neurons containing SP was observed when compared with untreated animals (34.6 ± 6.5% vs. 45.6 ± 1.3%), while the number of retrogradely traced cells immunolabeled for GAL, nNOS, and CB distinctly increased (12.3 ± 1.0% vs. 7.4 ± 0.6%, 11.9 ± 0.6% vs. 5.4 ± 0.5% and 8.6 ± 0.5% vs. 2.7 ± 0.4%, respectively). However, administration of GUA did not change the number of FB+ neurons containing CGRP, PACAP, or SOM. The present study provides evidence that GUA significantly modifies the sensory innervation of the porcine urinary bladder wall and thus may be considered a potential tool for studying the plasticity of this subdivision of the bladder innervation.


Asunto(s)
Ganglios Espinales/metabolismo , Guanetidina/farmacología , Vejiga Urinaria/inervación , Antagonistas Adrenérgicos/farmacología , Neuronas Adrenérgicas/efectos de los fármacos , Neuronas Adrenérgicas/metabolismo , Animales , Calbindinas/metabolismo , Péptido Relacionado con Gen de Calcitonina/metabolismo , Femenino , Galanina/metabolismo , Ganglios Espinales/efectos de los fármacos , Guanetidina/metabolismo , Neurotoxinas/farmacología , Óxido Nítrico Sintasa/metabolismo , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/metabolismo , Células Receptoras Sensoriales/metabolismo , Somatostatina/metabolismo , Sustancia P/metabolismo , Porcinos , Vejiga Urinaria/efectos de los fármacos
17.
Molecules ; 26(18)2021 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-34577088

RESUMEN

We report the first isolation of the alkaloid aaptamine from the Philippine marine sponge Stylissa sp. Aaptamine possessed weak antiproliferative activity against HCT116 colon cancer cells and inhibited the proteasome in vitro at 50 µM. These activities may be functionally linked. Due to its known, more potent activity on certain G-protein coupled receptors (GPCRs), including α-adrenergic and δ-opioid receptors, the compound was profiled more broadly at sub-growth inhibitory concentrations against a panel of 168 GPCRs to potentially reveal additional targets and therapeutic opportunities. GPCRs represent the largest class of drug targets. The primary screen at 20 µM using the ß-arrestin functional assay identified the antagonist, agonist, and potentiators of agonist activity of aaptamine. Dose-response analysis validated the α-adrenoreceptor antagonist activity of aaptamine (ADRA2C, IC50 11.9 µM) and revealed the even more potent antagonism of the ß-adrenoreceptor (ADRB2, IC50 0.20 µM) and dopamine receptor D4 (DRD4, IC50 6.9 µM). Additionally, aaptamine showed agonist activity on selected chemokine receptors, by itself (CXCR7, EC50 6.2 µM; CCR1, EC50 11.8 µM) or as a potentiator of agonist activity (CXCR3, EC50 31.8 µM; CCR3, EC50 16.2 µM). These GPCRs play a critical role in the treatment of cardiovascular disease, diabetes, cancer, and neurological disorders. The results of this study may thus provide novel preventive and therapeutic strategies for noncommunicable diseases (NCDs).


Asunto(s)
Alcaloides/farmacología , Naftiridinas/farmacología , Enfermedades no Transmisibles/tratamiento farmacológico , Poríferos/química , Antagonistas Adrenérgicos/farmacología , Alcaloides/química , Alcaloides/aislamiento & purificación , Regulación Alostérica/efectos de los fármacos , Animales , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Antagonistas de Dopamina/farmacología , Humanos , Naftiridinas/química , Naftiridinas/aislamiento & purificación , Filipinas , Receptores Adrenérgicos/efectos de los fármacos , Receptores de Quimiocina/agonistas , Receptores de Quimiocina/efectos de los fármacos , Receptores Dopaminérgicos/efectos de los fármacos , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Receptores Acoplados a Proteínas G/efectos de los fármacos , Saccharomyces cerevisiae/efectos de los fármacos
18.
Ter Arkh ; 93(10): 1193-1202, 2021 Oct 15.
Artículo en Ruso | MEDLINE | ID: mdl-36286821

RESUMEN

BACKGROUND: The main factors that increase the risk of cardiovascular accidents and mortality among patients with COVID-19 include hyperglycemia, arterial hypertension and dyslipidemia. Therefore, all patients with COVID-19 and metabolic syndrome should receive antihypertensive (AHT), hypolipidemic (GLT) and hypoglycemic therapy (GGT). Currently, there is a limited number of studies regarding the effectiveness and safety of this therapy in patients with COVID-19. AIM: Evaluate the clinical outcomes of patients with COVID-19, depending on the recipient of AHT, GLT and GGT. MATERIALS AND METHODS: A retrospective analysis of the clinical outcomes "discharged/died" of 1753 patients with COVID-19 was carried out depending on the received AHT, GLT and GGT. RESULTS: A significant reduction in the risk of mortality among patients with COVID-19 was observed during therapy with angiotensin-converting enzyme inhibitors/angiotensin II receptor blockers ACE inhibitors/ARBs (OR 0.39, 95% CI 0.210.72; p0.05) and b-adrenergic blockers b-AB (OR 0.53, 95% CI 0.281; p0.05). At the same time, against the background of therapy with ACE inhibitors/ARBs and b-ABs, the chance of mortality decreased more significantly among patients with type 2 diabetes mellitus (T2DM) compared with patients without T2DM. Diuretic therapy was associated with a 3-fold increase in the chances of death: OR 3.33, 95% CI 1.884.79; p0.05. Statin therapy did not affect clinical outcomes in COVID-19 patients. On the background of therapy with oral hypoglycemic drugs, the risk of mortality decreased 5-fold (OR 0.19, 95% CI 0.070.54; p0.05). Against the background of insulin therapy, there was an increase in mortality risk by 2.8 times (OR 2.81, 95% CI 1.55.29; p0.05). CONCLUSION: A significant reduction in mortality among patients with COVID-19 was observed during therapy with ACEI/ARB, b-AB, and oral hypoglycemic therapy. Increased risk of death was associated with insulin therapy and diuretic therapy.


Asunto(s)
Tratamiento Farmacológico de COVID-19 , Diabetes Mellitus Tipo 2 , Inhibidores de Hidroximetilglutaril-CoA Reductasas , Hipertensión , Insulinas , Humanos , Antihipertensivos/efectos adversos , Antagonistas de Receptores de Angiotensina/efectos adversos , Inhibidores de la Enzima Convertidora de Angiotensina/efectos adversos , Estudios Retrospectivos , Diabetes Mellitus Tipo 2/complicaciones , Hipertensión/tratamiento farmacológico , Hipertensión/complicaciones , Antagonistas Adrenérgicos/uso terapéutico , Hipoglucemiantes/efectos adversos , Diuréticos , Insulinas/uso terapéutico , Lípidos
19.
J Cell Mol Med ; 24(20): 11791-11799, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32885586

RESUMEN

Trauma-induced calcification is the pathological consequence of complex injuries which often affect the central nervous system and other parts of the body simultaneously. We demonstrated by an animal model recapitulating the calcification of the above condition that adrenaline transmits the stress signal of brain injury to the calcifying tissues. We have also found that although the level of plasma pyrophosphate, the endogenous inhibitor of calcification, was normal in calcifying animals, it could not counteract the acute calcification. However, externally added pyrophosphate inhibited calcification even when it was administered after the complex injuries. Our finding suggests a potentially powerful clinical intervention of calcification triggered by polytrauma injuries which has no effective treatment.


Asunto(s)
Lesiones Traumáticas del Encéfalo/complicaciones , Difosfatos/uso terapéutico , Osificación Heterotópica/complicaciones , Calcificación Vascular/etiología , Antagonistas Adrenérgicos/farmacología , Animales , Lesiones Traumáticas del Encéfalo/patología , Cardiotoxinas , Difosfatos/sangre , Modelos Animales de Enfermedad , Epinefrina , Femenino , Regulación de la Expresión Génica , Ratones Endogámicos C57BL , Músculo Esquelético/diagnóstico por imagen , Músculo Esquelético/patología , Osificación Heterotópica/sangre , Osificación Heterotópica/diagnóstico por imagen , Receptores Adrenérgicos/metabolismo , Calcificación Vascular/sangre , Calcificación Vascular/diagnóstico por imagen , Calcificación Vascular/genética , Microtomografía por Rayos X
20.
CA Cancer J Clin ; 63(3): 167-92, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23355109

RESUMEN

Hot flashes are prevalent and severe symptoms that can interfere with mood, sleep, and quality of life for women and men with cancer. The purpose of this article is to review existing literature on the risk factors, pathophysiology, and treatment of hot flashes in individuals with cancer. Electronic searches were conducted to identify relevant English-language literature published through June 15, 2012. Results indicated that risk factors for hot flashes in cancer include patient-related factors (eg, age, race/ethnicity, educational level, smoking history, cardiovascular risk including body mass index, and genetics) and disease-related factors (eg, cancer diagnosis and dose/type of treatment). In addition, although the pathophysiology of hot flashes has remained elusive, these symptoms are likely attributable to disruptions in thermoregulation and neurochemicals. Therapies that have been offered or tested fall into 4 broad categories: pharmacological, nutraceutical, surgical, and complementary/behavioral strategies. The evidence base for this broad range of therapies varies, with some treatments not yet having been fully tested or showing equivocal results. The evidence base surrounding all therapies is evaluated to enhance hot flash treatment decision-making by clinicians and patients.


Asunto(s)
Sofocos/etiología , Neoplasias/complicaciones , Antagonistas Adrenérgicos/uso terapéutico , Anticonvulsivantes/uso terapéutico , Antidepresivos/uso terapéutico , Bloqueo Nervioso Autónomo , Regulación de la Temperatura Corporal/fisiología , Antagonistas Colinérgicos/uso terapéutico , Terapia Cognitivo-Conductual , Terapias Complementarias , Sofocos/fisiopatología , Sofocos/terapia , Humanos , Neoplasias/fisiopatología , Neoplasias/terapia , Fitoterapia , Factores de Riesgo , Ganglio Estrellado/cirugía , Vitaminas/uso terapéutico
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda