Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29.012
Filter
1.
Life Sci ; 348: 122695, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38710285

ABSTRACT

AIMS: To evaluate the basal release of 6-nitrodopamine (6-ND) from human isolated seminal vesicles (HISV) and to characterize its action and origin. MAIN METHODS: Left HISV obtained from patients undergoing prostatectomy surgery was suspended in a 3-mL organ bath containing warmed (37 °C) and gassed (95%O2:5%CO2) Krebs-Henseleit's solution (KHS) with ascorbic acid. An aliquot of 2 mL of the supernatant was used to quantify catecholamines by LC-MS/MS. For functional studies, concentration-responses curves to catecholamines were obtained, and pEC50 and Emax values were calculated. Detection of tyrosine hydroxylase and S100 protein were also carried out by both immunohistochemistry and fluorescence in-situ hybridization assays (FISH). KEY FINDINGS: Basal release of 6-ND was higher than the other catecholamines (14.76 ± 14.54, 4.99 ± 6.92, 3.72 ± 4.35 and 5.13 ± 5.76 nM for 6-ND, noradrenaline, adrenaline, and dopamine, respectively). In contrast to the other catecholamines, the basal release of 6-ND was not affected by the sodium current (Nav) channel inhibitor tetrodotoxin (1 µM; 10.4 ± 8.9 and 10.4 ± 7.9 nM, before and after tetrodotoxin, respectively). All the catecholamines produced concentration-dependent HISV contractions (pEC50 4.1 ± 0.2, 4.9 ± 0.3, 5.0 ± 0.3, and 3.9 ± 0.8 for 6-ND, noradrenaline, adrenaline, and dopamine, respectively), but 6-ND was 10-times less potent than noradrenaline and adrenaline. However, preincubation with very low concentration of 6-ND (10-8 M, 30 min) produced significant leftward shifts of the concentration-response curves to noradrenaline. Immunohistochemical and FISH assays identified tyrosine hydroxylase in tissue epithelium of HISV strips. SIGNIFICANCE: Epithelium-derived 6-ND is the major catecholamine released from human isolated seminal vesicles and that modulates smooth muscle contractility by potentiating noradrenaline-induced contractions.


Subject(s)
Dopamine , Norepinephrine , Seminal Vesicles , Humans , Male , Norepinephrine/pharmacology , Norepinephrine/metabolism , Seminal Vesicles/drug effects , Seminal Vesicles/metabolism , Dopamine/metabolism , Dopamine/pharmacology , Middle Aged , Epithelium/metabolism , Epithelium/drug effects , Muscle Contraction/drug effects , Aged , Catecholamines/metabolism
2.
Virulence ; 15(1): 2350904, 2024 12.
Article in English | MEDLINE | ID: mdl-38725098

ABSTRACT

Fusobacterium nucleatum (F. nucleatum) is closely correlated with tumorigenesis in colorectal cancer (CRC). We aimed to investigate the effects of host norepinephrine on the carcinogenicity of F. nucleatum in CRC and reveal the underlying mechanism. The results revealed that both norepinephrine and bacterial quorum sensing (QS) molecule auto-inducer-2 (AI-2) were positively associated with the progression of F. nucleatum related CRC (p < 0.01). In vitro studies, norepinephrine induced upregulation of QS-associated genes and promoted the virulence and proliferation of F. nucleatum. Moreover, chronic stress significantly increased the colon tumour burden of ApcMin/+ mice infected with F. nucleatum (p < 0.01), which was decreased by a catecholamine inhibitor (p < 0.001). Our findings suggest that stress-induced norepinephrine may promote the progression of F. nucleatum related CRC via bacterial QS signalling. These preliminary data provide a novel strategy for the management of pathogenic bacteria by targeting host hormones-bacterial QS inter-kingdom signalling.


Subject(s)
Colorectal Neoplasms , Fusobacterium nucleatum , Norepinephrine , Quorum Sensing , Signal Transduction , Quorum Sensing/drug effects , Fusobacterium nucleatum/pathogenicity , Fusobacterium nucleatum/drug effects , Fusobacterium nucleatum/physiology , Animals , Colorectal Neoplasms/microbiology , Norepinephrine/pharmacology , Mice , Humans , Disease Progression , Fusobacterium Infections/microbiology , Virulence , Homoserine/analogs & derivatives , Homoserine/metabolism , Mice, Inbred C57BL , Male , Lactones
3.
Nat Commun ; 15(1): 3834, 2024 May 07.
Article in English | MEDLINE | ID: mdl-38714741

ABSTRACT

Sleep disorders increase the risk and mortality of heart disease, but the brain-heart interaction has not yet been fully elucidated. Cuproptosis is a copper-dependent type of cell death activated by the excessive accumulation of intracellular copper. Here, we showed that 16 weeks of sleep fragmentation (SF) resulted in elevated copper levels in the male mouse heart and exacerbated myocardial ischemia-reperfusion injury with increased myocardial cuproptosis and apoptosis. Mechanistically, we found that SF promotes sympathetic overactivity, increases the germination of myocardial sympathetic nerve terminals, and increases the level of norepinephrine in cardiac tissue, thereby inhibits VPS35 expression and leads to impaired ATP7A related copper transport and copper overload in cardiomyocytes. Copper overload further leads to exacerbated cuproptosis and apoptosis, and these effects can be rescued by excision of the sympathetic nerve or administration of copper chelating agent. Our study elucidates one of the molecular mechanisms by which sleep disorders aggravate myocardial injury and suggests possible targets for intervention.


Subject(s)
Apoptosis , Copper , Mice, Inbred C57BL , Myocardial Reperfusion Injury , Myocytes, Cardiac , Sleep Deprivation , Animals , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Male , Copper/metabolism , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Mice , Sleep Deprivation/physiopathology , Sleep Deprivation/metabolism , Sleep Deprivation/complications , Copper-Transporting ATPases/metabolism , Copper-Transporting ATPases/genetics , Norepinephrine/metabolism , Norepinephrine/pharmacology , Myocardium/metabolism , Myocardium/pathology , Sympathetic Nervous System/metabolism , Disease Models, Animal
4.
Int J Mol Sci ; 25(8)2024 Apr 17.
Article in English | MEDLINE | ID: mdl-38673988

ABSTRACT

In Parkinson's disease (PD), along with typical motor dysfunction, abnormal breathing is present; the cause of which is not well understood. The study aimed to analyze the effects of stimulation of the serotonergic system with 5-HT1A and 5-HT2A agonists in a model of PD induced by injection of 6-hydroxydopamine (6-OHDA). To model PD, bilateral injection of 6-OHDA into both striata was performed in male Wistar rats. Respiratory disturbances in response to 7% hypercapnia (CO2 in O2) in the plethysmographic chamber before and after stimulation of the serotonergic system and the incidence of apnea were studied in awake rats 5 weeks after 6-OHDA or vehicle injection. Administration of 6-OHDA reduced the concentration of serotonin (5-HT), dopamine (DA) and norepinephrine (NA) in the striatum and the level of 5-HT in the brainstem of treated rats, which have been associated with decreased basal ventilation, impaired respiratory response to 7% CO2 and increased incidence of apnea compared to Sham-operated rats. Intraperitoneal (i.p.) injection of the 5-HT1AR agonist 8-OH-DPAT and 5-HT2AR agonist NBOH-2C-CN increased breathing during normocapnia and hypercapnia in both groups of rats. However, it restored reactivity to hypercapnia in 6-OHDA group to the level present in Sham rats. Another 5-HT2AR agonist TCB-2 was only effective in increasing normocapnic ventilation in 6-OHDA rats. Both the serotonergic agonists 8-OH-DPAT and NBOH-2C-CN had stronger stimulatory effects on respiration in PD rats, compensating for deficits in basal ventilation and hypercapnic respiration. We conclude that serotonergic stimulation may have a positive effect on respiratory impairments that occur in PD.


Subject(s)
Hypercapnia , Parkinson Disease , Receptor, Serotonin, 5-HT1A , Receptor, Serotonin, 5-HT2A , Animals , Male , Rats , Disease Models, Animal , Dopamine/metabolism , Hypercapnia/metabolism , Hypercapnia/physiopathology , Norepinephrine/metabolism , Norepinephrine/pharmacology , Oxidopamine/pharmacology , Parkinson Disease/metabolism , Rats, Wistar , Receptor, Serotonin, 5-HT1A/metabolism , Receptor, Serotonin, 5-HT2A/metabolism , Respiration/drug effects , Serotonin/metabolism , Serotonin 5-HT1 Receptor Agonists/pharmacology , Serotonin 5-HT2 Receptor Agonists/pharmacology
5.
Am J Physiol Cell Physiol ; 326(5): C1334-C1344, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38557356

ABSTRACT

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


Subject(s)
Myocytes, Cardiac , Propranolol , Signal Transduction , Animals , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/drug effects , Mice , Propranolol/pharmacology , Receptors, Adrenergic, beta/metabolism , Mice, Inbred C57BL , Isoproterenol/pharmacology , Male , Heart/drug effects , Cells, Cultured , Adrenergic beta-Agonists/pharmacology , Norepinephrine/metabolism , Norepinephrine/pharmacology , Adrenergic beta-Antagonists/pharmacology
6.
Aging Cell ; 23(4): e14087, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38332648

ABSTRACT

Age-related dysfunctions in specific neurotransmitter systems likely play an important role in cognitive decline even in its most subtle forms. Therefore, preservation or improvement of cognition via augmentation of neurotransmission is a potential therapeutic strategy to prevent further cognitive deficits. Here we identified a particular neuronal vulnerability in the aged Fischer 344 rat brain, an animal model of neurocognitive aging. Specifically, we demonstrated a marked impairment in glutamate-stimulated release of norepinephrine (NE) in the hippocampus and cerebral cortex of aged rats, and established that this release was mediated by N-methyl-D-aspartate (NMDA) receptors. Further, we also demonstrated that this decrease in NE release is fully rescued by the psychostimulant drug amphetamine (AMPH). Moreover, we showed that AMPH increases dendritic spine maturation, and importantly shows preclinical efficacy in restoring memory deficits in the aged rat through its actions to potentiate NE neurotransmission at ß-adrenergic receptors. Taken together, our results suggest that deficits in glutamate-stimulated release of NE may contribute to and possibly be a determinant of neuronal vulnerability underlying cognitive decline during aging, and that these deficits can be corrected with currently available drugs. Overall these studies suggest that repurposing of psychostimulants for age-associated cognitive deficits is a potential avenue to delay or prevent cognitive decline and/or frank dementia later in life.


Subject(s)
Amphetamine , Central Nervous System Stimulants , Rats , Animals , Amphetamine/pharmacology , Norepinephrine/pharmacology , Rats, Sprague-Dawley , Dendritic Spines/metabolism , Central Nervous System Stimulants/pharmacology , Cerebral Cortex/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Hippocampus/metabolism , Rats, Inbred F344 , Glutamic Acid , Cognition
7.
Hypertens Res ; 47(4): 1017-1023, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38337004

ABSTRACT

Increased blood pressure variability (BPV) was shown to be associated with cardiovascular morbidities and/or mortalities. There are various types of BPV depending on time intervals of BP measurements, ranging from beat-to-beat to visit-to-visit or year-to-year. We previously found that continuous infusion of noradrenaline (NA) for 14 days increased short-term BPV every 15 min in rats. The aims of this study were to examine (1) whether NA infusion increases very short-term beat-to-beat BPV, (2) the effects of azelnidipine and hydralazine on NA-induced BPV, and (3) whether baroreceptor reflex sensitivity (BRS) is affected by NA or NA plus those vasodilators. Nine-week-old Wistar rats infused subcutaneously with 30 µg/h NA were orally treated with or without 9.7 mg/day azelnidipine or 5.9 mg/day hydralazine over 14 days. BP levels were continuously monitored via abdominal aortic catheter with a telemetry system in an unrestrained condition. Standard deviations (SDs) were used to evaluate beat-to-beat BPV and BPV every 15 min which was obtained by averaging BP levels for 10-s segment at each time point. BRS was determined by a sequence analysis. Continuous NA infusion over 14 days increased average BP, beat-to-beat BPV, and BPV every 15 min, lowering BRS. Comparing the two vasodilators, hydralazine reduced BP elevation by NA; meanwhile, azelnidipine alleviated BPV augmentation, preserving BRS, despite a smaller BP reduction. Thus, NA infusion increased both very short- and short-term BPV concomitantly with impaired BRS, while azelnidipine had an inhibitory effect, possibly independent of BP-lowering, on those types of BPV and impairment of BRS.


Subject(s)
Azetidinecarboxylic Acid/analogs & derivatives , Dihydropyridines , Norepinephrine , Vasodilator Agents , Rats , Animals , Blood Pressure , Vasodilator Agents/pharmacology , Norepinephrine/pharmacology , Rats, Wistar , Hydralazine/pharmacology
8.
J Trop Pediatr ; 70(2)2024 02 07.
Article in English | MEDLINE | ID: mdl-38324898

ABSTRACT

BACKGROUND: The incidence of neonatal septic shock in low-income countries is 26.8% with a mortality rate of 35.4%. The evidence of the hemodynamic effects of noradrenaline in neonates remains sparse. This study was carried out to evaluate the effects of noradrenaline in neonates with septic shock. METHODS: This was a single-center prospective cohort study in a tertiary care hospital's level III neonatal intensive care unit. Neonates with septic shock and those who received noradrenaline as a first-line vasoactive agent were included. Clinical and hemodynamic parameters were recorded before and after one hour of noradrenaline infusion. The primary outcomes were: response at the end of one hour after starting noradrenaline infusion and mortality rate. RESULTS: A total of 21 babies were analyzed. The cohort comprised 17 preterm neonates. The mean age of presentation with septic shock was 74.3 h. Resolution of shock at one hour after starting noradrenaline was achieved in 76.2% of cases. The median duration of hospital stay was 14 days. The mean blood pressure improved after the initiation of noradrenaline from 30.6 mm of Hg [standard deviation (SD) 6.1] to 37.8 mm of Hg (SD 8.22, p < 0.001). Fractional shortening improved after noradrenaline initiation from 29% (SD 13.5) to 45.1% (SD 21.1, p < 0.001). The mortality rate was 28.6% in our study. CONCLUSION: Noradrenaline is a potential drug for use in neonatal septic shock, with improvement in mean blood pressure and fractional shortening; however, further studies with larger sample sizes are needed to confirm our findings before it can be recommended as first-line therapy in neonatal septic shock.


Neonatal sepsis is one of the leading causes of neonatal mortality. In neonates with septic shock, mortality is high at 35.4% in low- and middle-income countries. The evidence of the hemodynamic effects of noradrenaline in neonates is still sparse, so we carried out a study in our tertiary care neonatal intensive care unit to evaluate the effects of noradrenaline in neonates with septic shock. Neonates with septic shock and those who received noradrenaline as a first-line vasoactive agent were included. Clinical and hemodynamic parameters were recorded before and after one hour of noradrenaline infusion. The primary outcomes were: response at the end of one hour after starting noradrenaline infusion and mortality rate. A total of 21 babies were analyzed. We found that there was a statistically significant improvement in the mean blood pressure and fractional shortening after noradrenaline initiation. The mortality rate was 28.6% in our study. We conclude that noradrenaline is a relatively safe and effective drug for the treatment of neonatal septic shock. However, further studies with larger sample sizes are needed to confirm our findings before it can be recommended as first-line therapy in neonatal septic shock.


Subject(s)
Mercury , Shock, Septic , Infant, Newborn , Humans , Norepinephrine/pharmacology , Norepinephrine/therapeutic use , Prospective Studies , Hemodynamics , Mercury/pharmacology
9.
Neuropharmacology ; 247: 109861, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38331315

ABSTRACT

Sleep is an instinct behavior, and its significance and functions are still an enigma. It is expressed throughout one's life and its loss affects psycho-somatic and physiological processes. We had proposed that it might maintain a fundamental property of the neurons and the brain. In that context, it was shown that sleep, rapid eye movement sleep (REMS) in particular, by regulating noradrenaline (NA), maintains the brain excitability. It was also reported that sleep-loss affected memory, reaction time and decision-making ability among others. However, as there was lack of clarity on the cause-and-effect relationship as to how the sleep-loss could affect these basic behaviors, their association was questioned and it was difficult to propose a cure or at least ways and means to ameliorate the symptoms. Also, we wanted to conduct the studies in a simpler model system so that conducting future molecular studies might be easier. Hence, using zebrafish as a model we evaluated if sleep-loss affected the basic decision-making ability, a cognitive process and if the effect was induced by NA. Indeed, our findings confirmed that upon sleep-deprivation, the cognitive decision-making ability of the prey zebrafish was compromised to protect itself by running away from the reach of the exposed predator Tiger Oscar (TO) fish. Also, we observed that upon sleep-loss the axonal arborization of the prey zebrafish brain was reduced. Interestingly, the effects were prevented by prazosin (PRZ), an α1-adrenoceptor (AR) antagonist and when the zebrafish recovered from the lost sleep.


Subject(s)
Norepinephrine , Zebrafish , Animals , Norepinephrine/pharmacology , Sleep Deprivation , Sleep , Neurons , Receptors, Adrenergic, alpha-1/physiology
10.
Auton Neurosci ; 252: 103154, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38330594

ABSTRACT

INTRODUCTION: Autonomic dysreflexia (AD) is a potentially life-threatening consequence in high (above T6) spinal cord injury that involves multiple incompletely understood mechanisms. While peripheral arteriolar vasoconstriction, which controls systemic vascular resistance, is documented to be pronounced during AD, the pathophysiological neurovascular junction mechanisms of this vasoconstriction are undefined. One hypothesized mechanism is increased neuronal release of norepinephrine and co-transmitters. We tested this by examining the effects of blockade of pre-synaptic neural release of norepinephrine and co-transmitters on cutaneous vasoconstriction during AD, using a novel non-invasive technique; bretylium (BT) iontophoresis followed by skin blood flow measurements via laser doppler flowmetry (LDF). METHODS: Bretylium, a sympathetic neuronal blocking agent (blocks release of norepinephrine and co-transmitters) was applied iontophoretically to the skin of a sensate (arm) and insensate (leg) area in 8 males with motor complete tetraplegia. An nearby untreated site served as control (CON). Cutaneous vascular conductance (CVC) was measured (CVC = LDF/mean arterial pressure) at normotension before AD was elicited by bladder stimulation. The percent drop in CVC values from pre-AD vs. AD was compared among BT and CON sites in sensate and insensate areas. RESULTS: There was a significant effect of treatment but no significant effect of limb/sensation or interaction of limb x treatment on CVC. The percent drop in CVC between BT and CON treated sites was 25.7±1.75 vs. 39.4±0.87, respectively (P = 0.004). CONCLUSION: Bretylium attenuates, but does not fully abolish vasoconstriction during AD. This suggests release of norepinephrine and cotransmitters from cutaneous sympathetic nerves is involved in cutaneous vasoconstriction during AD.


Subject(s)
Autonomic Dysreflexia , Bretylium Compounds , Vasoconstriction , Male , Humans , Skin Temperature , Skin/innervation , Norepinephrine/pharmacology , Neurotransmitter Agents/pharmacology , Regional Blood Flow
11.
JCI Insight ; 9(5)2024 Mar 08.
Article in English | MEDLINE | ID: mdl-38271085

ABSTRACT

High-grade serous carcinoma (HGSC) is the most lethal gynecological malignancy in the United States. Late diagnosis and the emergence of chemoresistance have prompted studies into how the tumor microenvironment, and more recently tumor innervation, may be leveraged for HGSC prevention and interception. In addition to stess-induced sources, concentrations of the sympathetic neurotransmitter norepinephrine (NE) in the ovary increase during ovulation and after menopause. Importantly, NE exacerbates advanced HGSC progression. However, little is known about the role of NE in early disease pathogenesis. Here, we investigated the role of NE in instigating anchorage independence and micrometastasis of preneoplastic lesions from the fallopian tube epithelium (FTE) to the ovary, an essential step in HGSC onset. We found that in the presence of NE, FTE cell lines were able to survive in ultra-low-attachment (ULA) culture in a ß-adrenergic receptor-dependent (ß-AR-dependent) manner. Importantly, spheroid formation and cell viability conferred by treatment with physiological sources of NE were abrogated using the ß-AR blocker propranolol. We have also identified that NE-mediated anoikis resistance may be attributable to downregulation of colony-stimulating factor 2. These findings provide mechanistic insight and identify targets that may be regulated by ovary-derived NE in early HGSC.


Subject(s)
Cystadenocarcinoma, Serous , Ovarian Neoplasms , Female , Humans , Ovarian Neoplasms/metabolism , Cystadenocarcinoma, Serous/drug therapy , Cystadenocarcinoma, Serous/metabolism , Cystadenocarcinoma, Serous/pathology , Fallopian Tubes/metabolism , Fallopian Tubes/pathology , Anoikis , Norepinephrine/pharmacology , Norepinephrine/metabolism , Tumor Microenvironment
12.
Fish Physiol Biochem ; 50(2): 705-719, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38294642

ABSTRACT

Color changes and pattern formations can represent strategies of the utmost importance for the survival of individuals or of species. Previous studies have associated capture with the formation of blotches (areas with light color) of coral trout, but the regulatory mechanisms link the two are lacking. Here, we report that capture induced blotches formation within 4-5 seconds. The blotches disappeared after anesthesia dispersed the pigment cells and reappeared after electrical stimulation. Subsequently, combining immunofluorescence, transmission electron microscopy and chemical sympathectomy, we found blotches formation results from activation of catecholaminergic neurons below the pigment layer. Finally, the in vitro incubation and intraperitoneal injection of norepinephrine (NE) induced aggregation of chromatosomes and lightening of body color, respectively, suggesting that NE, a neurotransmitter released by catecholaminergic nerves, mediates blotches formation. Our results demonstrate that acute stress response-induced neuronal activity can drive rapid changes in body color, which enriches our knowledge of physiological adaptations in coral reef fish.


Subject(s)
Anthozoa , Bass , Animals , Trout , Norepinephrine/pharmacology , Bass/physiology , Coral Reefs
13.
Behav Brain Res ; 462: 114872, 2024 Mar 28.
Article in English | MEDLINE | ID: mdl-38266779

ABSTRACT

The dorsal raphe nucleus (DRN) is essential for the control of food intake. Efferent projections from the DRN extend to several forebrain regions that are involved in the control of food intake. However, the neurotransmitters released in the DRN related to the control of food intake are not known. We have previously demonstrated that a tonic α1 action on DRN neurons contributes to satiety in the fed rats. In this study we investigated the participation of norepinephrine (NE) signaling in the DRN in the satiety response. Intra-DRN administration of NE causes an increase in the 2-hour food intake of sated mice, an effect that was blocked by previous administration of yohimbine, an α2 antagonist. Similarly, Intra-DRN administration of clonidine, an α2 agonist, increases food intake in sated mice. This result indicates that in the satiated mice exogenous NE acts on α2 receptors to increase food intake. Furthermore, administration of phenylephrine, an α1 agonist, decreases food intake in fasted mice and prazosin, an α1 antagonist, increases food intake in the sated mice. Taken together these results indicate that, in a satiated condition, a tonic α1 adrenergic action on the DRN neurons inhibits food intake and that exogenous NE administered to the DRN acts on α2 adrenergic receptors to increase food intake. These data reinforce the intricate neuronal functioning of the DRN and its effects on feeding.


Subject(s)
Dorsal Raphe Nucleus , Norepinephrine , Rats , Mice , Male , Animals , Norepinephrine/pharmacology , Neurons/physiology , Prazosin/pharmacology , Eating
14.
Neuropharmacology ; 246: 109849, 2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38244888

ABSTRACT

Cognitive impairment is a debilitating feature of psychiatric disorders including schizophrenia, mood disorders and substance use disorders for which there is a substantial lack of effective therapies. d-Govadine (d-GOV) is a tetrahydroprotoberberine recently shown to significantly enhance working memory and behavioural flexibility in several prefrontal cortex (PFC)-dependent rodent tasks. d-GOV potentiates dopamine (DA) efflux in the mPFC and not the nucleus accumbens, a unique pharmacology that sets it apart from many dopaminergic drugs and likely contributes to its effects on cognitive function. However, specific mechanisms involved in the preferential effects of d-GOV on mPFC DA function remain to be determined. The present study employs brain dialysis in male rats to deliver d-GOV into the mPFC or ventral tegmental area (VTA), while simultaneously sampling DA and norepinephrine (NE) efflux in the mPFC. Intra-PFC delivery or systemic administration of d-GOV preferentially potentiated medial prefrontal DA vs NE efflux. This differential effect of d-GOV on the primary catecholamines known to affect mPFC function further underscores its specificity for the mPFC DA system. Importantly, the potentiating effect of d-GOV on mPFC DA was disrupted when glutamatergic transmission was blocked in either the mPFC or the VTA. We hypothesize that d-GOV acts in the mPFC to engage the mesocortical feedback loop through which prefrontal glutamatergic projections activate a population of VTA DA neurons that specifically project back to the PFC. The activation of a PFC-VTA feedback loop to elevate PFC DA efflux without affecting mesolimbic DA release represents a novel approach to developing pro-cognitive drugs.


Subject(s)
Berberine Alkaloids , Dopamine , Nootropic Agents , Humans , Rats , Male , Animals , Dopamine/pharmacology , Nootropic Agents/pharmacology , Rats, Sprague-Dawley , Norepinephrine/pharmacology , Ventral Tegmental Area , Prefrontal Cortex
15.
Psychopharmacology (Berl) ; 241(4): 767-783, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38001266

ABSTRACT

RATIONALE: Win-paired stimuli can promote risk taking in experimental gambling paradigms in both rats and humans. We previously demonstrated that atomoxetine, a noradrenaline reuptake inhibitor, and guanfacine, a selective α2A adrenergic receptor agonist, reduced risk taking on the cued rat gambling task (crGT), a rodent assay of risky choice in which wins are accompanied by salient cues. Both compounds also decreased impulsive premature responding. OBJECTIVE: The key neural loci mediating these effects were unknown. The lateral orbitofrontal cortex (lOFC) and the medial prefrontal cortex (mPFC), which are highly implicated in risk assessment, action selection, and impulse control, receive dense noradrenergic innervation. We therefore infused atomoxetine and guanfacine directly into either the lOFC or prelimbic (PrL) mPFC prior to task performance. RESULTS: When infused into the lOFC, atomoxetine improved decision making score and adaptive lose-shift behaviour in males, but not in females, without altering motor impulsivity. Conversely, intra-PrL atomoxetine improved impulse control in risk preferring animals of both sexes, but did not alter decision making. Guanfacine administered into the PrL, but not lOFC, also altered motor impulsivity in all subjects, though in the opposite direction to atomoxetine. CONCLUSIONS: These data highlight a double dissociation between the behavioural effects of noradrenergic signaling across frontal regions with respect to risky choice and impulsive action. Given that the influence of noradrenergic manipulations on motor impulsivity could depend on baseline risk preference, these data also suggest that the noradrenaline system may function differently in subjects that are susceptible to the risk-promoting lure of win-associated cues.


Subject(s)
Cues , Guanfacine , Humans , Male , Female , Rats , Animals , Atomoxetine Hydrochloride/pharmacology , Guanfacine/pharmacology , Impulsive Behavior/physiology , Norepinephrine/pharmacology , Brain , Prefrontal Cortex , Decision Making , Choice Behavior
16.
Shock ; 61(3): 406-413, 2024 Mar 01.
Article in English | MEDLINE | ID: mdl-38010114

ABSTRACT

ABSTRACT: Background: High-dose vasopressors maintain blood pressure during septic shock but may adversely reduce microcirculation in vital organs. We assessed the effect of high-dose norepinephrine and vasopressin on the microcirculation of the brain, tongue, liver, and kidney during endotoxic shock using near-infrared spectroscopy (NIRS). Methods: Thirteen pigs (24.5 ± 1.8 kg) were anesthetized, and an NIRS probe was attached directly to each organ. Approximately 0.2, 0.5, 1, and 2 µg/kg/min of norepinephrine were administered in a stepwise manner, followed by 0.5, 1, 2, and 5 µg/kg/min of sodium nitroprusside in normal condition. Moreover, 1 µg/kg/h of lipopolysaccharide was administered continuously after 100 µg bolus to create endotoxic shock and after 1,000 mL of crystalloid infusion and high-dose norepinephrine (2, 5, 10, and 20 µg/kg/min) and vasopressin (0.6, 1.5, 3, and 6 U/min) were administered in a stepwise manner. The relationship between the MAP and each tissue oxygenation index (TOI) during vasopressor infusion was evaluated. Results: Three pigs died after receiving lipopolysaccharides, and 10 were analyzed. An increase of >20% from the baseline MAP induced by high-dose norepinephrine during endotoxic shock reduced the TOI in all organs except the liver. The elevation of MAP to baseline with vasopressin alone increased the kidney and liver TOIs and decreased the tongue TOI. Conclusion: Forced blood pressure elevation with high-dose norepinephrine during endotoxic shock decreased the microcirculation of vital organs, especially the kidney. Cerebral TOI may be useful for identifying the upper limit of blood pressure, at which norepinephrine impairs microcirculation.


Subject(s)
Shock, Septic , Swine , Animals , Shock, Septic/drug therapy , Microcirculation , Spectroscopy, Near-Infrared , Vasoconstrictor Agents/pharmacology , Vasoconstrictor Agents/therapeutic use , Kidney , Vasopressins/pharmacology , Norepinephrine/pharmacology , Lipopolysaccharides/pharmacology , Liver , Tongue
17.
Biochim Biophys Acta Mol Cell Res ; 1871(2): 119651, 2024 02.
Article in English | MEDLINE | ID: mdl-38086448

ABSTRACT

Hypertension is one of the major life-threatening complications of obesity. Recently adipose multipotent mesenchymal stromal cells (MSCs) were implicated to the pathogenesis of obesity-associated hypertension. These cells amplify noradrenaline-induced vascular cell contraction via cAMP-mediated signaling pathway. In this study we tested the ability of several cAMP-mediated hormones to affect the adrenergic sensitivity of MSCs and their associated contractility. Despite that adipose MSCs express a plethora of receptors capable of cAMP signaling activation, only 5-HT was able to elevate α1A-adrenoceptor-induced Ca2+ signaling in MSCs. Furthermore, 5-HT markedly enhanced noradrenaline-induced MSCs contractility. Using HTR isoform-specific antagonists followed by CRISPRi-mediated knockdown, we identified that the observed 5-HT effect on MSCs was mediated by the HTR6 isoform. This receptor was previously associated exclusively with 5-HT central nervous system activity. Discovered effect of HTR6 on MSCs contractility points to it as a potential therapeutic target for the prevention and treatment of obesity-associated hypertension.


Subject(s)
Hypertension , Serotonin , Humans , Norepinephrine/pharmacology , Hypertension/etiology , Obesity/complications , Protein Isoforms
18.
Microvasc Res ; 152: 104627, 2024 03.
Article in English | MEDLINE | ID: mdl-37963515

ABSTRACT

AIMS: Protein kinase D (PKD), once considered an effector of protein kinase C (PKC), now plays many pathophysiological roles in various tissues. However, little is known about role of PKD in vascular function. We investigated the role of PKD in contraction of rat aorta and human aortic smooth muscle cells (HASMCs) and in haemodynamics in rats. METHODS AND RESULTS: Isometric tension of rat aortic was measured to examine norepinephrine-induced contraction in the presence of PKD, PKC and Rho-kinase inhibitors. Phosphorylation of PKD1, myosin targeting subunit-1 (MYPT1), myosin light chain (MLC), CPI-17 and heat-shock protein 27 (HSP27), and actin polymerization were measured in the aorta. Phosphorylation of MYPT1 and MLC was also measured in HASMCs knocked down with specific siRNAs of PKD 1, 2 and 3. Intracellular calcium concentrations and cell shortening were measured in HASMCs. Norepinephrine-induced aortic contraction was accompanied by increased phosphorylation of PKD1, MYPT1 and MLC and actin polymerization, all of which were attenuated with PKD inhibitor CRT0066101. PKD1 phosphorylation was not inhibited by PKC inhibitor, chelerythrine or Rho kinase inhibitor, fasudil. In HASMCs, the phosphorylation of MYPT1 and MLC was attenuated by PKD1, but not PKD2, 3 knockdown. In HASMCs, CRT0066101 inhibited norepinephrine-induced cell shortening without affecting calcium concentration. Administration of CRT0066101 decreased systemic vascular resistance and blood pressure without affecting cardiac output in rats. CONCLUSIONS: PKD1 may play roles in aorta contraction and haemodynamics via phosphorylation of MYPT1 and actin polymerization in a calcium-independent manner.


Subject(s)
Actins , Vasoconstriction , Animals , Humans , Rats , Actins/metabolism , Calcium/metabolism , Muscle Contraction , Muscle, Smooth, Vascular/metabolism , Myosin Light Chains/metabolism , Norepinephrine/pharmacology , Norepinephrine/metabolism , Phosphorylation , Protein Kinase Inhibitors/pharmacology , rho-Associated Kinases/metabolism
19.
Neuroendocrinology ; 114(1): 25-41, 2024.
Article in English | MEDLINE | ID: mdl-37699381

ABSTRACT

INTRODUCTION: The ventromedial hypothalamic nucleus (VMN) is an estrogen receptor (ER)-rich structure that regulates glucostasis. The role of nuclear but not membrane G protein-coupled ER-1 (GPER) in that function has been studied. METHODS: Gene silencing and laser-catapult microdissection/immunoblot tools were used to examine whether GPER regulates transmitter and energy sensor function in dorsomedial (VMNdm) and/or ventrolateral (VMNvl) VMN counter-regulatory nitrergic and γ-Aminobutyric acid (GABA) neurons. RESULTS: Intra-VMN GPER siRNA administration to euglycemic animals did not affect VMNdm or -vl nitrergic neuron nitric oxide synthase (nNOS), but upregulated (VMNdm) or lacked influence on (VMNvl) GABA nerve cell glutamate decarboxylase65/67 (GAD) protein. Insulin-induced hypoglycemia (IIH) caused GPER knockdown-reversible augmentation of nNOS, 5'-AMP-activated protein kinase (AMPK), and phospho-AMPK proteins in nitrergic neurons in both divisions. IIH had dissimilar effects on VMNvl (unchanged) versus VMNdm (increased) GABAergic neuron GAD levels, yet GPER knockdown affected these profiles. GPER siRNA prevented hypoglycemic upregulation of VMNvl and -dm GABA neuron AMPK without altering pAMPK expression. CONCLUSIONS: Outcomes infer that GPER exerts differential control of VMNdm versus -vl GABA transmission during glucostasis and is required for hypoglycemic upregulated nitrergic (VMNdm and -vl) and GABA (VMNdm) signaling. Glycogen metabolism is reported to regulate VMN nNOS and GAD proteins. Data show that GPER limits VMNvl glycogen phosphorylase (GP) protein expression and glycogen buildup during euglycemia but mediates hypoglycemic augmentation of VMNvl GP protein and glycogen content; VMNdm glycogen mass is refractory to GPER control. GPER regulation of VMNvl glycogen metabolism infers that this receptor may govern local counter-regulatory transmission in part by astrocyte metabolic coupling.


Subject(s)
Hypoglycemia , Ventromedial Hypothalamic Nucleus , Rats , Animals , AMP-Activated Protein Kinases/metabolism , AMP-Activated Protein Kinases/pharmacology , Rats, Sprague-Dawley , Norepinephrine/metabolism , Norepinephrine/pharmacology , Receptors, Estrogen/metabolism , Hypoglycemia/metabolism , Glycogen/metabolism , Glycogen/pharmacology , Hypoglycemic Agents/pharmacology , gamma-Aminobutyric Acid/metabolism , RNA, Small Interfering/metabolism , RNA, Small Interfering/pharmacology , GTP-Binding Proteins/metabolism , GTP-Binding Proteins/pharmacology
20.
Eur J Neurosci ; 59(6): 1278-1295, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38052454

ABSTRACT

Astrocytes, the most abundant glial cells in the central nervous system, respond to a wide variety of neurotransmitters binding to metabotropic receptors. Here, we investigated the intracellular calcium responses of spinal cord astrocytes to dopamine and noradrenaline, two catecholamines released by specific descending pathways. In a slice preparation from the spinal cord of neonatal mice, puff application of dopamine resulted in intracellular calcium responses that remained in the endfeet. Noradrenaline induced stronger responses that also started in the endfeet but spread to neighbouring compartments. The intracellular calcium responses were unaffected by blocking neuronal activity or inhibiting various neurotransmitter receptors, suggesting a direct effect of dopamine and noradrenaline on astrocytes. The intracellular calcium responses induced by noradrenaline and dopamine were inhibited by the D1 receptor antagonist SCH 23390. We assessed the functional consequences of these astrocytic responses by examining changes in arteriole diameter. Puff application of dopamine or noradrenaline resulted in vasoconstriction of spinal arterioles. However, blocking D1 receptors or manipulating astrocytic intracellular calcium levels did not abolish the vasoconstrictions, indicating that the observed intracellular calcium responses in astrocyte endfeet were not responsible for the vascular changes. Our findings demonstrate a compartmentalized response of spinal cord astrocytes to catecholamines and expand our understanding of astrocyte-neurotransmitter interactions and their potential roles in the physiology of the central nervous system.


Subject(s)
Dopamine , Norepinephrine , Mice , Animals , Norepinephrine/pharmacology , Norepinephrine/metabolism , Dopamine/metabolism , Astrocytes/metabolism , Calcium/metabolism , Catecholamines/metabolism , Catecholamines/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...