Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Neuropharmacology ; 184: 108408, 2021 02 15.
Article in English | MEDLINE | ID: mdl-33220305

ABSTRACT

Ongoing activity in nociceptors, a driver of spontaneous pain, can be generated in dorsal root ganglion neurons in the absence of sensory generator potentials if one or more of three neurophysiological alterations occur - prolonged depolarization of resting membrane potential (RMP), hyperpolarization of action potential (AP) threshold, and/or increased amplitude of depolarizing spontaneous fluctuations of membrane potential (DSFs) to bridge the gap between RMP and AP threshold. Previous work showed that acute, sustained exposure to serotonin (5-HT) hyperpolarized AP threshold and potentiated DSFs, leading to ongoing activity if a separate source of maintained depolarization was present. Cellular signaling pathways that increase DSF amplitude and promote ongoing activity acutely in nociceptors are not known for any neuromodulator. Here, isolated DRG neurons from male rats were used to define the pathway by which low concentrations of 5-HT enhance DSFs, hyperpolarize AP threshold, and promote ongoing activity. A selective 5-HT4 receptor antagonist blocked these 5-HT-induced hyperexcitable effects, while a selective 5-HT4 agonist mimicked the effects of 5-HT. Inhibition of cAMP effectors, protein kinase A (PKA) and exchange protein activated by cAMP (EPAC), attenuated 5-HT's hyperexcitable effects, but a blocker of hyperpolarization-activated cyclic nucleotide-gated (HCN) channels had no significant effect. 5-HT4-dependent PKA activation was specific to DRG neurons that bind isolectin B4 (a nonpeptidergic nociceptor marker). 5-HT's effects on AP threshold, DSFs, and ongoing activity were mimicked by a cAMP analog. Sustained exposure to 5-HT promotes ongoing activity in nonpeptidergic nociceptors through the Gs-coupled 5-HT4 receptor and downstream cAMP signaling involving both PKA and EPAC.


Subject(s)
Cyclic AMP/metabolism , Ganglia, Spinal/metabolism , Neurons/metabolism , Receptors, Serotonin, 5-HT4/metabolism , Serotonin 5-HT4 Receptor Agonists/metabolism , Serotonin/metabolism , Action Potentials/drug effects , Action Potentials/physiology , Animals , Cells, Cultured , Dose-Response Relationship, Drug , Ganglia, Spinal/drug effects , Male , Neurons/drug effects , Rats , Rats, Sprague-Dawley , Serotonin/pharmacology , Serotonin 5-HT4 Receptor Agonists/pharmacology
2.
Sci Rep ; 7(1): 5522, 2017 07 17.
Article in English | MEDLINE | ID: mdl-28717248

ABSTRACT

Adenylyl cyclase type 9 (AC9) is found tightly associated with the scaffolding protein Yotiao and the IKs ion channel in heart. But apart from potential IKs regulation, physiological roles for AC9 are unknown. We show that loss of AC9 in mice reduces less than 3% of total AC activity in heart but eliminates Yotiao-associated AC activity. AC9-/- mice exhibit no structural abnormalities but show a significant bradycardia, consistent with AC9 expression in sinoatrial node. Global changes in PKA phosphorylation patterns are not altered in AC9-/- heart, however, basal phosphorylation of heat shock protein 20 (Hsp20) is significantly decreased. Hsp20 binds AC9 in a Yotiao-independent manner and deletion of AC9 decreases Hsp20-associated AC activity in heart. In addition, expression of catalytically inactive AC9 in neonatal cardiomyocytes decreases isoproterenol-stimulated Hsp20 phosphorylation, consistent with an AC9-Hsp20 complex. Phosphorylation of Hsp20 occurs largely in ventricles and is vital for the cardioprotective effects of Hsp20. Decreased Hsp20 phosphorylation suggests a potential baseline ventricular defect for AC9-/-. Doppler echocardiography of AC9-/- displays a decrease in the early ventricular filling velocity and ventricular filling ratio (E/A), indicative of grade 1 diastolic dysfunction and emphasizing the importance of local cAMP production in the context of macromolecular complexes.


Subject(s)
Adenylyl Cyclases/metabolism , HSP20 Heat-Shock Proteins/metabolism , Ventricular Function, Left/physiology , A Kinase Anchor Proteins/metabolism , Adenylyl Cyclases/genetics , Animals , Bradycardia/etiology , Bradycardia/veterinary , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Echocardiography , Female , Isoproterenol/pharmacology , KCNQ1 Potassium Channel/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocardium/metabolism , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Phosphorylation/drug effects , Sinoatrial Node/metabolism
3.
Mol Med ; 18: 556-64, 2012 May 09.
Article in English | MEDLINE | ID: mdl-22331028

ABSTRACT

The molecular mechanisms determining magnitude and duration of inflammatory pain are still unclear. We assessed the contribution of G protein-coupled receptor kinase (GRK)-6 to inflammatory hyperalgesia in mice. We showed that GRK6 is a critical regulator of severity and duration of cytokine-induced hyperalgesia. In GRK6⁻/⁻ mice, a significantly lower dose (100 times lower) of intraplantar interleukin (IL)-1ß was sufficient to induce hyperalgesia compared with wild-type (WT) mice. In addition, IL-1ß hyperalgesia lasted much longer in GRK6⁻/⁻ mice than in WT mice (8 d in GRK6⁻/⁻ versus 6 h in WT mice). Tumor necrosis factor (TNF)-α-induced hyperalgesia was also enhanced and prolonged in GRK6⁻/⁻ mice. In vitro, IL-1ß-induced p38 phosphorylation in GRK6⁻/⁻ dorsal root ganglion (DRG) neurons was increased compared with WT neurons. In contrast, IL-1ß only induced activation of the phosphatidylinositol (PI) 3-kinase/Akt pathway in WT neurons, but not in GRK6⁻/⁻ neurons. In vivo, p38 inhibition attenuated IL-1ß- and TNF-α-induced hyperalgesia in both genotypes. Notably, however, whereas PI 3-kinase inhibition enhanced and prolonged hyperalgesia in WT mice, it did not have any effect in GRK6-deficient mice. The capacity of GRK6 to regulate pain responses was also apparent in carrageenan-induced hyperalgesia, since thermal and mechanical hypersensitivity was significantly prolonged in GRK6⁻/⁻ mice. Finally, GRK6 expression was reduced in DRGs of mice with chronic neuropathic or inflammatory pain. Collectively, these findings underline the potential role of GRK6 in pathological pain. We propose the novel concept that GRK6 acts as a kinase that constrains neuronal responsiveness to IL-1ß and TNF-α and cytokine-induced hyperalgesia via biased cytokine-induced p38 and PI 3-kinase/Akt activation.


Subject(s)
Cytokines/toxicity , G-Protein-Coupled Receptor Kinases/metabolism , Hyperalgesia/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Cytokines/administration & dosage , Dinoprostone/administration & dosage , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , Female , G-Protein-Coupled Receptor Kinases/genetics , Gene Expression Regulation , Hyperalgesia/chemically induced , Hyperalgesia/genetics , Inflammation/genetics , Inflammation/metabolism , Interleukin-1beta/administration & dosage , Mice , Mice, Knockout , Neuralgia/genetics , Neuralgia/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Tumor Necrosis Factor-alpha/administration & dosage , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
4.
Brain Behav Immun ; 25(6): 1055-60, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21473908

ABSTRACT

Pain is a hallmark of tissue damage and inflammation promoting tissue protection and thereby contributing to repair. Therefore, transient acute pain is an important feature of the adaptive response to damage. However, in a significant number of cases, pain persists for months to years after the problem that originally caused the pain has resolved. Such chronic pain is maladaptive as it no longer serves a protective aim. Chronic pain is debilitating, both physiologically and psychologically, and treatments to provide relief from chronic pain are often ineffective. The neurobiological mechanisms underlying the transition from adaptive acute pain to maladaptive chronic pain are only partially understood. In this review, we will summarize recent evidence that a kinase known as G protein-coupled receptor kinase (GRK2) is a key regulator of the transition from acute to chronic inflammatory pain. Our recent studies have shown that mice with a reduction in the cellular level of GRK2 develop chronic hyperalgesia in response to inflammatory mediators that induce only transient hyperalgesia in WT mice. This finding is clinically relevant because rodent models of chronic pain are associated with reduced cellular levels of GRK2. We propose that GRK2 is a newly discovered major player in the regulation of chronic pain. The pathways regulated by this kinase may open up new avenues for development of treatment strategies that target the cause, and not the symptoms of chronic pain.


Subject(s)
Chronic Pain/enzymology , G-Protein-Coupled Receptor Kinase 2/physiology , Inflammation/physiopathology , Microglia/enzymology , Neuralgia/enzymology , Acute Pain/enzymology , Acute Pain/physiopathology , Animals , Chronic Pain/physiopathology , Cytokines/metabolism , Disease Models, Animal , Forecasting , G-Protein-Coupled Receptor Kinase 2/deficiency , G-Protein-Coupled Receptor Kinase 2/genetics , Gene Expression Regulation, Enzymologic , Humans , Hyperalgesia/enzymology , Hyperalgesia/physiopathology , Macrophages/enzymology , Mice , Mice, Knockout , Mice, Transgenic , Microglia/metabolism , Neuralgia/physiopathology , Receptors, Interleukin-1/physiology , Sciatic Nerve/injuries , Signal Transduction , Spinal Cord/pathology , Spinal Cord/physiopathology , Spinal Nerves/injuries
5.
Pain ; 152(7): 1649-1658, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21514055

ABSTRACT

Epinephrine (EPI) contributes to hyperalgesia in inflammatory and stress conditions. EPI signals via adrenoceptors, which are regulated by G protein-coupled receptor kinase 2 (GRK2). We previously reported that GRK2 is decreased in nociceptors during chronic inflammation. Herein, we investigated whether GRK2 modulates EPI-induced mechanical and thermal hyperalgesia by using GRK2(+/-) mice, which express 50% of the GRK2 protein. We demonstrate for the first time that EPI-induced mechanical as well as thermal hyperalgesia is prolonged to approximately 21 days in GRK2(+/-) mice, whereas it lasts only 3 to 4 days in wild-type mice. Using cell- specific GRK2-deficient mice, we further show that a low level of GRK2 in primary sensory neurons is critical for this prolongation of EPI-induced hyperalgesia. Low GRK2 in microglia had only a small effect on EPI-induced hyperalgesia. Low GRK2 in astrocytes did not alter EPI-induced hyperalgesia. EPI-induced hyperalgesia was prolonged similarly in mice with tamoxifen-induced homozygous or heterozygous deletion of GRK2. In terms of EPI signalling pathways, the protein kinase A (PKA) inhibitor H-89 inhibited EPI-induced mechanical hyperalgesia in wild-type mice, whereas H-89 had no effect in mice with low GRK2 in sensory neurons (SNS-GRK2(+/-) mice). Conversely, intraplantar injection of the protein kinase Cε PKCε inhibitor TAT-PKC(εv1-2) inhibited hyperalgesia in sensory neuron specific (SNS)-GRK2(+/-) mice and not in wild-type mice. These results indicate that low GRK2 in primary sensory neurons switches EPI-induced signalling from a protein kinase A-dependent toward a PKCε-dependent pathway that ultimately mediates prolonged EPI-induced hyperalgesia.


Subject(s)
Epinephrine/toxicity , G-Protein-Coupled Receptor Kinase 2/metabolism , Hyperalgesia/chemically induced , Pain Threshold/drug effects , Sensory Receptor Cells/metabolism , Signal Transduction/drug effects , Adrenergic alpha-Antagonists/therapeutic use , Adrenergic beta-Antagonists/therapeutic use , Animals , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Female , G-Protein-Coupled Receptor Kinase 2/deficiency , Ganglia, Spinal/pathology , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Hyperalgesia/drug therapy , Hyperalgesia/genetics , Hyperalgesia/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neuroglia/drug effects , Neuroglia/metabolism , Pain Measurement , Phentolamine/therapeutic use , Propanolamines/therapeutic use , Sensory Receptor Cells/drug effects , Signal Transduction/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...