Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.331
Filter
Add more filters

Publication year range
1.
Zhongguo Zhong Yao Za Zhi ; 47(17): 4691-4697, 2022 Sep.
Article in Chinese | MEDLINE | ID: mdl-36164876

ABSTRACT

To investigate the effect of Rehmanniae Radix on depression-like behavior and monoamine neurotransmitters of chronic unpredictable mild stress(CUMS) model rats. CUMS combined with isolated feeding was used to induce the depression model of rats. The depression-like behavior of rats was evaluated by sucrose preference test, open field test, and forced swim test. Hematoxylin-Eosin(HE) staining was used to investigate the pathological changes of neurons in the CA1 and CA3 area of hippocampus. Ultra performance liquid chromatography-tandem mass spectrometry(UPLC-MS) was used to detect the contents of 5-hydroxytryptamine(5-HT), 5-hydroxyindoleacetic acid(5-HIAA), dopamine(DA), 3,4-dihydroxyphenylacetic acid(DOPAC), homovanillic acid(HVA), norepinephrine(NE), and 3-methoxy-4-hydroxyphenyl glycol(MHPG) in rats. Western blot was used to detect the protein expressions of tryptophan hydroxylase 2(TPH2), serotonin transporter(SERT), and monoamine oxidase A(MAO-A) in the hippocampus of rats. Compared with the normal group, depressive-like behavior of rats was obvious in the model group. The arrangements of neurons in the CA1 and CA3 area of hippocampus were loose and disorderly. The levels of 5-HT, 5-HIAA, and 5-HT/5-HIAA in the hippocampal area were decreased(P<0.01). The protein expression of TPH2 was decreased(P<0.01), but those of SERT and MAO-A were increased(P<0.01). In the Rehmanniae Radix groups with 1.8 g·kg~(-1) and 7.2 g·kg~(-1), the depression-like behavior of CUMS rats and pathological changes of neurons in CA1, CA3 area of hippocampus were improved. The protein expression of TPH2(P<0.05, P<0.01) was increased, and those of SERT and MAO-A were down-regulated(P<0.05, P<0.01). The levels of 5-HT, 5-HIAA, and 5-HT/5-HIAA in hippocampus were increased(P<0.05, P<0.01). The changes in DA, DOPAC, HVA, DA/(DOPAC +HVA), NE, DHPG, and NE/DHPG were not statistically significant. The results suggested that Rehmanniae Radix improved depression-like behavior of CUMS rats, and the mechanism might be related to the regulation of synthesis, transportation, and metabolism of 5-HT neurotransmitter in the hippocampus.


Subject(s)
Antidepressive Agents , Depression , Hippocampus , Hydroxyindoleacetic Acid , Rehmannia , Serotonin , 3,4-Dihydroxyphenylacetic Acid/metabolism , 3,4-Dihydroxyphenylacetic Acid/pharmacology , Animals , Antidepressive Agents/pharmacology , Antidepressive Agents/therapeutic use , Chromatography, Liquid , Depression/drug therapy , Disease Models, Animal , Dopamine , Eosine Yellowish-(YS)/metabolism , Eosine Yellowish-(YS)/pharmacology , Hematoxylin/metabolism , Hematoxylin/pharmacology , Hippocampus/metabolism , Homovanillic Acid/metabolism , Homovanillic Acid/pharmacology , Hydroxyindoleacetic Acid/metabolism , Methoxyhydroxyphenylglycol/analogs & derivatives , Methoxyhydroxyphenylglycol/metabolism , Methoxyhydroxyphenylglycol/pharmacology , Monoamine Oxidase/metabolism , Neurotransmitter Agents/metabolism , Norepinephrine/metabolism , Norepinephrine/pharmacology , Plant Extracts , Rats , Rehmannia/chemistry , Serotonin/metabolism , Serotonin Plasma Membrane Transport Proteins/metabolism , Serotonin Plasma Membrane Transport Proteins/pharmacology , Stress, Psychological/drug therapy , Stress, Psychological/metabolism , Tandem Mass Spectrometry , Tryptophan Hydroxylase/metabolism
2.
Alcohol Clin Exp Res ; 46(8): 1384-1396, 2022 08.
Article in English | MEDLINE | ID: mdl-35791038

ABSTRACT

BACKGROUND: Individuals with alcohol use disorder (AUD) exhibit maladaptive responses of the hypothalamic-pituitary-adrenal (HPA) axis to stress, which has been linked to high rates of relapse to drinking among abstinent individuals. Corticotropin-releasing factor (CRF) parvocellular neuroendocrine cells (PNCs) within the paraventricular nucleus of the hypothalamus (PVN) are critical to stress-induced HPA axis activation. Here, we investigate sex differences in synaptic transmission and plasticity in PNCs following the application of the stress-associated neurotransmitter norepinephrine (NE) in a rat model of AUD. METHODS: Adult Sprague-Dawley rats were exposed to 40 days of chronic intermittent ethanol (CIE) vapor and 30 to 108 days of protracted withdrawal. We measured changes in holding current, evoked synaptic currents, and short-term glutamatergic plasticity (STP) in putative PNCs following the application of NE (10 µM) with and without the selective α1 adrenergic receptor (AR) antagonist prazosin (10 µM) or the α2AR antagonist atipamezole (10 µM). The experiments were performed using whole-cell patch clamp recordings in slices from CIE rats and air-exposed controls. RESULTS: NE application caused two distinct effects: a depolarizing, inward, postsynaptic current and a reduction in amplitude of an evoked glutamatergic excitatory postsynaptic current (eEPSC). Both effects were sex- and CIE-specific. Prazosin blocked the postsynaptic inward current, while atipamezole blocked the NE-mediated suppression of eEPSCs. Additionally, STP formation was facilitated following NE application only in stress-naïve males and this response was lost in stressed animals exposed to a 30-min restraint stress following CIE exposure. Furthermore, NE + prazosin restored STP formation in stressed CIE males. CONCLUSIONS: NE exerts excitatory and inhibitory effects on CRF PVN PNCs, and both effects are influenced by sex and CIE. Behavioral and hormonal responses to stress are influenced by STP formation within the PVN, which is lost following CIE and restored with the preapplication of prazosin. The selective blockade of α1AR may, therefore, ameliorate CIE-induced deficits in HPA responses to stress in a sex-specific manner.


Subject(s)
Alcoholism , Hypothalamo-Hypophyseal System , Animals , Corticotropin-Releasing Hormone/metabolism , Ethanol/toxicity , Female , Hypothalamo-Hypophyseal System/metabolism , Hypothalamus/metabolism , Male , Neuronal Plasticity , Norepinephrine/pharmacology , Paraventricular Hypothalamic Nucleus/metabolism , Pituitary-Adrenal System/metabolism , Prazosin/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Adrenergic, alpha , Sex Characteristics
3.
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
4.
Int J Med Mushrooms ; 24(4): 63-73, 2022.
Article in English | MEDLINE | ID: mdl-35695597

ABSTRACT

Our study aimed to investigate whether Agaricus brasiliensis water extract (AWE) possesses antidepressant activity. Depression as a result of chronic unpredictable mild stress (CUMS) was established in mice. The AWE group was administered 3.0 g/kg of AWE. The tail suspension test (TST) was conducted 1 h after the last administration. Then after fasting for 12 h, the mice were sacrificed by euthanasia and the brain and organs (liver, spleen, kidney, and thymus) were collected immediately. Biochemical indexes, including serotonin (5-HT), norepinephrine (NE), and dopamine (DA), were analyzed with biochemical reagent kits. In addition, 2,2-diphenyl-1-picrylhydrazyl free radical scavenging activity, inhibition of hydroxyl radical activity, and total antioxidant capacity were used to evaluate the antioxidant activity of AWE. The organ index analysis indicated that AWE had no adverse effect on mice at feeding time. The results suggested that AWE administration could significantly shorten the immobility time of mice in the TST. Particularly, the levels of 5-HT and NE appeared to increase significantly (P < 0.05) after AWE administration. At the same time, in vitro antioxidant experiments also revealed that AWE displayed better antioxidant activity. Collectively, these results suggest that AWE possesses good antidepressant activity, and these effects may be mediated by enhancing monoamine neurotransmitter content in the brain or antioxidant capacity to improve depression.


Subject(s)
Agaricus , Serotonin , Animals , Antidepressive Agents/chemistry , Antidepressive Agents/pharmacology , Antioxidants/pharmacology , Behavior, Animal , Depression/drug therapy , Disease Models, Animal , Mice , Norepinephrine/pharmacology , Serotonin/pharmacology , Water
5.
J Vasc Res ; 59(5): 303-313, 2022.
Article in English | MEDLINE | ID: mdl-35728582

ABSTRACT

INTRODUCTION: Adrenoceptor and endothelin (ET) receptor-mediated vasoconstriction as well as endothelium-dependent vasodilation of human saphenous veins were compared before and after 20 h of cold storage. METHODS: Contractile responses to potassium chloride (KCl), norepinephrine (NE), and ET-1 as well as vasodilator responses to acetylcholine (ACh) were evaluated. RESULTS: Storage in HEPES-supplemented Dulbecco's modified Eagle's medium (HDMEM) diminished KCl induced contractile forces to 71% (p = 0.002) and NE induced contractions to 80% (p = 0.037), in contrast to HEPES-supplemented Krebs-Henseleit solution (HKH) and TiProtec solution. KCl-normalized NE contractions were not affected by storage. NE EC50 values were slightly lower (7.1E-8 vs. 7.5E-8, p = 0.019) after storage in HKH, with no changes after storage in the other solutions. Endothelium-dependent responses to ACh were not affected by storage. ET-1 induced contractions were attenuated after storage in HDMEM (77%, p = 0.002), HKH (75%, p = 0.020), and TiProtec (73%, p = 0.010) with no changes in normalized constrictions. ET-1 EC50 values were not affected by storage. CONCLUSION: Loss of contractility after storage in HDMEM may reflect the lower content of dextrose. There was no specific attenuation of adrenoceptor, ET-receptor, or ACh receptor mediated signal transduction after storage in any of the media. HKH or TiProtec are equally suitable cold storage solutions for ex vivo measurements.


Subject(s)
Endothelium, Vascular , Receptors, Adrenergic , Receptors, Endothelin , Tissue Preservation , Vasoconstriction , Vasodilation , Humans , Acetylcholine/pharmacology , Endothelin-1/pharmacology , Endothelins/pharmacology , Endothelium , Endothelium, Vascular/physiopathology , Glucose/pharmacology , HEPES/pharmacology , Norepinephrine/pharmacology , Potassium Chloride/pharmacology , Receptors, Adrenergic/physiology , Receptors, Endothelin/physiology , Vasoconstriction/physiology , Vasodilation/physiology , Vasodilator Agents/pharmacology , Muscle Contraction/physiology , Tissue Preservation/methods , Cold Temperature/adverse effects , Receptors, Cholinergic/physiology
6.
Mol Cell Biochem ; 477(3): 663-672, 2022 Mar.
Article in English | MEDLINE | ID: mdl-34988854

ABSTRACT

Enhanced sympathetic system activation mediated by norepinephrine (NE) contributes to adverse cardiac remodeling leading to oxidative stress and cell death, progressing to heart failure. Natural antioxidants may help maintain redox balance, attenuating NE-mediated cardiac cell damage. In the present study, we evaluated the effect of a blueberry extract (BBE) on H9c2 cardiac cells exposed to NE on cell death, oxidative stress status and its major signaling pathways. H9c2 cells were pre-incubated with 50 µg/ml of BBE for 4 h and maintained in the presence of 100 µM NE for 24 h. NE exposure resulted in increased caspase 3/7 activity. This was associated with reduced protein expression of antioxidants catalase, superoxide dismutase and glutathione peroxidase and increase in 4-hydroxynonenal adduct formation. NE led to increased activity of Protein kinase B (Akt), Forkhead box O3a and AMP-activated protein kinase alpha and decreased activity of Signal transducer and activator of transcription 3. BBE prevented caspases activation and abrogated NE-induced increase in oxidative stress, as well as attenuated the increase in Akt. Based on these findings, it is concluded that BBE promoted cardioprotection of H9c2 cells in an in vitro model of NE-induced oxidative damage, suggesting a cardioprotective role for BBE in response to NE exposure.


Subject(s)
Apoptosis/drug effects , Blueberry Plants/chemistry , Myoblasts, Cardiac/metabolism , Norepinephrine/pharmacology , Oxidative Stress/drug effects , Plant Extracts/pharmacology , Animals , Cell Line , Plant Extracts/chemistry , Rats
7.
Mol Pharmacol ; 100(3): 203-216, 2021 09.
Article in English | MEDLINE | ID: mdl-34158361

ABSTRACT

Norepinephrine (NE) controls many vital body functions by activating adrenergic receptors (ARs). Average core body temperature (CBT) in mice is 37°C. Of note, CBT fluctuates between 36 and 38°C within 24 hours, but little is known about the effects of CBT changes on the pharmacodynamics of NE. Here, we used Peltier element-controlled incubators and challenged murine hypothalamic mHypoA -2/10 cells with temperature changes of ±1°C. We observed enhanced NE-induced activation of a cAMP-dependent luciferase reporter at 36 compared with 38°C. mRNA analysis and subtype specific antagonists revealed that NE activates ß 2- and ß 3-AR in mHypoA-2/10 cells. Agonist binding to the ß 2-AR was temperature insensitive, but measurements of cytosolic cAMP accumulation revealed an increase in efficacy of 45% ± 27% for NE and of 62% ± 33% for the ß 2-AR-selective agonist salmeterol at 36°C. When monitoring NE-promoted cAMP efflux, we observed an increase in the absolute efflux at 36°C. However, the ratio of exported to cytosolic accumulated cAMP is higher at 38°C. We also stimulated cells with NE at 37°C and measured cAMP degradation at 36 and 38°C afterward. We observed increased cAMP degradation at 38°C, indicating enhanced phosphodiesterase activity at higher temperatures. In line with these data, NE-induced activation of the thyreoliberin promoter was found to be enhanced at 36°C. Overall, we show that physiologic temperature changes fine-tune NE-induced cAMP signaling in hypothalamic cells via ß 2-AR by modulating cAMP degradation and the ratio of intra- and extracellular cAMP. SIGNIFICANCE STATEMENT: Increasing cytosolic cAMP levels by activation of G protein-coupled receptors (GPCR) such as the ß 2-adrenergic receptor (AR) is essential for many body functions. Changes in core body temperature are fundamental and universal factors of mammalian life. This study provides the first data linking physiologically relevant temperature fluctuations to ß 2-AR-induced cAMP signaling, highlighting a so far unappreciated role of body temperature as a modulator of the prototypic class A GPCR.


Subject(s)
Cyclic AMP/metabolism , Cytosol/metabolism , Receptors, Adrenergic, beta-2/physiology , 1-Methyl-3-isobutylxanthine/pharmacology , ARNTL Transcription Factors/metabolism , Aminopyridines/pharmacology , Animals , Cell Line , Cyclic AMP Response Element-Binding Protein/metabolism , Forkhead Transcription Factors/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/physiology , GTP-Binding Protein alpha Subunits, Gs/physiology , Hypothalamus/physiology , Mice , Neurons/physiology , Norepinephrine/pharmacology , Receptors, Adrenergic, beta-2/biosynthesis , Receptors, Adrenergic, beta-3/biosynthesis , Receptors, Adrenergic, beta-3/physiology , STAT Transcription Factors/metabolism , Salmeterol Xinafoate/pharmacology , Signal Transduction/physiology , Temperature , Thyrotropin-Releasing Hormone/genetics , Thyrotropin-Releasing Hormone/metabolism
8.
Sci Rep ; 10(1): 19358, 2020 11 09.
Article in English | MEDLINE | ID: mdl-33168874

ABSTRACT

5-HT inhibits cardiac sympathetic neurotransmission in normoglycaemic rats, via 5-HT1B, 5-HT1D and 5-HT5A receptor activation. Since type 1 diabetes impairs the cardiac sympathetic innervation leading to cardiopathies, this study aimed to investigate whether the serotonergic influence on cardiac noradrenergic control is altered in type 1 diabetic rats. Diabetes was induced in male Wistar rats by streptozotocin (50 mg/kg, i.p.). Four weeks later, the rats were anaesthetized, pithed and prepared for producing tachycardic responses by electrical preganglionic stimulation (C7-T1) of the cardioaccelerator sympathetic outflow or i.v. noradrenaline bolus injections. Immunohistochemistry was performed to study 5-HT1B, 5-HT1D and 5-HT5A receptor expression in the stellate ganglion from normoglycaemic and diabetic rats. In the diabetic group, i) i.v. continuous infusions of 5-HT induced a cardiac sympatho-inhibition that was mimicked by the 5-HT1/5A agonist 5-carboxamidotryptamine (without modifying noradrenaline-induced tachycardia), but not by the agonists indorenate (5-HT1A), CP 93,129 (5-HT1B), PNU 142633 (5-HT1D), or LY344864 (5-HT1F); ii) SB 699551 (5-HT5A antagonist; i.v.) completely reversed 5-CT-induced cardiac sympatho-inhibition; and iii) 5-HT5A receptors were more expressed in the stellate ganglion compared to normoglycaemic rats. These results show the prominent role of the peripheral 5-HT5A receptors prejunctionally inhibiting the cardiac sympathetic drive in type 1 diabetic rats.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Receptors, Serotonin/physiology , Sympathetic Nervous System/physiology , 5-Methoxytryptamine/analogs & derivatives , 5-Methoxytryptamine/pharmacology , Animals , Biphenyl Compounds/pharmacology , Carbazoles/pharmacology , Chromans/pharmacology , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Type 1/chemically induced , Diabetes Mellitus, Type 1/metabolism , Electric Stimulation Therapy , Fluorobenzenes/pharmacology , Immunohistochemistry , Male , Norepinephrine/pharmacology , Pyridines/pharmacology , Pyrroles/pharmacology , Rats , Rats, Wistar , Receptor, Serotonin, 5-HT1B/physiology , Receptor, Serotonin, 5-HT1D/physiology , Serotonin/analogs & derivatives , Serotonin/chemistry , Serotonin/metabolism
9.
Biomed Pharmacother ; 126: 110083, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32272432

ABSTRACT

Sepsis is a disease with high mortality rate worldwide and inducible nitric oxide (iNOS) induced vascular hyporeactivity plays a key role in it. There is no effective drug to treat vascular hyporeactivity specifically. Tubeimoside I (TBM) is a triterpenoid saponin isolated from Rhizoma Bolbostemmatis. In this study, we found that 4 mg/kg TBM intraperitoneally injected 1 h before cecal ligation and puncture (CLP) partially improved survival, ameliorated mean arterial pressure (MAP) and enhanced vascular responsiveness to norepinephrine (NE) and KCl in wild-type septic mice. CLP activated TLR4-MyD88-NF-κB-iNOS pathway was also inhibited by TBM both in vitro and in vivo. However, iNOS gene knockout counteracted the protection provided by TBM. We conclude that TBM protects mice in sepsis by reducing excessive NO production through inhibiting the TLR4-MyD88-NF-κB-iNOS pathway. Our study suggests a possible therapeutic application of TBM in sepsis.


Subject(s)
Drugs, Chinese Herbal/pharmacology , Gene Expression Regulation/drug effects , Nitric Oxide Synthase/genetics , Saponins/pharmacology , Sepsis/drug therapy , Sepsis/etiology , Triterpenes/pharmacology , Animals , Biomarkers , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Gene Expression , Male , Mesenteric Arteries/drug effects , Mesenteric Arteries/metabolism , Mice , Mice, Knockout , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Nitric Oxide/metabolism , Norepinephrine/pharmacology , Prognosis , Sepsis/mortality , Signal Transduction/drug effects , Treatment Outcome , Vasoconstrictor Agents/pharmacology
10.
Invest Ophthalmol Vis Sci ; 61(3): 26, 2020 03 09.
Article in English | MEDLINE | ID: mdl-32182332

ABSTRACT

Purpose: Elevated IOP can cause the development of glaucoma. The circadian rhythm of IOP depends on the dynamics of the aqueous humor and is synchronized with the circadian rhythm pacemaker, that is, the suprachiasmatic nucleus. The suprachiasmatic nucleus resets peripheral clocks via sympathetic nerves or adrenal glucocorticoids. However, the detailed mechanisms underlying IOP rhythmicity remain unclear. The purpose of this study was to verify this regulatory pathway. Methods: Adrenalectomy and/or superior cervical ganglionectomy were performed in C57BL/6J mice. Their IOP rhythms were measured under light/dark cycle and constant dark conditions. Ocular administration of corticosterone or norepinephrine was also performed. Localization of adrenergic receptors, glucocorticoid receptors, and clock proteins Bmal1 and Per1 were analyzed using immunohistochemistry. Period2::luciferase rhythms in the cultured iris/ciliary bodies of adrenalectomized and/or superior cervical ganglionectomized mice were monitored to evaluate the effect of the procedures on the local clock. The IOP rhythm of retina and ciliary epithelium-specific Bmal1 knockout mice were measured to determine the significance of the local clock. Results: Adrenalectomy and superior cervical ganglionectomy disrupted IOP rhythms and the circadian clock in the iris/ciliary body cultures. Instillation of corticosterone and norepinephrine restored the IOP rhythm. ß2-Adrenergic receptors, glucocorticoid receptors, and clock proteins were strongly expressed within the nonpigmented epithelia of the ciliary body. However, tissue-specific Bmal1 knock-out mice maintained their IOP rhythm. Conclusions: These findings suggest direct driving of the IOP rhythm by the suprachiasmatic nucleus, via the dual corticosterone and norepinephrine pathway, but not the ciliary clock, which may be useful for chronotherapy of glaucoma.


Subject(s)
Circadian Rhythm/physiology , Corticosterone/pharmacology , Intraocular Pressure/physiology , Norepinephrine/pharmacology , Sympathetic Nervous System/physiology , ARNTL Transcription Factors/metabolism , Administration, Ophthalmic , Adrenalectomy , Animals , Cells, Cultured , Ciliary Body/drug effects , Ciliary Body/metabolism , Circadian Rhythm/drug effects , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Ganglionectomy , Immunohistochemistry , Iris/drug effects , Iris/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Period Circadian Proteins/metabolism , Photoperiod , Receptors, Adrenergic, beta-2/metabolism , Receptors, Glucocorticoid/metabolism , Superior Cervical Ganglion/surgery , Tonometry, Ocular
11.
Nat Prod Res ; 34(23): 3369-3372, 2020 Dec.
Article in English | MEDLINE | ID: mdl-30698035

ABSTRACT

Oxalis pes-caprae L. is a plant of the Oxalidaceae family, from which several compounds have been previously identified. Recently, we showed that an Oxalis pes-caprae L. extract inhibits the vasopressor effect of noradrenaline. In this work we aimed to explore the mechanisms involved in this effect. The results confirmed that the flavonoid fraction present in the extract inhibits noradrenaline-induced contractions and that this effect is concentration-dependent. Also, a parallel shift to the right in the noradrenaline concentration-response curve was observed, suggesting a decrease in efficacy and also in potency. Together these results support the assumption that the extract could exert a non-competitive antagonism on the α-adrenergic receptors. However, experiments in the presence of competitive antagonists for α-adrenergic receptor sub-types (i.e. prazosin, yohimbine and phentolamine) showed that the effect may not be directly mediated by α-adrenergic receptors. Thus, the interaction of this extract with the adrenergic system remains to be confirmed.


Subject(s)
Oxalidaceae/chemistry , Plant Extracts/pharmacology , Polyphenols/analysis , Thoracic Arteries/drug effects , Vasoconstriction/drug effects , Adrenergic alpha-Agonists/pharmacology , Adrenergic alpha-Antagonists/pharmacology , Adult , Aged , Aged, 80 and over , Dose-Response Relationship, Drug , Humans , Middle Aged , Norepinephrine/pharmacology , Plant Extracts/chemistry , Plant Leaves/chemistry , Prazosin/pharmacology , Receptors, Adrenergic, alpha/drug effects , Receptors, Adrenergic, alpha/metabolism , Yohimbine/pharmacology
12.
Chem Biol Interact ; 316: 108923, 2020 Jan 25.
Article in English | MEDLINE | ID: mdl-31838051

ABSTRACT

Angina pectoris can be used as an early warning for coronary artery disease. Vasodilation is an important mechanism of angina pectoris. Traditional Chinese medicine - Compound Danshen Dripping Pill (CDDP) is widely used to improve the symptoms of cardiovascular diseases (CVDs). To investigate the influence of vasodilation effect and underlying mechanisms of CDDP, we determined the vasodilation effect of thoracic aorta ring on rat induced by norepinephrine (NE). Then targets-fishing method was used to predict the potential mechanism of CDDP on vasodilation, based on the structures of the main components. Then, iTRAQ-based quantitative proteomics analysis was used for verification of the candidate target proteins and pathways to illustrate the underlying mechanisms. Furthermore, the differentially expressed proteins in the enriched pathways were validated by western blotting. In this study, we found that CDDP could significantly inhibit NE induced aortic contraction tension, and the mechanism may be related to platelet activation, cGMP - PKG signaling pathway and vascular smooth muscle contraction. The method provides a new way to uncover the vasodilation mechanism of CDDP, as well as other multi-component herbal medicines.


Subject(s)
Drugs, Chinese Herbal/pharmacology , Proteome/analysis , Proteomics , Vasodilator Agents/pharmacology , Animals , Aorta, Thoracic/drug effects , Aorta, Thoracic/metabolism , Camphanes , Cyclic GMP/metabolism , Cyclic GMP-Dependent Protein Kinases/metabolism , Male , Medicine, Chinese Traditional , Muscle Contraction/drug effects , Norepinephrine/pharmacology , Panax notoginseng , Proteome/drug effects , Proteome/metabolism , Rats , Rats, Sprague-Dawley , Salvia miltiorrhiza , Signal Transduction/drug effects
13.
Cell Rep ; 29(13): 4349-4361.e4, 2019 12 24.
Article in English | MEDLINE | ID: mdl-31875545

ABSTRACT

In addition to their support role in neurotransmitter and ion buffering, astrocytes directly regulate neurotransmission at synapses via local bidirectional signaling with neurons. Here, we reveal a form of neuronal-astrocytic signaling that transmits retrograde dendritic signals to distal upstream neurons in order to activate recurrent synaptic circuits. Norepinephrine activates α1 adrenoreceptors in hypothalamic corticotropin-releasing hormone (CRH) neurons to stimulate dendritic release, which triggers an astrocytic calcium response and release of ATP; ATP stimulates action potentials in upstream glutamate and GABA neurons to activate recurrent excitatory and inhibitory synaptic circuits to the CRH neurons. Thus, norepinephrine activates a retrograde signaling mechanism in CRH neurons that engages astrocytes in order to extend dendritic volume transmission to reach distal presynaptic glutamate and GABA neurons, thereby amplifying volume transmission mediated by dendritic release.


Subject(s)
Adrenergic alpha-Agonists/pharmacology , Astrocytes/drug effects , Dendrites/drug effects , GABAergic Neurons/drug effects , Norepinephrine/pharmacology , Synaptic Transmission/drug effects , Action Potentials/drug effects , Action Potentials/physiology , Animals , Astrocytes/metabolism , Astrocytes/ultrastructure , Cell Communication , Channelrhodopsins/genetics , Channelrhodopsins/metabolism , Corticotropin-Releasing Hormone/genetics , Corticotropin-Releasing Hormone/metabolism , Dendrites/metabolism , Dendrites/ultrastructure , GABAergic Neurons/metabolism , GABAergic Neurons/ultrastructure , Gene Expression Regulation , Glutamic Acid/metabolism , Glutamic Acid/pharmacology , Hypothalamus/drug effects , Hypothalamus/metabolism , Hypothalamus/ultrastructure , Male , Mice , Mice, Transgenic , Microtomy , Receptors, Corticotropin/genetics , Receptors, Corticotropin/metabolism , Synapses/drug effects , Synapses/physiology , Synaptic Transmission/physiology , Tissue Culture Techniques , gamma-Aminobutyric Acid/metabolism , gamma-Aminobutyric Acid/pharmacology
14.
Kidney Int ; 96(5): 1150-1161, 2019 11.
Article in English | MEDLINE | ID: mdl-31530477

ABSTRACT

Norepinephrine exacerbates renal medullary hypoxia in experimental septic acute kidney injury. Here we examined whether dexmedetomidine, an α2-adrenergic agonist, can restore vasopressor responsiveness, decrease the requirement for norepinephrine and attenuate medullary hypoxia in ovine gram-negative sepsis. Sheep were instrumented with pulmonary and renal artery flow probes, and laser Doppler and oxygen-sensing probes in the renal cortex and medulla. Conscious sheep received an infusion of live Escherichia coli for 30 hours. Eight sheep in each group were randomized to receive norepinephrine, norepinephrine with dexmedetomidine, dexmedetomidine alone or saline vehicle, from 24-30 hours of sepsis. Sepsis significantly reduced the average mean arterial pressure (84 to 67 mmHg), average renal medullary perfusion (1250 to 730 perfusion units), average medullary tissue pO2 (40 to 21 mmHg) and creatinine clearance (2.50 to 0.78 mL/Kg/min). Norepinephrine restored baseline mean arterial pressure (to 83 mmHg) but worsened medullary hypoperfusion (to 330 perfusion units) and medullary hypoxia (to 9 mmHg). Dexmedetomidine (0.5 µg/kg/h) co-administration significantly reduced the norepinephrine dose (0.8 to 0.4 µg/kg/min) required to restore baseline mean arterial pressure, attenuated medullary hypoperfusion (to 606 perfusion units), decreased medullary tissue hypoxia (to 29 mmHg), and progressively increased creatinine clearance (to 1.8 mL/Kg/min). Compared with vehicle time-control, dexmedetomidine given alone significantly prevented the temporal reduction in mean arterial pressure, but had no significant effects on medullary perfusion and oxygenation or creatinine clearance. Thus, in experimental septic acute kidney injury, dexmedetomidine reduced norepinephrine requirements, attenuated its adverse effects on the renal medulla, and maintained renal function.


Subject(s)
Acute Kidney Injury/drug therapy , Adrenergic alpha-2 Receptor Agonists/therapeutic use , Adrenergic alpha-Agonists/therapeutic use , Dexmedetomidine/therapeutic use , Norepinephrine/therapeutic use , Acute Kidney Injury/blood , Acute Kidney Injury/etiology , Adrenergic alpha-2 Receptor Agonists/pharmacology , Adrenergic alpha-Agonists/pharmacology , Animals , Cytokines/blood , Dexmedetomidine/pharmacology , Drug Evaluation, Preclinical , Escherichia coli , Hemodynamics/drug effects , Kidney/drug effects , Kidney/metabolism , Norepinephrine/pharmacology , Oxygen/metabolism , Sepsis/complications , Sheep
15.
Biomolecules ; 9(6)2019 06 11.
Article in English | MEDLINE | ID: mdl-31212721

ABSTRACT

Despite pharmacotherapeutic advances, cardiovascular disease (CVD) remains the primary cause of global mortality. Alternative approaches, such as herbal medicine, continue to be sought to reduce this burden. Origanum majorana is recognized for many medicinal values, yet its vasculoprotective effects remain poorly investigated. Here, we subjected rat thoracic aortae to increasing doses of an ethanolic extract of Origanummajorana (OME). OME induced relaxation in a dose-dependent manner in endothelium-intact rings. This relaxation was significantly blunted in denuded rings. N(ω)-nitro-l-arginine methyl ester (L-NAME) or 1H-[1,2,4]oxadiazolo[4,3,-a]quinoxalin-1-one (ODQ) significantly reduced the OME-induced vasorelaxation. Cyclic guanosine monophosphate (cGMP) levels were also increased by OME. Moreover, wortmannin or LY294002 significantly reduced OME-induced vasorelaxation. Blockers of ATP-sensitive or Ca2+-activated potassium channels such as glibenclamide or tetraethylamonium (TEA), respectively, did not significantly affect OME-induced relaxation. Similarly, verapamil, a Ca2+ channel blocker, indomethacin, a non-selective cyclooxygenase inhibitor, and pyrilamine, a H1 histamine receptor blocker, did not significantly modulate the observed relaxation. Taken together, our results show that OME induces vasorelaxation via an endothelium-dependent mechanism involving the phosphoinositide 3-kinase (PI3-K)/ endothelial nitric oxide (NO) synthase (eNOS)/cGMP pathway. Our findings further support the medicinal value of marjoram and provide a basis for its beneficial intake. Although consuming marjoram may have an antihypertensive effect, further studies are needed to better determine its effects in different vascular beds.


Subject(s)
Aorta, Thoracic/drug effects , Aorta, Thoracic/physiology , Cyclic GMP/metabolism , Nitric Oxide Synthase Type III/metabolism , Origanum/chemistry , Phosphatidylinositol 3-Kinase/metabolism , Plant Extracts/pharmacology , Animals , Aorta, Thoracic/cytology , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Male , Norepinephrine/pharmacology , Potassium Channel Blockers/pharmacology , Prostaglandin-Endoperoxide Synthases/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Vasoconstriction/drug effects , Vasodilation/drug effects , Vasodilator Agents/pharmacology
16.
Biol Pharm Bull ; 42(6): 1048-1053, 2019.
Article in English | MEDLINE | ID: mdl-31155582

ABSTRACT

Previous research has indicated that high insulin affects vascular function. Equol is an active metabolite of daidzein, an isoflavone produced from soy by intestinal microbial flora, with beneficial effects on the vascular system. This study investigated whether equol was beneficial for vascular function under high insulin conditions. Using organ culture techniques, rat carotid arteries were treated for 23 ± 1 h with a vehicle, high insulin (100 nM), or equol (100 µM) plus high insulin (100 nM). Vascular isometric forces were measured by the organ bath technique. In each endothelium-intact ring, the contractions induced by high-K+, noradrenaline, or by serotonin (5-HT) were similar for the vehicle, insulin, and equol + insulin treatments. Contractions induced by a selective 5-HT2A receptor agonist (TCB2) increased with insulin treatment (vs. vehicle), but less so with equol + insulin. Under basal conditions, a selective 5-HT2B receptor agonist (BW723C86) did not induce contraction; following precontraction by a thromboxane analog, it induced contraction but not relaxation. These responses were similar across the three treatments. Acetylcholine-induced relaxations were also similar for the three treatments. In the endothelium-denuded preparations, 5-HT-induced contraction was augmented with insulin treatment (vs. vehicle) but less so by equol + insulin treatment. These differences in 5-HT-induced contractions were eliminated by iberiotoxin, a large-conductance calcium-activated K+ channel (BKCa) inhibitor. These results suggest that equol exerts a preventive effect on the enhancement of 5-HT-induced contraction by high insulin (possibly mediated by the 5-HT2A receptor), and that these effects may be attributed to the activation of BKCa channels in vascular smooth muscle.


Subject(s)
Carotid Arteries/drug effects , Equol/pharmacology , Insulin/pharmacology , Vasoconstriction/drug effects , Animals , Carotid Arteries/physiology , Large-Conductance Calcium-Activated Potassium Channels/physiology , Male , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/physiology , Norepinephrine/pharmacology , Phytoestrogens/pharmacology , Potassium/pharmacology , Rats, Wistar , Serotonin/pharmacology
17.
Clin Toxicol (Phila) ; 57(11): 1073-1079, 2019 Nov.
Article in English | MEDLINE | ID: mdl-30806099

ABSTRACT

Context: Although cerebral perfusion (CP) is preserved across a wide range of mean arterial pressures (MAP) through cerebral-vascular autoregulation, the relationship between MAP and CP in refractory poison-induced cardiogenic shock (PICS) has never been studied. We compared the effects of therapies used in PICS: high-dose insulin (HDI), HDI plus norepinephrine (NE), and vasopressors alone (NE plus epinephrine (Epi)) on cerebral tissue oxygenation (PtO2). Methods: Fifteen swine were randomized to either HDI, HDI + NE, or NE + Epi. All animals received a propranolol infusion using an established model of toxicity. At primary toxicity (P1), defined as a 25% reduction in heart rate (HR) multiplied by MAP, the HDI and HDI + NE groups received HDI and the NE + Epi group received NE. Once a sustained MAP < 55 mmHg was reached (P2), the HDI group received saline (NS), the HDI + NE group received NE and the NE + Epi group received Epi until death or censoring. PtO2 and hemodynamic parameters including MAP, cardiac output (CO) and central venous pressure (CVP) were measured every 10 minutes. Glucose and potassium were measured at predetermined intervals. Results: Animals treated with HDI + NE maintained PtO2 over time more than the HDI-alone group. Due to rapid hemodynamic collapse, we were unable to analyze PtO2 data in the vasopressor only animals. Mean survival time was 1.9, 2.9 and 0.1 hours for the HDI, HDI + NE and NE + Epi groups, respectively. Survival time from P2 (sustained MAP <55 mmHg) to death or censoring was not different between HDI and HDI + NE groups. Conclusions: HDI + NE treatment was superior to HDI-alone at preserving PtO2 when MAP < 55 mmHg. We were unable to compare the PtO2 between the NE + Epi to the HDI or HDI + NE due to rapid decline in CO and death. If MAP is sustained at < 55 mmHg after maximizing HDI, adjunctive treatment with NE should be considered to preserve PtO2.


Subject(s)
Insulin/administration & dosage , Propranolol/toxicity , Shock, Cardiogenic/drug therapy , Vasoconstrictor Agents/pharmacology , Adrenergic beta-Antagonists/adverse effects , Animals , Arterial Pressure/drug effects , Disease Models, Animal , Drug Therapy, Combination , Epinephrine/pharmacology , Kaplan-Meier Estimate , Norepinephrine/pharmacology , Oxygen/metabolism , Random Allocation , Shock, Cardiogenic/chemically induced , Shock, Cardiogenic/mortality , Swine , Time Factors
18.
Clin Exp Pharmacol Physiol ; 46(5): 427-434, 2019 05.
Article in English | MEDLINE | ID: mdl-30697784

ABSTRACT

Many hypertensive animal models have been developed and used to elucidate the pathophysiology of hypertension and to develop antihypertensive drugs. Among them, the spontaneous hypertensive rat (SHR), deoxycorticosterone acetate (DOCA)-treated and high salt intake rat (DOCA-salt), and high sodium-fed Dahl salt-sensitive rat (HS) models are commonly used. Multiple studies have been conducted, however, elevation in blood pressure in these models due to the reactivity of adrenergic vasoconstriction has not been well characterized in a centralized experiment. In this study, the pressor responses to periarterial nerve stimulation (PNS) or exogenous noradrenaline (NA) infusion were measured in the isolated mesenteric vascular bed with the intestinal tract to investigate the reactivity of mesenteric adrenergic vasoconstriction. The systemic arterial blood pressure of the hypertensive rat models was uniformly elevated compared with their respective controls. However, the changes in perfusion pressure in the mesenteric vascular bed in response to PNS and exogenous NA infusion were quite different depending on the model. The pressor responses to PNS in SHRs and Dahl S HS rats were significantly higher, and those in DOCA-salt rats were significantly lower than those in the controls. The pressor responses to exogenous NA infusion in SHRs were significantly higher, and those in Dahl S HS rats were significantly lower than those in their respective controls. No difference was observed in the pressor responses to the exogenous NA between the DOCA-salt and sham groups. These results demonstrate that the reactivity of adrenergic vasoconstriction is different for each type of experimental hypertensive model rat.


Subject(s)
Electric Stimulation Therapy , Hypertension/therapy , Intestines/blood supply , Mesentery/drug effects , Norepinephrine/pharmacology , Animals , Blood Pressure/drug effects , Disease Models, Animal , Hypertension/drug therapy , Hypertension/physiopathology , Male , Mesentery/physiopathology , Norepinephrine/administration & dosage , Norepinephrine/therapeutic use , Rats
19.
J Vet Med Sci ; 81(1): 35-41, 2019 Jan 08.
Article in English | MEDLINE | ID: mdl-30429409

ABSTRACT

Eukaryotic elongation factor 2 (eEF2) kinase (eEF2K) inhibits protein translation through the phosphorylation of its specific substrate, eEF2. We previously demonstrated that eEF2K expression increases in superior mesenteric artery from spontaneously hypertensive rats (SHR) and that eEF2K mediates development of hypertension in SHR. In addition, we recently revealed that A484954, a selective eEF2K inhibitor induced relaxation via opening smooth muscle inward rectifier K+ (Kir) channel in rat isolated superior mesenteric artery. Here, we further examined the effects of A484954 on contractility and blood pressure (BP) in rats. Isometric contraction of rat isolated superior mesenteric artery was measured. BP was measured by a carotid cannulation method. A484954 (10 µM) inhibited noradrenaline (NA)-induced contraction in a biphasic manner (magnitude of inhibition higher at high dose NA). A484954 also inhibited an α1-receptor agonist, phenylephrine-induced contraction, while it was not biphasic. Specifically, a ß-receptor antagonist, propranolol (1 µM) prevented the A484954-mediated inhibition of NA (high-dose)-induced contraction. A484954 (10 µM) potentiated a ß-receptor agonist, isoproterenol-induced relaxation, which was completely prevented by BaCl2 (1 mM), a Kir channel blocker. In vivo, A484954 (122 µg/kg) inhibited NA-induced increase of BP in rats. Another eEF2K inhibitor, NH125 (22 µg/kg) also inhibited the NA-induced BP increase in rats. In summary, it was concluded that A484954 lowers NA-induced BP rise perhaps through activation of ß2-receptor-Kir channel and subsequent vasorelaxation via inhibiting eEF2K activity.


Subject(s)
Blood Pressure/drug effects , Cyclopropanes/pharmacology , Elongation Factor 2 Kinase/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , Pyrrolidines/pharmacology , Animals , Isometric Contraction/drug effects , Male , Norepinephrine/pharmacology , Potassium Channels, Inwardly Rectifying/metabolism , Pyridines , Rats, Inbred SHR , Rats, Wistar , Vasodilation/drug effects
20.
Bull Exp Biol Med ; 165(5): 597-601, 2018 Sep.
Article in English | MEDLINE | ID: mdl-30225714

ABSTRACT

Single injection of muscarinic cholinoceptor blocker atropine (1 mg/kg) to outbred male rats reduced ß-adrenergic responsiveness of erythrocytes (by 2.2 times) and the content of epinephrine granules on erythrocytes (by 1.5 times), significantly increased HR and rigidity of the heart rhythm, and manifold decreased the power of all spectral components of heart rhythm variability. Stimulation of the central neurotransmitter systems increased ß-adrenergic responsiveness of erythrocytes (by 15-26%), decreased the number of epinephrine granules on erythrocytes (by 25-40%), and increased HR and cardiac rhythm intensity. These changes were most pronounced after stimulation of the serotoninergic system. Administration of atropine against the background of activation of central neurotransmitter systems did not decrease ß-adrenergic responsiveness of erythrocytes (this parameter remained at a stably high level and even increased during stimulation of the dopaminergic system), but decreased the number of epinephrine granules on erythrocytes, increased HR, and dramatically decreased the power of all components of heart rhythm variability spectrum. The response to atropine was maximum against the background of noradrenergic system activation and less pronounced during stimulation of the serotoninergic system. Thus, substances that are complementary to cholinergic receptors modulated adrenergic effect on the properties of red blood cells, which, in turn, can modulate the adrenergic influences on the heart rhythm via the humoral channel of regulation. Stimulation of central neurotransmitter systems that potentiates the growth of visceral adrenergic responsiveness weakens the cholinergic modulation of the adrenergic influences, especially with respect to erythrocyte responsiveness. Hence, changes in the neurotransmitter metabolism in the body can lead to coupled modulation of reception and reactivity to adrenergic- and choline-like regulatory factors at the level of erythrocyte membranes, which can be important for regulation of heart rhythm.


Subject(s)
Atropine/pharmacology , Erythrocytes/drug effects , Heart Rate/drug effects , Heart/drug effects , Muscarinic Antagonists/pharmacology , Receptors, Muscarinic/metabolism , Acetylcholine/pharmacology , Adrenergic Uptake Inhibitors/pharmacology , Adrenergic alpha-Agonists/pharmacology , Animals , Animals, Outbred Strains , Cardiotonic Agents/pharmacology , Cholinergic Agonists/pharmacology , Dopamine/pharmacology , Epinephrine/pharmacology , Erythrocytes/metabolism , Heart/physiology , Heart Rate/physiology , Maprotiline/pharmacology , Norepinephrine/pharmacology , Rats
SELECTION OF CITATIONS
SEARCH DETAIL