Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters








Publication year range
1.
Pain ; 164(7): 1524-1536, 2023 Jul 01.
Article in English | MEDLINE | ID: mdl-36972485

ABSTRACT

ABSTRACT: Low-frequency sinusoidal current applied to human skin evokes local axon reflex flare and burning pain, indicative of C-fibre activation. Because topical cooling works well as a local analgesic, we examined the effect of cooling on human pain ratings to sinusoidal and rectangular profiles of constant current stimulation. Unexpectedly, pain ratings increased upon cooling the skin from 32 to 18°C. To explore this paradoxical observation, the effects of cooling on C-fibre responses to stimulation with sinusoidal and rectangular current profiles were determined in ex vivo segments of mouse sural and pig saphenous nerve. As expected by thermodynamics, the absolute value of electrical charge required to activate C-fibre axons increased with cooling from 32°C to 20°C, irrespective of the stimulus profile used. However, for sinusoidal stimulus profiles, cooling enabled a more effective integration of low-intensity currents over tens of milliseconds resulting in a delayed initiation of action potentials. Our findings indicate that the paradoxical cooling-induced enhancement of electrically evoked pain in people can be explained by an enhancement of C-fibre responsiveness to slow depolarization at lower temperatures. This property may contribute to symptoms of enhanced cold sensitivity, especially cold allodynia, associated with many forms of neuropathic pain.


Subject(s)
Capillaries , Neuralgia , Humans , Animals , Mice , Swine , Skin/innervation , Nerve Fibers, Unmyelinated/physiology , Hyperalgesia
2.
Front Cell Neurosci ; 16: 843225, 2022.
Article in English | MEDLINE | ID: mdl-35496916

ABSTRACT

TRPM3 is a calcium-permeable cation channel expressed in a range of sensory neurons that can be activated by heat and the endogenous steroid pregnenolone sulfate (PS). During inflammation, the expression and function of TRPM3 are both augmented in somatosensory nociceptors. However, in isolated dorsal root ganglion (DRG) neurons application of inflammatory mediators like prostaglandins and bradykinin (BK) inhibit TRPM3. Therefore, the aim of this study was to examine the effect of preceding activation of cultured 1 day old mouse DRG neurons by the inflammatory mediator BK on TRPM3-mediated calcium responses. Calcium signals were recorded using the intensity-based dye Fluo-8. We found that TRPM3-mediated calcium responses to PS were enhanced by preceding application of BK in cells that responded to BK with a calcium signal, indicating BK receptor (BKR) expression. The majority of cells that co-expressed TRPM3 and BKRs also expressed TRPV1, however, only a small fraction co-expressed TRPA1, identified by calcium responses to capsaicin and supercinnamaldehyde, respectively. Signaling and trafficking pathways responsible for sensitization of TRPM3 following BK were characterized using inhibitors of second messenger signaling cascades and exocytosis. Pharmacological blockade of protein kinase C, calcium-calmodulin-dependent protein kinase II and diacylglycerol (DAG) lipase did not affect BK-induced sensitization, but inhibition of DAG kinase did. In addition, release of calcium from intracellular stores using thapsigargin also resulted in TRPM3 sensitization. Finally, BK did not sensitize TRPM3 in the presence of exocytosis inhibitors. Collectively, we show that preceding activation of DRG neurons by BK sensitized TRPM3-mediated calcium responses to PS. Our results indicate that BKR-mediated activation of intracellular signaling pathways comprising DAG kinase, calcium and exocytosis may contribute to TRPM3 sensitization during inflammation.

4.
Sci Rep ; 11(1): 21490, 2021 11 02.
Article in English | MEDLINE | ID: mdl-34728705

ABSTRACT

Single intradermal injections of nerve growth factor (NGF) evoke prolonged but temporally distinct sensitization patterns to somatosensory stimuli. Focal administration of the non-histaminergic pruritogen cowhage but not histamine resulted in elevated itch at day 21 after NGF administration. Here, we injected bovine adrenal medulla peptide 8-22 (BAM8-22), ß-alanine (ß-ALA) and endothelin-1 (ET-1) into NGF-treated skin of 11 healthy volunteers and investigated the corresponding itch/pain and flare reactions. ß-ALA was the weakest pruritogen, while BAM8-22 and ET-1 were equally potent as histamine. NGF did not sensitize itch or flare reactions induced by any compound, but injection and evoked pain were increased at day 21 and 49. The involvement of histamine H1 receptors in itch was explored in eight subjects after oral cetirizine. ET-1-induced itch and flare were significantly reduced. BAM8-22 and ß-ALA itch were not affected, but flare responses after BAM8-22 reduced by 50%. The results indicate that a single NGF injection does not sensitize for experimentally induced itch but increases pain upon pruritogen injection. In healthy humans, pruritic and algetic processing appear differentially regulated by NGF. However, in patients suffering chronic itch, prolonged elevation of NGF-levels under inflammatory conditions may contribute to elevated itch.


Subject(s)
Endothelin-1/pharmacology , Nerve Growth Factor/adverse effects , Pain/drug therapy , Peptide Fragments/pharmacology , Pruritus/drug therapy , Skin/drug effects , beta-Alanine/pharmacology , Adult , Animals , Cattle , Female , Humans , Male , Pain/chemically induced , Pain/pathology , Pruritus/chemically induced , Pruritus/pathology , Skin/pathology
5.
Elife ; 102021 04 23.
Article in English | MEDLINE | ID: mdl-33891544

ABSTRACT

In humans, intradermal administration of ß-alanine (ALA) and bovine adrenal medulla peptide 8-22 (BAM8-22) evokes the sensation of itch. Currently, it is unknown which human dorsal root ganglion (DRG) neurons express the receptors of these pruritogens, MRGPRD and MRGPRX1, respectively, and which cutaneous afferents these pruritogens activate in primate. In situ hybridization studies revealed that MRGPRD and MRGPRX1 are co-expressed in a subpopulation of TRPV1+ human DRG neurons. In electrophysiological recordings in nonhuman primates (Macaca nemestrina), subtypes of polymodal C-fiber nociceptors are preferentially activated by ALA and BAM8-22, with significant overlap. When pruritogens ALA, BAM8-22, and histamine, which activate different subclasses of C-fiber afferents, are administered in combination, human volunteers report itch and nociceptive sensations similar to those induced by a single pruritogen. Our results provide evidence for differences in pruriceptive processing between primates and rodents, and do not support the spatial contrast theory of coding of itch and pain.


Subject(s)
Ganglia, Spinal/physiopathology , Nociceptors/physiology , Peptide Fragments/adverse effects , Pruritus/physiopathology , Receptors, G-Protein-Coupled/genetics , beta-Alanine/adverse effects , Adult , Animals , Female , Ganglia, Spinal/drug effects , Histamine/administration & dosage , Humans , Macaca nemestrina/physiology , Male , Middle Aged , Nociceptors/drug effects , Pruritus/chemically induced , Receptors, G-Protein-Coupled/metabolism , Young Adult
6.
Front Med (Lausanne) ; 7: 627617, 2020.
Article in English | MEDLINE | ID: mdl-33553220

ABSTRACT

Administration of chemicals (pruritogens) into the skin evokes itch based on signal transduction mechanisms that generate action potentials mainly in mechanically sensitive and insensitive primary afferent C-fibers (pruriceptors). These signals from peripheral neurons are processed in spinal and supra-spinal centers of the central nervous system and finally generate the sensation of itch. Compared to chemical stimulation, electrical activation of pruriceptors would allow for better temporal control and thereby a more direct functional assessment of their activation. Here, we review the electrical stimulation paradigms which were used to evoke itch in humans in the past. We further evaluate recent attempts to explore electrically induced itch in atopic dermatitis patients. Possible mechanisms underlying successful pruritus generation in chronic itch patients by transdermal slowly depolarizing electrical stimulation are discussed.

7.
Sci Transl Med ; 11(500)2019 07 10.
Article in English | MEDLINE | ID: mdl-31292265

ABSTRACT

There is a major clinical need for new therapies for the treatment of chronic itch. Many of the molecular components involved in itch neurotransmission are known, including the neuropeptide NPPB, a transmitter required for normal itch responses to multiple pruritogens in mice. Here, we investigated the potential for a novel strategy for the treatment of itch that involves the inhibition of the NPPB receptor NPR1 (natriuretic peptide receptor 1). Because there are no available effective human NPR1 (hNPR1) antagonists, we performed a high-throughput cell-based screen and identified 15 small-molecule hNPR1 inhibitors. Using in vitro assays, we demonstrated that these compounds specifically inhibit hNPR1 and murine NPR1 (mNPR1). In vivo, NPR1 antagonism attenuated behavioral responses to both acute itch- and chronic itch-challenged mice. Together, our results suggest that inhibiting NPR1 might be an effective strategy for treating acute and chronic itch.


Subject(s)
Ganglia, Spinal/metabolism , Pruritus/drug therapy , Receptors, Atrial Natriuretic Factor/antagonists & inhibitors , Animals , Behavior, Animal , Cell-Free System , Dermatitis, Contact/drug therapy , Disease Models, Animal , Ganglia, Spinal/pathology , Humans , Mice, Inbred C57BL , Mice, Knockout , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Pruritus/pathology , Receptors, Atrial Natriuretic Factor/agonists , Receptors, Atrial Natriuretic Factor/metabolism , Reproducibility of Results , Signal Transduction/drug effects , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Small Molecule Libraries/therapeutic use
8.
Cell Rep ; 26(13): 3561-3573.e4, 2019 03 26.
Article in English | MEDLINE | ID: mdl-30917312

ABSTRACT

Itch is an unpleasant skin sensation that can be triggered by exposure to many chemicals, including those released by mast cells. The natriuretic polypeptide b (Nppb)-expressing class of sensory neurons, when activated, elicits scratching responses in mice, but it is unclear which itch-inducing agents stimulate these cells and the receptors involved. Here, we identify receptors expressed by Nppb neurons and demonstrate the functional importance of these receptors as sensors of endogenous pruritogens released by mast cells. Our search for receptors in Nppb neurons reveals that they express leukotriene, serotonin, and sphingosine-1-phosphate receptors. Targeted cell ablation, calcium imaging of primary sensory neurons, and conditional receptor knockout studies demonstrate that these receptors induce itch by the direct stimulation of Nppb neurons and neurotransmission through the canonical gastrin-releasing peptide (GRP)-dependent spinal cord itch pathway. Together, our results define a molecular and cellular pathway for mast cell-induced itch.


Subject(s)
Mast Cells/physiology , Pruritus , Receptors, Atrial Natriuretic Factor/physiology , Receptors, Cell Surface/physiology , Sensory Receptor Cells/physiology , Animals , Cells, Cultured , Female , Male , Mice , Receptors, Leukotriene/physiology , Receptors, Serotonin, 5-HT1/physiology , Sensory Receptor Cells/metabolism , Sphingosine-1-Phosphate Receptors/physiology , Transcriptome
9.
Neurosci Lett ; 690: 167-170, 2019 01 18.
Article in English | MEDLINE | ID: mdl-30355519

ABSTRACT

How is temperature detected and how is the resulting sensory information synthesized to produce appropriate thermosensory responses? Research in the last few years has gone a long way to answering the first part of this question. Excitingly, recent research has uncovered some of the ways sensory inputs are processed spinally, as well as identifying supra-spinal centers involved in processing responses to thermal stimuli. In this review, we explore the new areas of research that have contributed to our comprehension of the way the peripheral sensory neurons are tuned in addition to the receptors used to differentiate thermal stimuli. We also describe recent work which begins to illuminate the processing of primary sensory signals by the spinal cord and regions of the brain.


Subject(s)
Mammals/physiology , Thermoreceptors/physiology , Thermosensing/physiology , Animals , Body Temperature/physiology , Brain/physiology , Neural Pathways/physiology , Spinal Cord/physiology , Transient Receptor Potential Channels/physiology
10.
Nat Neurosci ; 21(6): 894, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29674654

ABSTRACT

In the version of this article initially published online, the labels were switched for the right-hand pair of bars in Fig. 4e. The left one of the two should be Chloroquine + veh, the right one Chloroquine + CNO. The error has been corrected in the print, HTML and PDF versions of the article.

11.
Nat Neurosci ; 21(5): 707-716, 2018 05.
Article in English | MEDLINE | ID: mdl-29556030

ABSTRACT

Stimuli that elicit itch are detected by sensory neurons that innervate the skin. This information is processed by the spinal cord; however, the way in which this occurs is still poorly understood. Here we investigated the neuronal pathways for itch neurotransmission, particularly the contribution of the neuropeptide somatostatin. We find that in the periphery, somatostatin is exclusively expressed in Nppb+ neurons, and we demonstrate that Nppb+somatostatin+ cells function as pruriceptors. Employing chemogenetics, pharmacology and cell-specific ablation methods, we demonstrate that somatostatin potentiates itch by inhibiting inhibitory dynorphin neurons, which results in disinhibition of GRPR+ neurons. Furthermore, elimination of somatostatin from primary afferents and/or from spinal interneurons demonstrates differential involvement of the peptide released from these sources in itch and pain. Our results define the neural circuit underlying somatostatin-induced itch and characterize a contrasting antinociceptive role for the peptide.


Subject(s)
Neural Pathways/physiopathology , Pain/physiopathology , Pruritus/physiopathology , Somatostatin/metabolism , Animals , Dynorphins/metabolism , Female , Ganglia, Spinal/metabolism , Male , Mice , Mice, Inbred C57BL , Optogenetics , Pain/metabolism , Pruritus/metabolism , Receptors, Atrial Natriuretic Factor/biosynthesis , Receptors, Atrial Natriuretic Factor/metabolism , Receptors, Purinergic/metabolism , Receptors, Somatostatin/antagonists & inhibitors , Receptors, Somatostatin/genetics , Sensory Receptor Cells , Somatostatin/biosynthesis , Spinal Cord/cytology , Spinal Cord/physiopathology
12.
Cell Rep ; 21(10): 2760-2771, 2017 Dec 05.
Article in English | MEDLINE | ID: mdl-29212024

ABSTRACT

Piezo2 is a mechanically activated ion channel required for touch discrimination, vibration detection, and proprioception. Here, we discovered that Piezo2 is extensively spliced, producing different Piezo2 isoforms with distinct properties. Sensory neurons from both mice and humans express a large repertoire of Piezo2 variants, whereas non-neuronal tissues express predominantly a single isoform. Notably, even within sensory ganglia, we demonstrate the splicing of Piezo2 to be cell type specific. Biophysical characterization revealed substantial differences in ion permeability, sensitivity to calcium modulation, and inactivation kinetics among Piezo2 splice variants. Together, our results describe, at the molecular level, a potential mechanism by which transduction is tuned, permitting the detection of a variety of mechanosensory stimuli.


Subject(s)
Ion Channels/metabolism , Mechanotransduction, Cellular/physiology , Alternative Splicing/genetics , Animals , Electrophysiology , Female , HEK293 Cells , Humans , In Situ Hybridization , Ion Channels/genetics , Male , Mechanotransduction, Cellular/genetics , Mice , Mice, Inbred C57BL , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA Splicing/genetics , Reverse Transcriptase Polymerase Chain Reaction
13.
Mol Endocrinol ; 29(3): 445-59, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25584415

ABSTRACT

Transcriptional activity of signal transducer and activator of transcription-3 (STAT-3) is a key element in the central regulation of appetite and energy homeostasis. Activation of hypothalamic STAT-3 has been attributed to cytokine-promoted phosphorylation at tyrosine-705 (Tyr-705). In nonhypothalamic cells, STAT-3 is also phosphorylated at serine-727 (Ser-727), but the functional significance of Ser-727 in the regulation of hypothalamic STAT-3 is not known. We used 2 hypothalamic cell lines and analyzed the effects of various hormones on STAT-3-dependent reporter gene activity and observed that IFN-γ, epidermal growth factor (EGF), and bradykinin (BK) induce similar STAT-3 reporter activation. EGF and BK solely increased Ser-727 and IFN-γ increased Tyr-705 phosphorylation of STAT-3. Specific inhibition of ERK-1/2 activity blocked EGF- and BK-induced STAT-3 activation and Ser-727 phosphorylation. BK-induced ERK-1/2 activation occurred via EGF receptor transactivation. Consequently, the BK-mediated effects on STAT-3 were blocked by a specific EGF receptor antagonist. Next, we analyzed the effects of IFN-γ and EGF on the expression of the STAT-3-dependent genes thyroliberin-releasing hormone and suppressors of cytokine signaling-3. EGF but not IFN-γ enhanced thyroliberin-releasing hormone expression via STAT-3. With regard to suppressors of cytokine signaling-3, we observed prolonged expression induced by IFN-γ and a transient effect of EGF that required coactivation of the activator protein-1. Thus, EGF-promoted Ser-727 phosphorylation by ERK-1/2 is not only sufficient to fully activate hypothalamic STAT-3, but, in terms of targeted genes and required cofactors, entails distinct modes of STAT-3 actions compared with IFN-γ-induced Tyr-705 phosphorylation.


Subject(s)
Hypothalamus/metabolism , Phosphoserine/metabolism , Phosphotyrosine/metabolism , STAT3 Transcription Factor/metabolism , Animals , Bradykinin/pharmacology , Cell Line , Epidermal Growth Factor/pharmacology , ErbB Receptors/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Genes, Reporter , Humans , Interferon-gamma/pharmacology , Ligands , Melanocyte-Stimulating Hormones/pharmacology , Mice , Neurons/drug effects , Neurons/metabolism , Neuropeptide Y/metabolism , Phosphorylation/drug effects , Promoter Regions, Genetic/genetics , Receptors, Cytokine/metabolism , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins/metabolism , Thyrotropin-Releasing Hormone/genetics , Thyrotropin-Releasing Hormone/metabolism , Transcriptional Activation/drug effects
15.
Pharmacol Rev ; 66(3): 570-97, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24867890

ABSTRACT

Signaling by heptahelical G protein-coupled receptors (GPCR) regulates many vital body functions. Consequently, dysfunction of GPCR signaling leads to pathologic states, and approximately 30% of all modern clinical drugs target GPCR. One decade ago, an entire new GPCR family was discovered, which was recently named MAS-related G protein-coupled receptors (MRGPR) by the HUGO Gene Nomenclature Committee. The MRGPR family consists of ∼40 members that are grouped into nine distinct subfamilies (MRGPRA to -H and -X) and are predominantly expressed in primary sensory neurons and mast cells. All members are formally still considered "orphan" by the Committee on Receptor Nomenclature and Drug Classification of the International Union of Basic and Clinical Pharmacology. However, several distinct peptides and amino acids are discussed as potential ligands, including ß-alanine, angiotensin-(1-7), alamandine, GABA, cortistatin-14, and cleavage products of proenkephalin, pro-opiomelanocortin, prodynorphin, or proneuropeptide-FF-A. The full spectrum of biologic roles of all MRGPR is still ill-defined, but there is evidence pointing to a role of distinct MRGPR subtypes in nociception, pruritus, sleep, cell proliferation, circulation, and mast cell degranulation. This review article summarizes findings published in the last 10 years on the phylogenetic relationships, pharmacology, signaling, physiology, and agonist-promoted regulation of all MRGPR subfamilies. Furthermore, we highlight interactions between MRGPR and other hormonal systems, paying particular attention to receptor multimerization and morphine tolerance. Finally, we discuss the challenges the field faces presently and emphasize future directions of research.


Subject(s)
Receptors, G-Protein-Coupled/metabolism , Signal Transduction/physiology , Animals , Humans , Ligands , Mast Cells/metabolism , Molecular Targeted Therapy , Receptors, G-Protein-Coupled/drug effects , Receptors, G-Protein-Coupled/physiology , Sensory Receptor Cells/metabolism
16.
PLoS One ; 8(3): e58756, 2013.
Article in English | MEDLINE | ID: mdl-23505557

ABSTRACT

Primate-specific Mas-related G protein-coupled receptors-X1 (MRGPR-X1) are highly enriched in dorsal root ganglia (DRG) neurons and induce acute pain. Herein, we analyzed effects of MRGPR-X1 on serum response factors (SRF) or nuclear factors of activated T cells (NFAT), which control expression of various markers of chronic pain. Using HEK293, DRG neuron-derived F11 cells and cultured rat DRG neurons recombinantly expressing human MRGPR-X1, we found activation of a SRF reporter gene construct and induction of the early growth response protein-1 via extracellular signal-regulated kinases-1/2 known to play a significant role in the development of inflammatory pain. Furthermore, we observed MRGPR-X1-induced up-regulation of the chemokine receptor 2 (CCR2) via NFAT, which is considered as a key event in the onset of neuropathic pain and, so far, has not yet been described for any endogenous neuropeptide. Up-regulation of CCR2 is often associated with increased release of its endogenous agonist chemokine ligand 2 (CCL2). We also found MRGPR-X1-promoted release of CCL2 in a human connective tissue mast cell line endogenously expressing MRGPR-X1. Thus, we provide first evidence to suggest that MRGPR-X1 induce expression of chronic pain markers in DRG neurons and propose a so far unidentified signaling circuit that enhances chemokine signaling by acting on two distinct yet functionally co-operating cell types. Given the important role of chemokine signaling in pain chronification, we propose that interruption of this signaling circuit might be a promising new strategy to alleviate chemokine-promoted pain.


Subject(s)
Chemokine CCL2/metabolism , Ganglia, Spinal/metabolism , Mast Cells/metabolism , Receptors, CCR2/genetics , Receptors, G-Protein-Coupled/metabolism , Sensory Receptor Cells/metabolism , Animals , Bradykinin/pharmacology , Calcineurin/metabolism , Calcium/metabolism , Cell Line , Early Growth Response Protein 1/genetics , Early Growth Response Protein 1/metabolism , Enzyme Activation/drug effects , Gene Expression Regulation/drug effects , Genes, fos , HEK293 Cells , Humans , Inositol 1,4,5-Trisphosphate Receptors/genetics , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , NFATC Transcription Factors/metabolism , Peptide Fragments/pharmacology , Rats , Receptors, CCR2/metabolism , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/genetics , Serum Response Factor/metabolism , Ternary Complex Factors/metabolism
17.
J Biol Chem ; 287(49): 40956-71, 2012 Nov 30.
Article in English | MEDLINE | ID: mdl-23074220

ABSTRACT

Sensory neuron-specific Mas-related G protein-coupled receptors-X1 (MRGPR-X1) are primate-specific proteins that are exclusively expressed in primary sensory neurons and provoke pain in humans. Hence, MRGPR-X1 represent promising targets for future pain therapy, but signaling pathways activated by MRGPR-X1 are poorly understood. The transient receptor potential cation channel V1 (TRPV1) is also expressed in primary sensory neurons and detects painful stimuli such as protons and heat. G(q)-promoted signaling has been shown to sensitize TRPV1 via protein kinase C (PKC)-dependent phosphorylation. In addition, recent studies suggested TRPV1 activation via a G(q)-mediated mechanism involving diacylglycerol (DAG) or phosphatidylinositol-4,5-bisphosphate (PIP(2)). However, it is not clear if DAG-promoted TRPV1 activation occurs independently from classic TRPV1 activation modes induced by heat and protons. Herein, we analyzed putative functional interactions between MRGPR-X1 and TRPV1 in a previously reported F11 cell line stably over-expressing MRGPR-X1. First, we found that MRGPR-X1 sensitized TRPV1 to heat and protons in a PKC-dependent manner. Second, we observed direct MRGPR-X1-mediated TRPV1 activation independent of MRGPR-X1-induced Ca(2+)-release and PKC activity or other TRPV1 affecting enzymes such as lipoxygenase, extracellular signal-regulated kinases-1/2, sarcoma, or phosphoinositide 3-kinase. Investigating several TRPV1 mutants, we observed that removal of the TRPV1 binding site for DAG and of the putative PIP(2) sensor decreased MRGPR-X1-induced TRPV1 activation by 71 and 43%, respectively. Therefore, we demonstrate dual functional interactions between MRGPR-X1 and TRPV1, resulting in PKC-dependent TRPV1 sensitization and DAG/PIP(2)-mediated activation. The molecular discrimination between TRPV1 sensitization and activation may help improve the specificity of current pain therapies.


Subject(s)
GTP-Binding Proteins/metabolism , Gene Expression Regulation , Neurons/metabolism , Receptors, G-Protein-Coupled/metabolism , Sensory Receptor Cells/metabolism , TRPV Cation Channels/metabolism , Analgesics/pharmacology , Animals , Cell Line , Chronic Pain/drug therapy , Diglycerides/metabolism , Enzyme Activation , Enzyme Inhibitors/pharmacology , Genetic Vectors , Humans , Manganese/pharmacology , Mice , Pain Management , Phosphatidylinositols/metabolism , Protein Kinase C/metabolism , Rats , Signal Transduction
18.
PLoS One ; 7(5): e35955, 2012.
Article in English | MEDLINE | ID: mdl-22570693

ABSTRACT

The dorsal cochlear nucleus (DCN) is a first relay of the central auditory system as well as a site for integration of multimodal information. Vesicular glutamate transporters VGLUT-1 and VGLUT-2 selectively package glutamate into synaptic vesicles and are found to have different patterns of organization in the DCN. Whereas auditory nerve fibers predominantly co-label with VGLUT-1, somatosensory inputs predominantly co-label with VGLUT-2. Here, we used retrograde and anterograde transport of fluorescent conjugated dextran amine (DA) to demonstrate that the lateral vestibular nucleus (LVN) exhibits ipsilateral projections to both fusiform and deep layers of the rat DCN. Stimulating the LVN induced glutamatergic synaptic currents in fusiform cells and granule cell interneurones. We combined the dextran amine neuronal tracing method with immunohistochemistry and showed that labeled projections from the LVN are co-labeled with VGLUT-2 by contrast to VGLUT-1. Wistar rats were exposed to a loud single tone (15 kHz, 110 dB SPL) for 6 hours. Five days after acoustic overexposure, the level of expression of VGLUT-1 in the DCN was decreased whereas the level of expression of VGLUT-2 in the DCN was increased including terminals originating from the LVN. VGLUT-2 mediated projections from the LVN to the DCN are likely to play a role in the head position in response to sound. Amplification of VGLUT-2 expression after acoustic overexposure could be a compensatory mechanism from vestibular inputs in response to hearing loss and to a decrease of VGLUT-1 expression from auditory nerve fibers.


Subject(s)
Acoustic Stimulation , Cell Surface Extensions/metabolism , Cochlear Nucleus/metabolism , Vesicular Glutamate Transport Protein 2/metabolism , Vestibular Nucleus, Lateral/metabolism , Animals , Cell Surface Extensions/genetics , Gene Expression , Gene Expression Regulation , Presynaptic Terminals/metabolism , Rats , Rats, Wistar , Synapses/genetics , Synapses/metabolism , Vesicular Glutamate Transport Protein 1/genetics , Vesicular Glutamate Transport Protein 1/metabolism , Vesicular Glutamate Transport Protein 2/genetics
19.
J Pharmacol Exp Ther ; 342(1): 41-52, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22454534

ABSTRACT

Morphine-induced signaling via opioid receptors (ORs) in dorsal root ganglia (DRG) neurons, the spinal cord, and various brain regions has been shown to modulate gene activity. Hitherto, little attention has been paid to extracellular signal-regulated kinases-1/2 (ERK-1/2)-mediated activation of the serum response factor (SRF) and ternary complex factors (TCFs) such as the E twenty six-like transcription factor-1 (ELK-1) in this context. Using TCF/SRF-dependent reporter gene constructs, a specific ERK-1/2 inhibitor and a dominant-negative ELK-1 mutant, we show herein that morphine activates ELK-1 via ERK-1/2 in DRG-derived F11 cells endogenously expressing µ and δ ORs. Previous studies with glioma cell lines such as NG108-15 cells attributed morphine-induced gene expression to the activation of the cAMP-responsive element binding protein (CREB). Thus, we also analyzed morphine-dependent activation of CREB in F11 and NG108-15 cells. In contrast to the CREB stimulation found in NG108-15 cells, we observed an inhibitory effect of morphine in F11 cells, indicating cell type-specific regulation of CREB by morphine. To obtain data about putative target genes of morphine-induced ELK-1/SRF activation, we analyzed mRNA levels of 15 ELK-1/SRF-dependent genes in cultured rat DRG neurons and F11 cells. We identified the early growth response protein-4 (EGR-4) as the strongest up-regulated gene in both cell types and observed ELK-1 activity-dependent activation of an EGR-4-driven reporter in F11 cells. Overall, we reveal an important role of ELK-1 for morphine-dependent gene induction in DRG-derived cells and propose that ELK-1 and EGR-4 contribute to the effects of morphine on neuronal plasticity.


Subject(s)
Ganglia, Spinal/drug effects , MAP Kinase Signaling System/drug effects , Morphine/pharmacology , Neurons/drug effects , Serum Response Factor/metabolism , ets-Domain Protein Elk-1/metabolism , Animals , Cells, Cultured , Cyclic AMP Response Element-Binding Protein/genetics , Cyclic AMP Response Element-Binding Protein/metabolism , Early Growth Response Transcription Factors/genetics , Early Growth Response Transcription Factors/metabolism , Ganglia, Spinal/metabolism , Mice , Neurons/metabolism , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Receptors, Opioid, delta/genetics , Receptors, Opioid, delta/metabolism , Receptors, Opioid, mu/genetics , Receptors, Opioid, mu/metabolism , Serum Response Factor/genetics , Signal Transduction/drug effects , Ternary Complex Factors/genetics , Ternary Complex Factors/metabolism , Transcription, Genetic/drug effects , Transcriptional Activation/drug effects , Up-Regulation/drug effects , ets-Domain Protein Elk-1/genetics
20.
Mol Cell Endocrinol ; 331(2): 232-40, 2011 Jan 15.
Article in English | MEDLINE | ID: mdl-20674667

ABSTRACT

The melanocortin-4 receptor (MC4R) is a prototypical G protein-coupled receptor (GPCR) that plays a considerable role in controlling appetite and energy homeostasis. Signalling initiated by MC4R is orchestrated by multiple agonists, inverse agonism and by interactions with accessory proteins. The exact molecular events translating MC4R signalling into its physiological role, however, are not fully understood. This review is an attempt to summarize new aspects of MC4R signalling in the context of its recently discovered alternative G protein coupling, and to give a perspective on how future research could improve our knowledge about the intertwining molecular mechanisms that are responsible for the regulation of energy homeostasis by the melanocortin system.


Subject(s)
GTP-Binding Proteins/agonists , GTP-Binding Proteins/metabolism , Receptor, Melanocortin, Type 4/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Homeostasis , Humans , Protein Binding , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL