Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.154
Filter
2.
Nature ; 623(7989): 992-1000, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37968397

ABSTRACT

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.


Subject(s)
Brain Edema , Brain Injuries, Traumatic , Glymphatic System , Norepinephrine , Animals , Mice , Adrenergic Antagonists/pharmacology , Adrenergic Antagonists/therapeutic use , Brain Edema/complications , Brain Edema/drug therapy , Brain Edema/metabolism , Brain Edema/prevention & control , Brain Injuries, Traumatic/complications , Brain Injuries, Traumatic/drug therapy , Brain Injuries, Traumatic/metabolism , Disease Models, Animal , Glymphatic System/drug effects , Glymphatic System/metabolism , Inflammation/complications , Inflammation/drug therapy , Inflammation/metabolism , Inflammation/prevention & control , Lymphatic Vessels/metabolism , Norepinephrine/metabolism , Phosphorylation , Receptors, Adrenergic/metabolism
3.
Rev Mal Respir ; 40(8): 716-722, 2023 Oct.
Article in French | MEDLINE | ID: mdl-37633811

ABSTRACT

INTRODUCTION: Ectopic Cushing's syndrome (CS) is a rare condition nevertheless well-known to endocrinologists. The pneumologist may be called upon to treat CS not only because bronchial carcinoid tumors are the most frequent source of ectopic ACTH secretion, but also due to the fact that the immunosuppression induced by hypercorticism favors lower respiratory tract infections. CASE REPORT: We report the case of a female patient presenting with acute respiratory failure secondary to Enterobacter cloacae pneumonia exacerbated by SC. Further investigations confirmed ectopic ACTH secretion and revealed a right upper lobe pulmonary nodule. After appropriate antibiotic therapy, the patient received preoperative adrenolytic treatment. Management by right upper lobectomy resulted in the extraction of a 12mm tumor. Pathological analysis was consistent with the diagnosis of a typical carcinoid tumor. Immunohistochemistry confirmed ACTH secretion by the tumor. Even though the postoperative course showed CS regression, the patient developed adrenal insufficiency. CONCLUSION: Ectopic CS induces immunosuppression, which aggravates lower respiratory tract infections. Search for a pulmonary neuroendocrine tumor should be systematic. Following control of the secretory syndrome by adrenolytic treatment, and if the diagnosis of carcinoid tumor is confirmed, surgical treatment is the preferred option.


Subject(s)
ACTH Syndrome, Ectopic , Bronchial Neoplasms , Carcinoid Tumor , Cushing Syndrome , Pneumonia , Respiratory Tract Infections , Humans , Female , Cushing Syndrome/etiology , Cushing Syndrome/complications , ACTH Syndrome, Ectopic/complications , ACTH Syndrome, Ectopic/diagnosis , Bronchial Neoplasms/complications , Bronchial Neoplasms/diagnosis , Bronchial Neoplasms/surgery , Carcinoid Tumor/complications , Carcinoid Tumor/diagnosis , Carcinoid Tumor/surgery , Respiratory Tract Infections/complications , Adrenocorticotropic Hormone , Adrenergic Antagonists
4.
Int J Neurosci ; 133(5): 496-504, 2023 May.
Article in English | MEDLINE | ID: mdl-33998961

ABSTRACT

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.


Subject(s)
Epilepsy , Receptors, Adrenergic, alpha , Rats , Animals , Rats, Wistar , Hippocampus , Epilepsy/therapy , Receptors, Adrenergic, alpha-2 , Adrenergic Antagonists/pharmacology , Electric Stimulation
5.
Commun Biol ; 5(1): 1283, 2022 11 23.
Article in English | MEDLINE | ID: mdl-36418492

ABSTRACT

Vesicular monoamine transporter 2 (VMAT2) is responsible for packing monoamine neurotransmitters into synaptic vesicles for storage and subsequent neurotransmission. VMAT2 inhibitors are approved for symptomatic treatment of tardive dyskinesia and Huntington's chorea, but despite being much-studied inhibitors their exact binding site and mechanism behind binding and inhibition of monoamine transport are not known. Here we report the identification of several approved drugs, notably ß2-adrenergic agonists salmeterol, vilanterol and formoterol, ß2-adrenergic antagonist carvedilol and the atypical antipsychotic ziprasidone as inhibitors of rat VMAT2. Further, plausible binding modes of the established VMAT2 inhibitors reserpine and tetrabenazine and hit compounds salmeterol and ziprasidone were identified using molecular dynamics simulations and functional assays using VMAT2 wild-type and mutants. Our findings show VMAT2 as a potential off-target of treatments with several approved drugs in use today and can also provide important first steps in both drug repurposing and therapy development targeting VMAT2 function.


Subject(s)
Antipsychotic Agents , Animals , Rats , Adrenergic Agonists , Antipsychotic Agents/pharmacology , Piperazines , Vesicular Monoamine Transport Proteins/genetics , Adrenergic Antagonists/pharmacology
7.
Int J Mol Sci ; 23(21)2022 Oct 24.
Article in English | MEDLINE | ID: mdl-36361620

ABSTRACT

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.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Carcinoma, Non-Small-Cell Lung/metabolism , Cisplatin/pharmacology , Cisplatin/therapeutic use , Nicotine/pharmacology , Brain-Derived Neurotrophic Factor/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , ErbB Receptors/metabolism , Propranolol/pharmacology , Propranolol/therapeutic use , Adrenergic Antagonists/pharmacology , Drug Resistance, Neoplasm , Phosphatidylinositol 3-Kinases/metabolism , Receptors, Adrenergic, beta , Cell Line, Tumor
9.
Ter Arkh ; 94(2): 200-208, 2022 Feb 15.
Article in Russian | MEDLINE | ID: mdl-36286743

ABSTRACT

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.


Subject(s)
Hypertension, Portal , Propranolol , Humans , Adrenergic Antagonists/therapeutic use , Cytochrome P-450 CYP2D6/genetics , Cytochrome P-450 CYP2D6/therapeutic use , Hemodynamics , Hypertension, Portal/diagnosis , Hypertension, Portal/drug therapy , Hypertension, Portal/etiology , Liver Cirrhosis/complications , Liver Cirrhosis/diagnosis , Liver Cirrhosis/drug therapy , Propranolol/therapeutic use , Polymorphism, Genetic
10.
J Prev Alzheimers Dis ; 9(4): 679-691, 2022.
Article in English | MEDLINE | ID: mdl-36281672

ABSTRACT

BACKGROUND: Arterial hypertension is among factors with the potential for increasing the risk of cognitive impairment in elderly subjects. However, studies investigating the effects of antihypertensives on cognitive function have reported mixed results. METHODS: We have used the National Alzheimer's Coordinating Center (NACC) Uniform Data Set (UDS) to investigate the effect of each class of antihypertensives, both as single and combined, in reducing the rate of conversion from normal to mild cognitive impairment (MCI). RESULTS: The use of antihypertensive drugs was associated with 21% (Hazard ratio: 0.79, p<01001) delay in the rate of conversion to MCI. This effect was modulated by age, gender, and genotypic APOE4 allele. Among different antihypertensive subclasses, calcium channel blockers (CCBs) (24%, HR: 0.76, P=0.004), diuretics (21%, HR: 0.79, P=0.006), and α1-adrenergic blockers (α1-ABs) (23%, HR: 0.77, P=0.034) significantly delayed the rate of MCI conversion. A significant effect was observed with the selective L-type voltage-gated CCBs, dihydropyridines, amlodipine (47%, HR=0.53, P<0.001) and nifedipine (49%, HR=0.51, P=0.012), whereas non-DHPs showed insignificant effect. Loop diuretics, potassium sparing diuretics, and thiazides all significantly reduced the rate of MCI conversion. Combination of α1-AB and diuretics led to synergistic effects; combination of vasodilators plus ß-blockers (ßBs), and α1-AB plus ßBs led to additive effect in delaying the rate of MCI conversion, when compared to a single drug. CONCLUSION: Our results could have implications for the more effective treatment of hypertensive elderly adults who are likely to be at high risk of cognitive decline and dementia. The choice of combination of antihypertensive therapy should also consider the combination which would lead to an optimum benefit on cognitive function.


Subject(s)
Dihydropyridines , Hypertension , Adult , Humans , Aged , Antihypertensive Agents/therapeutic use , Calcium Channel Blockers/therapeutic use , Cohort Studies , Nifedipine/therapeutic use , Apolipoprotein E4 , Hypertension/drug therapy , Hypertension/genetics , Hypertension/complications , Thiazides/therapeutic use , Diuretics/therapeutic use , Amlodipine/therapeutic use , Dihydropyridines/therapeutic use , Cognition , Diuretics, Potassium Sparing/therapeutic use , Genotype , Vasodilator Agents/therapeutic use , Adrenergic Antagonists/therapeutic use
11.
Int J Mol Sci ; 23(18)2022 Sep 08.
Article in English | MEDLINE | ID: mdl-36142287

ABSTRACT

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.


Subject(s)
Anti-Arrhythmia Agents , Antihypertensive Agents , Aconitine/adverse effects , Adrenergic Antagonists , Animals , Anti-Arrhythmia Agents/pharmacology , Anti-Arrhythmia Agents/therapeutic use , Antihypertensive Agents/pharmacology , Arrhythmias, Cardiac/chemically induced , Arrhythmias, Cardiac/drug therapy , Arrhythmias, Cardiac/prevention & control , Calcium Chloride , Epinephrine/pharmacology , Epinephrine/therapeutic use , Methoxamine , Pyrrolidinones/pharmacology , Rats , Rats, Wistar , Receptors, Adrenergic, alpha-1 , Receptors, Adrenergic, beta-1
12.
Toxicon ; 218: 57-65, 2022 Oct 30.
Article in English | MEDLINE | ID: mdl-36113683

ABSTRACT

Rhinella marina toad is abundant in Brazil. Its poison contains cardiac glycosides called bufadienolides, which are extensively investigated for their bioactivity. Our aim was to characterize the vasoactivity of Rhinella marina poison (RmP) on the aorta of male Wistar rats. For this, the RmP was first collected and processed to obtain an alcoholic extract. To determine cardiovascular effects of RmP, we performed in vivo tests by administering RmP intravenously in doses of 0.1-0.8 mg/kg. Vascular reactivity was also performed through concentration-response curves to RmP (10 ng/mL to 200 µg/mL) in aortic segments with and without endothelium. RmP induced a concentration-dependent contraction in rat aorta which was partly endothelium-mediated. Nitric oxide contributes with this response in view that incubation with L-NAME increased the contractile response. Additionally, treatment with indomethacin [cyclooxygenase, (COX) inhibitor], nifedipine (L-type voltage-gated calcium channels blocker), and BQ-123 (ETA receptors antagonist) decreased maximum response, and ketanserin (5-HT2 receptors antagonist) decreased pEC50, suggesting active participation of these pathways in the contractile response. On the other hand, apocynin (NADPH oxidase inhibitor) did not alter contractility. Incubation with prazosin (α1-adrenergic receptor antagonist) abolished the contractile response, suggesting that the RmP-induced contraction is dependent on the adrenergic pathway. In the Na+/K+ ATPase protocol, a higher Emax was observed in the RmP experimental group, suggesting that RmP potentiated Na+/K+ATPase hyperpolarizing response. When this extract was injected (i.v.) in vivo, increase in blood pressure and decrease in heart rate were observed. The results were immediate and transitory, and occurred in a dose-dependent manner. Overall, these data suggest that the poison extract of R. marina toad has an important vasoconstrictor action and subsequent vasopressor effects, and its use can be investigated to some cardiovascular disorders.


Subject(s)
Bufanolides , Poisons , Adenosine Triphosphatases/metabolism , Adenosine Triphosphatases/pharmacology , Adrenergic Agents/pharmacology , Adrenergic Antagonists/pharmacology , Animals , Bufanolides/toxicity , Bufo marinus/metabolism , Calcium Channels , Endothelium, Vascular , Hemodynamics , Indomethacin/pharmacology , Ketanserin/pharmacology , Male , Methanol/pharmacology , NADPH Oxidases , NG-Nitroarginine Methyl Ester , Nifedipine/pharmacology , Nitric Oxide/metabolism , Prazosin/pharmacology , Prostaglandin-Endoperoxide Synthases/metabolism , Rats , Rats, Wistar , Serotonin/pharmacology , Vasoconstrictor Agents
13.
Psychiatry Res ; 317: 114840, 2022 11.
Article in English | MEDLINE | ID: mdl-36162349

ABSTRACT

Algorithms for posttraumatic stress disorder were published by this team in 1999 and 2011. Developments since then warrant revision. New studies and review articles from January 2011 to November 2021 were identified via PubMed and analyzed for evidence supporting changes. Following consideration of variations required by special patient populations, treatment of sleep impairments remains as the first recommended step. Nightmares and non-nightmare disturbed awakenings are best addressed with the anti-adrenergic agent prazosin, with doxazosin and clonidine as alternatives. First choices for difficulty initiating sleep include hydroxyzine and trazodone. If significant non-sleep PTSD symptoms remain, an SSRI should be tried, followed by a second SSRI or venlafaxine as a third step. Second generation antipsychotics can be considered, particularly for SSRI augmentation when PTSD-associated psychotic symptoms are present, with the caveat that positive evidence is limited and side effects are considerable. Anti-adrenergic agents can also be considered for general PTSD symptoms if not already tried, though evidence for daytime use lags that available for sleep. Regarding other pharmacological and procedural options, e.g., transcranial magnetic stimulation, cannabinoids, ketamine, psychedelics, and stellate ganglion block, evidence does not yet support firm inclusion in the algorithm. An interactive version of this work can be found at www.psychopharm.mobi.


Subject(s)
Psychopharmacology , Sleep Initiation and Maintenance Disorders , Sleep Wake Disorders , Stress Disorders, Post-Traumatic , Humans , Stress Disorders, Post-Traumatic/diagnosis , Prazosin/therapeutic use , Prazosin/pharmacology , Dreams , Sleep Wake Disorders/therapy , Sleep Initiation and Maintenance Disorders/drug therapy , Adrenergic Antagonists/pharmacology , Adrenergic Antagonists/therapeutic use
14.
Neurochem Res ; 47(11): 3440-3453, 2022 Nov.
Article in English | MEDLINE | ID: mdl-35945306

ABSTRACT

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.


Subject(s)
Cognitive Dysfunction , Dexmedetomidine , Neuroprotective Agents , Reperfusion Injury , Adrenergic Antagonists/therapeutic use , Animals , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Benzylamines , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/etiology , Dexmedetomidine/pharmacology , Dexmedetomidine/therapeutic use , Eosine Yellowish-(YS)/therapeutic use , Hematoxylin/therapeutic use , Ischemia , Locus Coeruleus/metabolism , Malondialdehyde , Mice , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Neurotoxins , Norepinephrine , Reperfusion , Reperfusion Injury/complications , Reperfusion Injury/drug therapy , Tumor Necrosis Factor-alpha/metabolism , Tyrosine 3-Monooxygenase/metabolism , Yohimbine/therapeutic use
15.
Mol Biol Rep ; 49(10): 9565-9573, 2022 Oct.
Article in English | MEDLINE | ID: mdl-35970968

ABSTRACT

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.


Subject(s)
Aphrodisiacs , Mouth Neoplasms , Adrenergic Antagonists/pharmacology , Aphrodisiacs/pharmacology , Apoptosis , Cell Line, Tumor , Humans , Membrane Potential, Mitochondrial , Mouth Neoplasms/drug therapy , Reactive Oxygen Species/metabolism , Yohimbine/pharmacology
16.
Andrology ; 10(8): 1540-1547, 2022 Nov.
Article in English | MEDLINE | ID: mdl-35934935

ABSTRACT

BACKGROUND: Rat isolated vas deferens releases 6-nitrodopamine (6-ND), and the spasmogenic activity of this novel catecholamine is significantly reduced by tricyclic compounds such as amitriptyline, desipramine, and carbamazepine and by antagonists of the α1 -adrenergic receptors such as doxazosin, tamsulosin, and prazosin. OBJECTIVES: To investigate the liberation of 6-ND by human epididymal vas deferens (HEVDs) and its pharmacological actions. METHODS: The in vitro liberation of 6-ND, dopamine, noradrenaline, and adrenaline from human vas deferens was evaluated by LC-MS/MS. The contractile effect of the catecholamines in HEVDs was investigated in vitro. The action of tricyclic antidepressants was evaluated on the spasmogenic activity ellicited by the catecholamines and by the electric-field stimulation (EFS). The tissue was also incubated with the inhibitor of nitric oxide (NO) synthase L-NAME and the release of catecholamines and the contractile response to EFS were assessed. RESULTS: 6-ND is the major catecholamine released from human vas deferens and its synthesis/release is inhibited by NO inhibition. The spasmogenic activity elicited by EFS in the human vas deferens was blocked by tricyclic antidepressants only at concentrations that selectively antagonize 6-ND induced contractions of the human vas deferens, without affecting the spasmogenic activity induced by dopamine, noradrenaline, and adrenaline in this tissue. Incubation of the vas deferens with L-NAME reduced both the 6-ND release and the contractions induced by EFS. DISCUSSION AND CONCLUSION: 6-ND should be considered a major endogenous modulator of human vas deferens contractility and possibly plays a pivotal role in the emission process of ejaculation. It offers a novel and shared mechanism of action for tricyclic antidepressants and α1 -adrenergic receptor antagonists.


Subject(s)
Dopamine , Vas Deferens , Adrenergic Antagonists/pharmacology , Amitriptyline/pharmacology , Animals , Antidepressive Agents, Tricyclic/pharmacology , Carbamazepine/pharmacology , Chromatography, Liquid , Desipramine/pharmacology , Dopamine/analogs & derivatives , Dopamine/pharmacology , Doxazosin/pharmacology , Epinephrine/pharmacology , Humans , Male , Muscle Contraction , Muscle, Smooth , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide , Norepinephrine/pharmacology , Prazosin/pharmacology , Rats , Receptors, Adrenergic , Tamsulosin/pharmacology , Tandem Mass Spectrometry
17.
Arch Razi Inst ; 77(1): 141-149, 2022 02.
Article in English | MEDLINE | ID: mdl-35891757

ABSTRACT

The current study was conducted to investigate the interaction between the central adrenergic and histaminergic systems and the broiler chick's feed intake. In the first experiment, the intracerebroventricular (ICV) injection of solutions was conducted which included 10 nmol of prazosin (an α1-receptor antagonist), 300 nmol of histamine, co-injection of prazosin and histamine. Experiments two to five were conducted similarly the same as the first experiment, in which chickens were ICV injected with 13 nmol of yohimbine (an α2-receptor antagonist), 24 nmol of metoprolol (a ß1 adrenergic receptor antagonist), 5 nmol of ICI 118,551 (a ß2 adrenergic receptor antagonist), and 20 nmol of SR 59230R (a ß3 adrenergic receptor antagonist). The injected solutions in the sixth experiment included 300 nmol of noradrenaline, 250 nmol of α-FMH (an alpha fluoromethyl histidine), noradrenaline, and α-FMH. Seventh to ninth experiments were similar to the sixth experiment, except that the chickens were ICV injected with 300 nmol of chlorpheniramine (a histamine H1 receptors antagonist), 82 nmol of famotidine (a histamine H2 receptors antagonist), and 300 nmol of thioperamide (a histamine H3 receptors antagonist), rather than α-FMH. Afterward, the cumulative food intake was measured 120 min after injection. Based on the obtained results, both histamine ICV injection and noradrenaline injection reduced food intake (P<0.05). Moreover, co-injection of histamine and ICI 118,551 (P<0.05), and co-injection of noradrenaline and Chlorpheniramine reduced food intake (P<0.05). In addition, noradrenaline and Thioperamide co-injection improved hypophagic effect of noradrenaline in neonatal chicken (P<0.05). These findings suggested the effect of interconnection between adrenergic and histaminergic systems, which may be mediated by H1 and H3 histaminergic and ß2 adrenergic receptors, on the regulation of food intake in the neonatal broiler chicken.


Subject(s)
Appetite , Chickens , Adrenergic Agents/pharmacology , Adrenergic Antagonists/pharmacology , Animals , Animals, Newborn , Chlorpheniramine/pharmacology , Feeding Behavior/physiology , Histamine/pharmacology , Norepinephrine/pharmacology , Prazosin/pharmacology , Receptors, Adrenergic , Receptors, Histamine
18.
J Acupunct Meridian Stud ; 15(1): 25-36, 2022 Feb 28.
Article in English | MEDLINE | ID: mdl-35770571

ABSTRACT

Background: Hyperplastic morphological changes associated with erythropoiesis have been reported in the primo vascular system (PVS) tissue on the surface of abdominal organs in rats with heart failure (HF) or hemolytic anemia (HA). Objectives: Since adrenergic activity is commonly activated in both HF and HA, we investigated whether adrenergic signaling mediates the abovementioned morphological changes. Methods: We compared the effects of adrenolytic treatments (exercise training and 6-hydroxydopamine) on the gross morphology of the PVS tissues isolated from organ surfaces in HF or HA rats. HF and HA were induced by ligating the left coronary artery and injecting phenylhydrazine, respectively. We further compared the effects of norepinephrine and norepinephrine plus α- or ß-adrenoceptor blockers. Results: The number of samples per rat, PN size, and proportion of red-colored samples in the PVS tissue increased in the HF and HA rats. These changes were reversed by adrenolytic treatments. Interestingly, 6-hydroxydopamine also reversed phenylhydrazineinduced hemolytic changes in erythrocytes. Subcutaneous administration of norepinephrine (3 mg/kg/d) increased the sampling frequency per rat and the PN size, but these effects were blunted at a higher dose (10 mg/kg/d). Norepinephrine administration had little effect on the proportion of red-colored tissues. Norepinephrine-induced morphological changes were completely blocked by a ß-adrenoceptor antagonist (propranolol) but increased slightly by an α-adrenoceptor antagonist (phentolamine). Conclusion: Adrenergic signaling controls hyperplastic changes in the organ surface PVS in rats. These findings may explain the morphological dynamics of the PVS tissues proposed by Bong Han Kim and further clarify the pathophysiological roles of the PVS.


Subject(s)
Adrenergic Agents , Norepinephrine , Adrenergic Antagonists , Animals , Norepinephrine/pharmacology , Oxidopamine , Rats , Receptors, Adrenergic
19.
J Neurotrauma ; 39(13-14): 964-978, 2022 07.
Article in English | MEDLINE | ID: mdl-35412843

ABSTRACT

Disruption of endogenous pain control mechanisms including descending pain inhibition has been linked to several forms of pain including chronic pain after traumatic brain injury (TBI). The locus coeruleus (LC) is the principal noradrenergic (NA) nucleus participating in descending pain inhibition. We therefore hypothesized that selectively stimulating LC neurons would reduce nociception after TBI. All experiments used a well-characterized rat lateral fluid percussion model of TBI. NA neurons were stimulated by administering clozapine N-oxide (CNO) to rats selectively expressing a designer receptor exclusively activated by designer drug (DREADD) viral construct in their LC's. Mechanical nociceptive thresholds were measured using von Frey fibers. The efficacy of diffuse noxious inhibitory control (DNIC), a critical endogenous pain control mechanism, was assessed using the hindpaw administration of capsaicin. Immunohistochemical analyses demonstrated the selective expression of the DREADD construct in LC neurons after stereotactic injection. During the 1st week after TBI, when rats demonstrated hindlimb (HL) nociceptive sensitization, CNO administration provided transient anti-allodynia in DREADD-expressing rats but not in rats injected with control virus. Seven weeks after TBI we observed a complete loss of DNIC in response to capsaicin. However, CNO administration largely restored DNIC in TBI DREADD-expressing rats but not those injected with control virus. Unexpectedly, the effects of LC activation in the DREADD-expressing rats were blocked by the α-1 adrenergic receptor antagonist prazosin, but not the α-2 adrenergic receptor antagonist atipamezole. These results suggest that directly stimulating the LC after TBI can reduce both early and late manifestations of dysfunctional endogenous pain regulation. Clinical approaches to activating descending pain circuits may reduce suffering in those with pain after TBI.


Subject(s)
Brain Injuries, Traumatic , Chronic Pain , Designer Drugs , Adrenergic Antagonists , Animals , Brain Injuries, Traumatic/complications , Brain Injuries, Traumatic/drug therapy , Capsaicin , Designer Drugs/pharmacology , Locus Coeruleus , Nociception , Rats
20.
Abdom Radiol (NY) ; 47(12): 4032-4041, 2022 12.
Article in English | MEDLINE | ID: mdl-35347385

ABSTRACT

Bladder pheochromocytomas (PCCs) are rare tumors that account for 0.06% of all bladder tumors and makeup 1% of all PCCs. Most PCCs are functional, and they secrete catecholamines that lead to clinical symptoms such as paroxysmal hypertension, headaches, palpitations, and sweating. However, some are nonfunctional and asymptomatic and are hence difficult to diagnose. Cystoscopy and biopsy should not be performed when bladder PCCs are suspected. They may provoke a hypertensive crisis if preventative antiadrenergic blockers are not administered prior to the procedure. The diagnostic workup begins with obtaining blood or urine catecholamine and catecholamine metabolite values to make a presumptive diagnosis of bladder PCC. Computed tomography (C.T.) and magnetic resonance imaging (MRI) are then used to localize and stage the tumor for surgical resection. MRI, due to its superior soft tissue resolution and the ability to use multiparametric MRI (mpMRI) to differentiate between layers of the bladder wall and from other bladder masses, is the optimal imaging modality to detect extra-adrenal bladder PCCs and determine locoregional staging. Once antiadrenergic medications are given, the tumor is resected, and the diagnosis is confirmed histologically. However, the differential diagnosis of bladder PCC often gets overlooked, leading to surgical resection in the absence of antiadrenergic medications, increasing the chances of a fatal hypertensive crisis. This makes MRI an essential diagnostic tool for staging bladder PCCs before surgery. This review discusses the indications for MRI in bladder PCCs and describes findings from these tumors on various MRI sequences and when to use them. We also discuss how MRI can differentiate bladder PCCs from other bladder neoplasms.


Subject(s)
Adrenal Gland Neoplasms , Pheochromocytoma , Urinary Bladder Neoplasms , Humans , Pheochromocytoma/pathology , Urinary Bladder/pathology , Adrenal Gland Neoplasms/surgery , Magnetic Resonance Imaging/methods , Catecholamines , Urinary Bladder Neoplasms/pathology , Adrenergic Antagonists
SELECTION OF CITATIONS
SEARCH DETAIL
...