Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 5.179
Filter
1.
Neuroscience ; 516: 91-99, 2023 04 15.
Article in English | MEDLINE | ID: mdl-36858308

ABSTRACT

Targeting reconsolidation with propranolol, a blocker of ß-adrenergic receptors (ß-ARs), emerged as a potential treatment for maladaptive memories such as those involved in posttraumatic stress disorder (PTSD). Reconsolidation targeting treatments for PTSD are becoming a common practice in the clinic and it is important to unveil any side effects upon 'non-targeted' memories. While previous studies have focused on propranolol's effects on the reconsolidation of emotional/distressful memories, the present study asked whether propranolol is involved in the reconsolidation of recognition memories - by assessing its effects on distinct memory components and the role of the dorsal hippocampus. Rats performed an object recognition (OR) task where they were exposed to different objects: A and B presented during the sample phase; A and C presented during the reactivation phase; and D in combination of either A, B, or C during a final test. Intra-hippocampal injections of propranolol (5 µg or 10 µg) were conducted immediately after the reactivation session. Propranolol infusions consistently impaired the addition of novel information to the previously consolidated memory trace regardless of dose, and the retention of familiar objects was not affected. Higher doses of propranolol also hindered memory of a familiar object that was not presented during the reactivation session, but was previously placed at the same location where novel information was presented during reactivation. The present results shed light on the role of ß-ARs on the reconsolidation of different memory components and argue for the need for further studies examining possible recognition memory deficits following propranolol treatment.


Subject(s)
Adrenergic beta-Antagonists , Propranolol , Rats , Animals , Propranolol/pharmacology , Adrenergic beta-Antagonists/pharmacology , Emotions , Recognition, Psychology , Hippocampus/metabolism , Receptors, Adrenergic, beta/physiology
2.
Pflugers Arch ; 474(12): 1311-1321, 2022 12.
Article in English | MEDLINE | ID: mdl-36131146

ABSTRACT

Atrial fibrillation (AF) from elevated adrenergic activity may involve increased atrial L-type Ca2+ current (ICaL) by noradrenaline (NA). However, the contribution of the adrenoceptor (AR) sub-types to such ICaL-increase is poorly understood, particularly in human. We therefore investigated effects of various broad-action and sub-type-specific α- and ß-AR antagonists on NA-stimulated atrial ICaL. ICaL was recorded by whole-cell-patch clamp at 37 °C in myocytes isolated enzymatically from atrial tissues from consenting patients undergoing elective cardiac surgery and from rabbits. NA markedly increased human atrial ICaL, maximally by ~ 2.5-fold, with EC75 310 nM. Propranolol (ß1 + ß2-AR antagonist, 0.2 microM) substantially decreased NA (310 nM)-stimulated ICaL, in human and rabbit. Phentolamine (α1 + α2-AR antagonist, 1 microM) also decreased NA-stimulated ICaL. CGP20712A (ß1-AR antagonist, 0.3 microM) and prazosin (α1-AR antagonist, 0.5 microM) each decreased NA-stimulated ICaL in both species. ICI118551 (ß2-AR antagonist, 0.1 microM), in the presence of NA + CGP20712A, had no significant effect on ICaL in human atrial myocytes, but increased it in rabbit. Yohimbine (α2-AR antagonist, 10 microM), with NA + prazosin, had no significant effect on human or rabbit ICaL. Stimulation of atrial ICaL by NA is mediated, based on AR sub-type antagonist responses, mainly by activating ß1- and α1-ARs in both human and rabbit, with a ß2-inhibitory contribution evident in rabbit, and negligible α2 involvement in either species. This improved understanding of AR sub-type contributions to noradrenergic activation of atrial ICaL could help inform future potential optimisation of pharmacological AR-antagonism strategies for inhibiting adrenergic AF.


Subject(s)
Calcium Channels, L-Type , Myocytes, Cardiac , Norepinephrine , Receptors, Adrenergic, alpha , Receptors, Adrenergic, beta , Animals , Humans , Rabbits , Atrial Fibrillation/physiopathology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/physiology , Norepinephrine/pharmacology , Norepinephrine/physiology , Prazosin/pharmacology , Receptors, Adrenergic, alpha-2 , Heart Atria/cytology , Receptors, Adrenergic, beta/physiology , Receptors, Adrenergic, alpha/physiology , Adrenergic alpha-Antagonists/pharmacology , Adrenergic beta-Antagonists/pharmacology , Calcium Channels, L-Type/physiology
3.
Pharmacol Res Perspect ; 10(3): e00975, 2022 06.
Article in English | MEDLINE | ID: mdl-35643970

ABSTRACT

A2A and A2B adenosine receptors produce regionally selective regulation of vascular tone and elicit differing effects on mean arterial pressure (MAP), whilst inducing tachycardia. The tachycardia induced by the stimulation of A2A or A2B receptors has been suggested to be mediated by a reflex increase in sympathetic activity. Here, we have investigated the role of ß1 - and ß2 -adrenoceptors in mediating the different cardiovascular responses to selective A2A and A2B receptor stimulation. Hemodynamic variables were measured in conscious male Sprague-Dawley rats (350-450 g) via pulsed Doppler flowmetry. The effect of intravenous infusion (3 min per dose) of the A2A -selective agonist CGS 21680 (0.1, 0.3, 1.0 µg.kg-1 .min-1 ) or the A2B -selective agonist BAY 60-6583 (4.0, 13.3, 40.0 µg.kg-1 .min-1 ) in the absence or following pre-treatment with the non-selective ß-antagonist propranolol (1.0 mg.kg-1 ), the selective ß1 -antagonist CGP 20712A (200 µg.kg-1 ), or the selective ß2 -antagonist ICI 118,551 (2.0 mg.kg-1 ) was investigated (maintenance doses also administered). CGP 20712A and propranolol significantly reduced the tachycardic response to CGS 21680, with no change in the effect on MAP. ICI 118,551 increased BAY 60-6583-mediated renal and mesenteric flows, but did not affect the heart rate response. CGP 20712A attenuated the BAY 60-6583-induced tachycardia. These data imply a direct stimulation of the sympathetic activity via cardiac ß1 -adrenoceptors as a mechanism for the A2A - and A2B -induced tachycardia. However, the regionally selective effects of A2B agonists on vascular conductance were independent of sympathetic activity and may be exploitable for the treatment of acute kidney injury and mesenteric ischemia.


Subject(s)
Adrenergic beta-Antagonists , Propranolol , Adenosine/pharmacology , Adrenergic beta-Antagonists/pharmacology , Animals , Blood Pressure , Male , Propranolol/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Adrenergic, beta/physiology , Tachycardia/chemically induced
4.
Hippocampus ; 32(6): 449-465, 2022 06.
Article in English | MEDLINE | ID: mdl-35478421

ABSTRACT

Patterned stimulation of the locus coeruleus (LC, 100 Hz), in conjunction with test-pulse stimulation of hippocampal afferents, results in input-specific long-term depression (LTD) of synaptic plasticity in the hippocampus. Effects are long-lasting and have been described in Schaffer-collateral-CA1 and perforant path-dentate gyrus synapses in behaving rats. To what extent LC-mediated hippocampal LTD (LC-LTD) is frequency-dependent is unclear. Here, we report that LC-LTD can be triggered by LC stimulation with 2 and 5 Hz akin to tonic activity, 10 Hz equivalent to phasic activity, and 100 Hz akin to high-phasic activity in the dentate gyrus (DG) of freely behaving rats. LC-LTD at both 2 and 100 Hz can be significantly prevented by an NMDA receptor antagonist. The LC releases both noradrenaline (NA) and dopamine (DA) from its hippocampal terminals and may also trigger hippocampal DA release by activating the ventral tegmental area (VTA). Unclear is whether both neurotransmitters contribute equally to hippocampal LTD triggered by LC stimulation (LC-LTD). Both DA D1/D5 receptors (D1/D5R) and beta-adrenergic receptors (ß-AR) are critically required for hippocampal LTD that is induced by patterned stimulation of hippocampal afferents, or is facilitated by spatial learning. We, therefore, explored to what extent these receptor subtypes mediate frequency-dependent hippocampal LC-LTD. LC-LTD elicited by 2, 5, and 10 Hz stimulation was unaffected by antagonism of ß-AR with propranolol, whereas LC-LTD induced by these frequencies was prevented by D1/D5R-antagonism using SCH23390. By contrast, LC-LTD evoked at 100 Hz was prevented by ß-AR-antagonism and only mildly affected by D1/D5R-antagonism. Taken together, these findings support that LC-LTD can be triggered by LC activity at a wide range of frequencies. Furthermore, the contribution of D1/D5R and ß-AR to hippocampal LTD that is triggered by LC activity is frequency-dependent and suggests that D1/D5R may be involved in LC-mediated hippocampal tonus.


Subject(s)
Locus Coeruleus , Receptors, Adrenergic, beta , Receptors, Dopamine D1 , Receptors, Dopamine D5 , Animals , Dopamine , Hippocampus/physiology , Locus Coeruleus/metabolism , Long-Term Synaptic Depression/physiology , Neuronal Plasticity , Rats , Receptors, Adrenergic, beta/physiology , Receptors, Dopamine D1/physiology , Receptors, Dopamine D5/physiology
5.
Psychoneuroendocrinology ; 133: 105421, 2021 11.
Article in English | MEDLINE | ID: mdl-34592505

ABSTRACT

A large amount of literature has demonstrated that Perceived Criticism (PC)-that is, how critical a person believes a given relative is of him or her-is associated with negative clinical outcomes in a broad range of psychiatric disorders (e.g., relapse or recurrence of symptoms). A possible mechanism behind the predictive value of PC might be its association with the stress regulation process. This is the first study to investigate differences in the psychophysiological response to a social stress task in young women (mean age = 21.66, SD = 4.33) with high (n = 40) and low (n = 39) PC. The physiological response was investigated by measuring two markers of sympathetic activity mediated by acetylcholine (skin conductance levels; SCL) and adrenaline (pre-ejection period; PEP) levels, respectively, and one marker of the vagally-mediated parasympathetic system (heart rate variability; HRV). Moreover, we investigated the anticipation and perception of social threat, in the form of criticism, during the stressor. No differences in HRV and SCL were observed. However, individuals high in PC mobilized fewer cardiovascular resources to deal with the stressor, reflected in an attenuated beta-adrenergic response (i.e., lower PEP response). Women high in PC also expected and perceived more criticism during the stress task. Together, our results indicate that women high in PC make heightened social threat anticipation and interpretations, and they tend to engage in less active coping when exposed to socially evaluated stressful events. Our findings indicate that PC is associated with underlying stress-related psychobiological vulnerabilities that may contribute to its association with negative clinical outcomes.


Subject(s)
Anticipation, Psychological , Receptors, Adrenergic, beta , Social Perception , Stress, Psychological , Adult , Anticipation, Psychological/physiology , Female , Humans , Receptors, Adrenergic, beta/physiology , Social Perception/psychology , Stress, Psychological/physiopathology , Young Adult
6.
PLoS One ; 16(7): e0253139, 2021.
Article in English | MEDLINE | ID: mdl-34214097

ABSTRACT

Keratinocyte migration into skin wounds is the step of the healing process that correlates with the wound closure rate. Keratinocyte migration, and wound epithelialization are decreased when beta 2-adrenergic receptors (B2AR) are activated by 1 µM epinephrine/adrenaline, resulting in delayed wound healing in human and mouse skin. In the present study, we found paradoxically, that in a subset of keratinocyte strains exposure to low concentrations of epinephrine (0.1 nM) increased, rather than decreased, their migratory rate. We find that both the alpha- and the beta-adrenergic receptors are expressed in human keratinocytes, and expression of alpha-2 AR subtypes demonstrated for the first time. Therefore, we tested if the alpha-AR could be modulating the increased migratory response observed in these cell strains. By using specific inhibitors to alpha-AR, we demonstrated that blocking A2B-AR could reverse the rapid cell migration induced by the 0.1 nM epinephrine. Phosphorylation of ERK was elevated after 1-10 minutes of the low epinephrine treatment and the A2B-AR inhibitor blocked the ERK phosphorylation. The results suggest that both the A2B-AR and B2AR mediate keratinocyte migration, in which with a low level of epinephrine treatment, A2B-AR could alter the B2AR signals and regulate the migration rate.


Subject(s)
Cell Movement , Keratinocytes/physiology , Receptors, Adrenergic, alpha/physiology , Receptors, Adrenergic, beta/physiology , Humans , Infant, Newborn , Male , Receptors, Adrenergic, alpha/metabolism , Receptors, Adrenergic, beta/metabolism , Wound Healing
7.
J Pharmacol Sci ; 145(3): 228-240, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33602503

ABSTRACT

Astrocytes are glial cells with numerous fine processes which are important for the functions of the central nervous system. The activation of ß-adrenoceptors induces process formation of astrocytes via cyclic AMP (cAMP) signaling. However, the role of α-adrenoceptors in the astrocyte morphology has not been elucidated. Here, we examined it by using cultured astrocytes from neonatal rat spinal cords and cortices. Exposure of these cells to noradrenaline and the ß-adrenoceptor agonist isoproterenol increased intracellular cAMP levels and induced the formation of processes. Noradrenaline-induced process formation was enhanced with the α1-adrenoceptor antagonist prazosin and α2-adrenoceptor antagonist atipamezole. Atipamezole also enhanced noradrenaline-induced cAMP elevation. Isoproterenol-induced process formation was not inhibited by the α1-adrenoceptor agonist phenylephrine but was inhibited by the α2-adrenoceptor agonist dexmedetomidine. Dexmedetomidine also inhibited process formation induced by the adenylate cyclase activator forskolin and the membrane-permeable cAMP analog dibutyryl-cAMP. Moreover, dexmedetomidine inhibited cAMP-independent process formation induced by adenosine or the Rho-associated kinase inhibitor Y27632. In the presence of propranolol, noradrenaline inhibited Y27632-induced process formation, which was abolished by prazosin or atipamezole. These results demonstrate that α-adrenoceptors inhibit both cAMP-dependent and -independent astrocytic process formation.


Subject(s)
Astrocytes/drug effects , Astrocytes/metabolism , Receptors, Adrenergic, alpha/physiology , Receptors, Adrenergic, beta/physiology , Adrenergic alpha-Agonists/pharmacology , Adrenergic alpha-Antagonists/pharmacology , Adrenergic beta-Agonists/pharmacology , Animals , Cells, Cultured , Cyclic AMP/metabolism , Dexmedetomidine/pharmacology , Imidazoles/pharmacology , Isoproterenol/pharmacology , Norepinephrine/pharmacology , Prazosin/pharmacology , Rats, Wistar , Signal Transduction
8.
J Pharmacol Sci ; 145(3): 253-261, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33602505

ABSTRACT

Facilitation of cardiac function in response to signals from the sympathetic nervous system is initiated by the phosphorylation of L-type voltage-dependent Ca2+ channels (VDCCs) by protein kinase A (PKA), which in turn is activated by ß-adrenoceptors. Among the five subunits (α1, ß, α2/δ, and γ) of VDCCs, the α1 subunit and the family of ß subunits are substrates for PKA-catalyzed phosphorylation; however, the subunit responsible for ß-adrenergic augmentation of Ca2+ channel function has yet to be specifically identified. Here we show that the VDCC ß2 subunit is required for PKA phosphorylation upon sympathetic acceleration. In mice with ß2 subunit-null mutations, cardiac muscle contraction in response to isoproterenol was reduced and there was no significant increase in Ca2+ channel currents upon PKA-dependent phosphorylation. These findings indicate that within the sympathetic nervous system the ß2 subunit of VDCCs is required for physiological PKA-dependent channel phosphorylation.


Subject(s)
Calcium Channels, L-Type/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic AMP-Dependent Protein Kinases/physiology , Receptors, Adrenergic, beta/physiology , Sympathetic Nervous System/metabolism , Sympathetic Nervous System/physiology , Animals , Catalysis , Cells, Cultured , Isoproterenol/pharmacology , Mice , Mutation , Myocardial Contraction/drug effects , Phosphorylation , Receptors, Adrenergic, beta/genetics
9.
FEBS J ; 288(23): 6603-6622, 2021 12.
Article in English | MEDLINE | ID: mdl-33415835

ABSTRACT

Heart failure with preserved ejection fraction (HFpEF) will soon take over as the predominant form of heart failure. This is largely driven by the continuing increased incidences of obesity and type 2 diabetes (T2D), which promote HFpEF in the absence of pressure overload stresses. With beta-blockers showing little effectiveness in treating obesity/T2D HFpEF and with no HFpEF-targeted drugs currently available, we are in urgent need of a better understanding of how obesity/T2D HFpEF develops and how we may treat this condition. An exciting emerging field aiming to do this focuses on the investigation of 3',5'-cyclic adenosine monophosphate (cAMP) microdomains in the heart. The previous work has largely focused on the investigation of cAMP microdomain remodelling in heart failure with reduced ejection fraction (HFrEF), with this work uncovering potential new targets for intervention strategies that otherwise would have been overlooked when studying changes in cAMP dynamics at the whole-cell level. In this review, we aimed to discuss current advancements in our understanding of cAMP microdomain remodelling in HFrEF vs that in obesity/T2D-associated HFpEF, with particular focus on the unresolved questions and limitations we face in being able to translate this knowledge.


Subject(s)
Cyclic AMP/physiology , Diabetes Mellitus, Type 2/physiopathology , Heart Failure/physiopathology , Obesity/physiopathology , Stroke Volume/physiology , Ventricular Remodeling/physiology , Animals , Cyclic AMP/metabolism , Diabetes Mellitus, Type 2/complications , Heart Failure/diagnosis , Heart Failure/etiology , Humans , Models, Cardiovascular , Obesity/complications , Receptors, Adrenergic, beta/metabolism , Receptors, Adrenergic, beta/physiology
10.
Front Immunol ; 12: 715102, 2021.
Article in English | MEDLINE | ID: mdl-34987499

ABSTRACT

Background: Epstein-Barr virus (EBV) causes infectious mononucleosis (IM) that can lead to chronic fatigue syndrome. The CEBA-project (Chronic fatigue following acute EBV infection in Adolescents) has followed 200 patients with IM and here we present an immunological profiling of adolescents with IM related to clinical characteristics. Methods: Patients were sampled within 6 weeks of debut of symptoms and after 6 months. Peripheral blood mononuclear cells (PBMC) were cultured and stimulated in vitro (n=68), and supernatants analyzed for cytokine release. Plasma was analyzed for inflammatory markers (n=200). The Chalder Fatigue Questionnaire diagnosed patients with and without chronic fatigue at 6 months (CF+ and CF- group, respectively) (n=32 and n=91, in vitro and plasma cohorts, respectively. Results: Broad activation of PBMC at baseline, with high levels of RANTES (Regulated on activation, normal T-cell expressed and secreted) in the CF+ group, and broad inflammatory response in plasma with high levels of T-cell markers was obeserved. At 6 months, there was an increased ß-agonist response and RANTES was still elevated in cultures from the CF+ group. Plasma showed decrease of inflammatory markers except for CRP which was consistently elevated in the CF+ group. Conclusion: Patients developing chronic fatigue after IM have signs of T-cell activation and low-grade chronic inflammation at baseline and after 6 months. Clinical Trial Registration: https://clinicaltrials.gov/, identifier NCT02335437.


Subject(s)
Chemokine CCL5/blood , Fatigue Syndrome, Chronic/etiology , Infectious Mononucleosis/immunology , Inflammation/etiology , Lymphocyte Activation , T-Lymphocytes/immunology , Adolescent , Antibodies, Viral/blood , Biomarkers , Cells, Cultured , Chemokine CCL5/biosynthesis , Chronic Disease , Convalescence , Cross-Sectional Studies , Cytokines/blood , Fatigue Syndrome, Chronic/blood , Fatigue Syndrome, Chronic/immunology , Female , Follow-Up Studies , Herpesvirus 4, Human/immunology , Humans , Infectious Mononucleosis/blood , Infectious Mononucleosis/complications , Inflammation/blood , Inflammation/immunology , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Male , Prospective Studies , Receptors, Adrenergic, beta/physiology , T-Lymphocytes/metabolism
11.
Sci Rep ; 10(1): 18801, 2020 11 02.
Article in English | MEDLINE | ID: mdl-33139790

ABSTRACT

Myocardial infarction (MI) can result in sympathetic nerve loss in the infarct region. However, the contribution of hypo-innervation to electrophysiological remodeling, independent from MI-induced ischemia and fibrosis, has not been comprehensively investigated. We present a novel mouse model of regional cardiac sympathetic hypo-innervation utilizing a targeted-toxin (dopamine beta-hydroxylase antibody conjugated to saporin, DBH-Sap), and measure resulting electrophysiological and Ca2+ handling dynamics. Five days post-surgery, sympathetic nerve density was reduced in the anterior left ventricular epicardium of DBH-Sap hearts compared to control. In Langendorff-perfused hearts, there were no differences in mean action potential duration (APD80) between groups; however, isoproterenol (ISO) significantly shortened APD80 in DBH-Sap but not control hearts, resulting in a significant increase in APD80 dispersion in the DBH-Sap group. ISO also produced spontaneous diastolic Ca2+ elevation in DBH-Sap but not control hearts. In innervated hearts, sympathetic nerve stimulation (SNS) increased heart rate to a lesser degree in DBH-Sap hearts compared to control. Additionally, SNS produced APD80 prolongation in the apex of control but not DBH-Sap hearts. These results suggest that hypo-innervated hearts have regional super-sensitivity to circulating adrenergic stimulation (ISO), while having blunted responses to SNS, providing important insight into the mechanisms of arrhythmogenesis following sympathetic nerve loss.


Subject(s)
Cardiac Electrophysiology , Heart/innervation , Receptors, Adrenergic, beta/physiology , Sympathetic Nervous System/pathology , Sympathetic Nervous System/physiopathology , Action Potentials/drug effects , Animals , Arrhythmias, Cardiac/etiology , Calcium/metabolism , Isoproterenol/pharmacology , Male , Mice, Inbred C57BL , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardium/metabolism
12.
J Neurosci ; 40(45): 8604-8617, 2020 11 04.
Article in English | MEDLINE | ID: mdl-33046543

ABSTRACT

The second messenger cAMP is an important determinant of synaptic plasticity that is associated with enhanced neurotransmitter release. Long-term potentiation (LTP) at parallel fiber (PF)-Purkinje cell (PC) synapses depends on a Ca2+-induced increase in presynaptic cAMP that is mediated by Ca2+-sensitive adenylyl cyclases. However, the upstream signaling and the downstream targets of cAMP involved in these events remain poorly understood. It is unclear whether cAMP generated by ß-adrenergic receptors (ßARs) is required for PF-PC LTP, although noradrenergic varicosities are apposed in PF-PC contacts. Guanine nucleotide exchange proteins directly activated by cAMP [Epac proteins (Epac 1-2)] are alternative cAMP targets to protein kinase A (PKA) and Epac2 is abundant in the cerebellum. However, whether Epac proteins participate in PF-PC LTP is not known. Immunoelectron microscopy demonstrated that ßARs are expressed in PF boutons. Moreover, activation of these receptors through their agonist isoproterenol potentiated synaptic transmission in cerebellar slices from mice of either sex, an effect that was insensitive to the PKA inhibitors (H-89, KT270) but that was blocked by the Epac inhibitor ESI 05. Interestingly, prior activation of these ßARs occluded PF-PC LTP, while the ß1AR antagonist metoprolol blocked PF-PC LTP, which was also absent in Epac2-/- mice. PF-PC LTP is associated with an increase in the size of the readily releasable pool (RRP) of synaptic vesicles, consistent with the isoproterenol-induced increase in vesicle docking in cerebellar slices. Thus, the ßAR-mediated modulation of the release machinery and the subsequent increase in the size of the RRP contributes to PF-PC LTP.SIGNIFICANCE STATEMENT G-protein-coupled receptors modulate the release machinery, causing long-lasting changes in synaptic transmission that influence synaptic plasticity. Nevertheless, the mechanisms underlying synaptic responses to ß-adrenergic receptor (ßAR) activation remain poorly understood. An increase in the number of synaptic vesicles primed for exocytosis accounts for the potentiation of neurotransmitter release driven by ßARs. This effect is not mediated by the canonical protein kinase A pathway but rather, through direct activation of the guanine nucleotide exchange protein Epac by cAMP. Interestingly, this ßAR signaling via Epac is involved in long term potentiation at cerebellar granule cell-to-Purkinje cell synapses. Thus, the pharmacological activation of ßARs modulates synaptic plasticity and opens therapeutic opportunities to control this phenomenon.


Subject(s)
Guanine Nucleotide Exchange Factors/physiology , Long-Term Potentiation/physiology , Receptors, Adrenergic, beta/physiology , Synaptic Vesicles/physiology , Adrenergic beta-Agonists/pharmacology , Adrenergic beta-Antagonists/pharmacology , Animals , Cerebellum/cytology , Cerebellum/metabolism , Cyclic AMP/physiology , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic AMP-Dependent Protein Kinases/metabolism , Female , Guanine Nucleotide Exchange Factors/genetics , Guanine Nucleotide Exchange Factors/metabolism , Male , Mice , Mice, Knockout , Protein Kinase Inhibitors/pharmacology , Purkinje Cells/physiology , Receptors, Adrenergic, beta/genetics , Receptors, Adrenergic, beta/metabolism , Signal Transduction/genetics , Signal Transduction/physiology , Synaptic Transmission/drug effects , Synaptic Vesicles/ultrastructure
13.
Cereb Cortex ; 30(12): 6135-6151, 2020 11 03.
Article in English | MEDLINE | ID: mdl-32607551

ABSTRACT

Release of the neuromodulator noradrenaline signals salience during wakefulness, flagging novel or important experiences to reconfigure information processing and memory representations in the hippocampus. Noradrenaline is therefore expected to enhance hippocampal responses to synaptic input; however, noradrenergic agonists have been found to have mixed and sometimes contradictory effects on Schaffer collateral synapses and the resulting CA1 output. Here, we examine the effects of endogenous, optogenetically driven noradrenaline release on synaptic transmission and spike output in mouse hippocampal CA1 pyramidal neurons. We show that endogenous noradrenaline release enhances the probability of CA1 pyramidal neuron spiking without altering feedforward excitatory or inhibitory synaptic inputs in the Schaffer collateral pathway. ß-adrenoceptors mediate this enhancement of excitation-spike coupling by reducing the charge required to initiate action potentials, consistent with noradrenergic modulation of voltage-gated potassium channels. Furthermore, we find the likely effective concentration of endogenously released noradrenaline is sub-micromolar. Surprisingly, although comparable concentrations of exogenous noradrenaline cause robust depression of slow afterhyperpolarization currents, endogenous release of noradrenaline does not, indicating that endogenous noradrenaline release is targeted to specific cellular locations. These findings provide a mechanism by which targeted endogenous release of noradrenaline can enhance information transfer in the hippocampus in response to salient events.


Subject(s)
Action Potentials , CA1 Region, Hippocampal/physiology , Locus Coeruleus/physiology , Norepinephrine/physiology , Pyramidal Cells/physiology , Receptors, Adrenergic, beta/physiology , Animals , Excitatory Postsynaptic Potentials , Male , Mice, Inbred C57BL
14.
Cell Physiol Biochem ; 54(4): 665-681, 2020 Jul 08.
Article in English | MEDLINE | ID: mdl-32639114

ABSTRACT

BACKGROUND/AIMS: Aortic stenosis-induced chronic pressure overload leads to cardiac dysfunction and congestive heart failure. The pathophysiological mechanisms of the myocardial impairment are multifactorial and include maladaptive ß-adrenergic signaling. Exercise training (ET) has been used as a non-pharmacological therapy for heart failure management. The present study tested the hypothesis that exercise training attenuates diastolic dysfunction through ß-adrenergic signaling preservation. METHODS: Wistar rats were submitted to ascending aortic stenosis (AS) surgery, and after 18 weeks, a moderate aerobic exercise training protocol was performed for ten weeks. RESULTS: ET attenuated diastolic dysfunction, evaluated by echocardiogram and isolated papillary muscle (IPM) assay. Also, ET reduced features of heart failure, cross-sectional cardiomyocyte area, and exercise intolerance, assessed by treadmill exercise testing. The ß2 adrenergic receptor protein expression was increased in AS rats independently of exercise. Interestingly, ET restored the protein levels of phosphorylated phospholamban at Serine 16 and preserved the ß-adrenergic receptor responsiveness as visualized by the lower myocardial compliance decline and time to 50% tension development and relaxation during ß-adrenergic stimulation in the IPM than untrained rats. Additionally, AS rats presented higher levels of TNFα and iNOS, which were attenuated by ET. CONCLUSION: Moderate ET improves exercise tolerance, reduces heart failure features, and attenuates diastolic dysfunction. In the myocardium, ET decreases the cross-sectional area of the cardiomyocyte and preserves the ß-adrenergic responsiveness, which reveals that the adjustments in ß-adrenergic signaling contribute to the amelioration of cardiac dysfunction by mild exercise training in aortic stenosis rats.


Subject(s)
Aortic Stenosis, Supravalvular/metabolism , Heart Failure, Diastolic/therapy , Myocytes, Cardiac/metabolism , Physical Conditioning, Animal/physiology , Receptors, Adrenergic, beta/metabolism , Animals , Aortic Stenosis, Supravalvular/therapy , Calcium-Binding Proteins/metabolism , Echocardiography , Exercise Test , Male , Myocardium/metabolism , Myocytes, Cardiac/physiology , Nitric Oxide Synthase Type II/metabolism , Papillary Muscles/physiology , Phosphorylation , Rats , Rats, Wistar , Receptors, Adrenergic, beta/physiology , Signal Transduction/physiology , Tumor Necrosis Factor-alpha/metabolism
15.
Bull Exp Biol Med ; 169(1): 24-28, 2020 May.
Article in English | MEDLINE | ID: mdl-32495169

ABSTRACT

We studied the effect of metformin (100 and 200 mg/kg/day, 4 weeks) on the adenylyl cyclasestimulating effects of ß-agonists and relaxin in the myocardial membranes and on activities of Akt-kinase, an effector component of insulin signaling, and AMP-activated protein kinase (AMPK), a cellular energy sensor, in the myocardium of rats with type 2 diabetes mellitus induced by high-fat diet and streptozotocin. Metformin normalized the ratio of adenylyl cyclase effects of ß1/2- and ß3-agonists in the myocardial membranes, that is reduced in DM2, and restored phosphorylation of Akt-kinase by Ser473 and AMPK by Thr172 in the myocardium of diabetic rats. The effect of metformin in a dose of 200 mg/kg/day was more pronounced. Thus, the cardioprotective effect of metformin is due to its ability to restore the adrenergic and insulin regulation in cardiomyocytes and their energy status.


Subject(s)
Diabetes Mellitus, Experimental/drug therapy , Metformin/therapeutic use , Myocardium/metabolism , Receptors, Adrenergic, beta/drug effects , AMP-Activated Protein Kinases/drug effects , AMP-Activated Protein Kinases/metabolism , Animals , Diabetes Mellitus, Experimental/etiology , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/metabolism , Diet, High-Fat , Heart/drug effects , Male , Metformin/pharmacology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Proto-Oncogene Proteins c-akt/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Wistar , Receptors, Adrenergic, beta/physiology , Signal Transduction/drug effects , Streptozocin
16.
Mol Metab ; 40: 101026, 2020 10.
Article in English | MEDLINE | ID: mdl-32473405

ABSTRACT

OBJECTIVE: Increasing energy expenditure through activation of brown adipose tissue (BAT) thermogenesis is an attractive approach to counteract obesity. It is therefore essential to understand the molecular mechanisms that control BAT functions. Until now several members of the Janus kinase (JAK) - signal transducer and activator of transcription (STAT) pathway have been implicated as being relevant for BAT physiology. However, whether the STAT family member STAT5 is important for the thermogenic property of adipose tissues is unknown. Therefore, we have investigated the role of STAT5 in thermogenic fat in this paper. METHODS: We performed metabolic and molecular analyses using mice that harbor an adipocyte-specific deletion of Stat5a/b alleles. RESULTS: We found that STAT5 is necessary for acute cold-induced temperature maintenance and the induction of lipid mobilization in BAT following ß3-adrenergic stimulation. Moreover, mitochondrial respiration of primary differentiated brown adipocytes lacking STAT5 was diminished. Increased sensitivity to cold stress upon STAT5 deficiency was associated with reduced expression of thermogenic markers including uncoupling protein 1 (UCP1), while decreased stimulated lipolysis was linked to decreased protein kinase A (PKA) activity. Additionally, brown remodeling of white adipose tissue was diminished following chronic ß3-adrenergic stimulation, which was accompanied by a decrease in mitochondrial performance. CONCLUSION: We conclude that STAT5 is essential for the functionality and the ß-adrenergic responsiveness of thermogenic adipose tissue.


Subject(s)
Adipose Tissue, Brown/metabolism , STAT5 Transcription Factor/metabolism , Thermogenesis/physiology , Adipocytes/metabolism , Adipose Tissue, White/metabolism , Animals , Cold-Shock Response/physiology , Energy Metabolism , Female , Lipid Metabolism/physiology , Lipids/physiology , Lipolysis , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/metabolism , Obesity/metabolism , Receptors, Adrenergic, beta/metabolism , Receptors, Adrenergic, beta/physiology , STAT5 Transcription Factor/physiology
17.
J Am Coll Cardiol ; 75(10): 1159-1174, 2020 03 17.
Article in English | MEDLINE | ID: mdl-32164890

ABSTRACT

BACKGROUND: Although cardiomyopathy has emerged as a leading cause of death in Duchenne muscular dystrophy (DMD), limited studies and therapies have emerged for dystrophic heart failure. OBJECTIVES: The purpose of this study was to model DMD cardiomyopathy using DMD patient-specific human induced pluripotent stem cell (hiPSC)-derived cardiomyocytes and to identify physiological changes and future drug therapies. METHODS: To explore and define therapies for DMD cardiomyopathy, the authors used DMD patient-specific hiPSC-derived cardiomyocytes to examine the physiological response to adrenergic agonists and ß-blocker treatment. The authors further examined these agents in vivo using wild-type and mdx mouse models. RESULTS: At baseline and following adrenergic stimulation, DMD hiPSC-derived cardiomyocytes had a significant increase in arrhythmic calcium traces compared to isogenic controls. Furthermore, these arrhythmias were significantly decreased with propranolol treatment. Using telemetry monitoring, the authors observed that mdx mice, which lack dystrophin, had an arrhythmic death when stimulated with isoproterenol; the lethal arrhythmias were rescued, in part, by propranolol pre-treatment. Using single-cell and bulk RNA sequencing (RNA-seq), the authors compared DMD and control hiPSC-derived cardiomyocytes, mdx mice, and control mice (in the presence or absence of propranolol and isoproterenol) and defined pathways that were perturbed under baseline conditions and pathways that were normalized after propranolol treatment in the mdx model. The authors also undertook transcriptome analysis of human DMD left ventricle samples and found that DMD hiPSC-derived cardiomyocytes have dysregulated pathways similar to the human DMD heart. The authors further determined that relatively few patients with DMD see a cardiovascular specialist or receive ß-blocker therapy. CONCLUSIONS: The results highlight mechanisms and therapeutic interventions from human to animal and back to human in the dystrophic heart. These results may serve as a prelude for an adequately powered clinical study that examines the impact of ß-blocker therapy in patients with dystrophinopathies.


Subject(s)
Adrenergic beta-Antagonists/therapeutic use , Cardiomyopathies/drug therapy , Cardiomyopathies/physiopathology , Induced Pluripotent Stem Cells/physiology , Muscular Dystrophy, Duchenne/drug therapy , Muscular Dystrophy, Duchenne/physiopathology , Adolescent , Adrenergic beta-Antagonists/pharmacology , Adult , Animals , Cell Line , Child , Child, Preschool , Female , Humans , Induced Pluripotent Stem Cells/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Receptors, Adrenergic, beta/physiology , Young Adult
18.
Neurosci Lett ; 725: 134878, 2020 04 23.
Article in English | MEDLINE | ID: mdl-32119942

ABSTRACT

The beta-adrenergic receptors of the basolateral amygdala (BLA) are involved in modulating emotional memory formation in the hippocampus. The molecular mechanisms of this involvement are still unclear. In this study, we investigate the effects of the beta-adrenergic receptors of the BLA involvements during the major cellular mechanisms that underlie memory formation in hippocampal sub-regions. Wistar rats were injected with the selective beta receptor agonist, clenbuterol (15 ng/0.5 µl) bilaterally into the BLA. Then, the long-term potentiation (LTP) and the paired-pulse responses were recorded. Control rats were injected by saline at the same volume. Our data indicated that the injection of clenbuterol into the BLA area enhanced the amplitude of the population spike (PS) but not the slope of the excitatory postsynaptic potential (EPSP). In addition, short-term plasticity in the dentate gyrus (DG) region was significantly changed at 500 ms inter-pulse interval. These results suggest that the activation of the beta-adrenergic receptors of the BLA can improve LTP induction, which depends on the short-term plasticity with a mechanism related to the GABAergic transmission. Thus, it can be concluded that the adrenergic system of the BLA engages with long-term and short-term plasticity.


Subject(s)
Adrenergic beta-Agonists/administration & dosage , Basolateral Nuclear Complex/physiology , Dentate Gyrus/cytology , Dentate Gyrus/physiology , Neuronal Plasticity/physiology , Receptors, Adrenergic, beta/physiology , Animals , Basolateral Nuclear Complex/drug effects , Dentate Gyrus/drug effects , Long-Term Potentiation/drug effects , Long-Term Potentiation/physiology , Male , Microinjections/methods , Neuronal Plasticity/drug effects , Rats , Rats, Wistar
19.
Shock ; 53(4): 476-484, 2020 04.
Article in English | MEDLINE | ID: mdl-31232864

ABSTRACT

Severely burned patients suffer from a hypermetabolic syndrome that can last for years after the injury has resolved. The underlying cause of these metabolic alterations most likely involves the persistent elevated catecholamine levels that follow the surge induced by thermal injury. At the cellular level, endoplasmic reticulum (ER) stress in metabolic tissues is a hallmark observed in patients following burn injury and is associated with several detrimental effects. Therefore, ER stress could be the underlying cellular mechanism of persistent hypermetabolism in burned patients. Here, we show that catecholamines induce ER stress and that adreno-receptor blockers reduce stress responses in the HepG2 hepatocyte cell line. Our results also indicate that norepinephrine (NE) significantly induces ER stress in HepG2 cells and 3T3L1 mouse adipocytes. Furthermore, we demonstrate that the alpha-1 blocker, prazosin, and beta blocker, propranolol, block ER stress induced by NE. We also show that the effects of catecholamines in inducing ER stress are cell type-specific, as NE treatment failed to evoke ER stress in human fibroblasts. Thus, these findings reveal the mechanisms used by catecholamines to alter metabolism and suggest inhibition of the receptors utilized by these agents should be further explored as a potential target for the treatment of ER stress-mediated disease.


Subject(s)
Catecholamines/physiology , Endoplasmic Reticulum Stress/physiology , Fibroblasts/physiology , Hep G2 Cells/physiology , Receptors, Adrenergic, alpha/physiology , Receptors, Adrenergic, beta/physiology , Adrenergic alpha-1 Receptor Antagonists/pharmacology , Adrenergic beta-Antagonists/pharmacology , Cell Culture Techniques , Fibroblasts/drug effects , Hep G2 Cells/drug effects , Humans , Prazosin/pharmacology , Propranolol/pharmacology
20.
Behav Pharmacol ; 31(6): 535-543, 2020 09.
Article in English | MEDLINE | ID: mdl-31860562

ABSTRACT

Posttraumatic stress disorder is a mental disorder with a known cause, yet effective behavioral and pharmacotherapies remain elusive for many afflicted patients. Propranolol is suggested to be effective as a fear-reducing agent when paired with behavioral therapy soon after trauma when psychological stress is high, possibly dampening or preventing the later development of posttraumatic stress disorder. In our previous study, we found propranolol efficaciously reduced fear retention induced by reactivation via ß-adrenergic receptors in lateral amygdala. However, it is unclear which subtypes of ß-adrenergic receptors dominate the function of adrenergic activation in lateral amygdala. In this study, we investigated the action of ß1-adrenergic receptor antagonist-metoprolol and ß2-adrenergic receptor antagonist-butoxamine on the retention of conditioned fear memory and synaptic adaptation in the lateral amygdala of rats. We found metoprolol not butoxamine attenuated the reactivation-induced strengthening of fear retention and restored the impaired long-term depression in lateral amygdala. Intra-amygdala infusion of metoprolol not butoxamine attenuated reactivation-induced enhancement of fear retention. Our results suggest that ß1-adrenergic receptor antagonist-metoprolol may be more suitable for the treatment of posttraumatic stress disorder.


Subject(s)
Adrenergic beta-1 Receptor Antagonists/pharmacology , Amygdala/drug effects , Fear/drug effects , Long-Term Synaptic Depression/drug effects , Memory/drug effects , Metoprolol/pharmacology , Stress Disorders, Post-Traumatic/drug therapy , Amygdala/physiology , Animals , Butoxamine/pharmacology , Male , Metoprolol/therapeutic use , Rats , Rats, Sprague-Dawley , Receptors, Adrenergic, beta/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...