Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Mov Disord ; 39(4): 733-738, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38357797

ABSTRACT

BACKGROUND: SAGE-324/BIIB124 is an investigational positive allosteric modulator of GABAA receptors. OBJECTIVE: KINETIC (NCT04305275), a double-blind, randomized, placebo-controlled, phase 2 study, evaluated SAGE-324/BIIB124 in individuals with essential tremor (ET). METHODS: Individuals aged 18 to 80 years were randomly assigned 1:1 to orally receive 60 mg of SAGE-324/BIIB124 or placebo once daily for 28 days. The primary endpoint was change from baseline in The Essential Tremor Rating Assessment Scale-Performance Subscale (TETRAS-PS) Item 4 (upper-limb tremor) at day 29 with SAGE-324/BIIB124 versus placebo. RESULTS: Between May 2020 and February 2021, 69 U.S. participants were randomly assigned to receive SAGE-324/BIIB124 (n = 34) or placebo (n = 35). There was a significant reduction from baseline in TETRAS-PS Item 4 at day 29 with SAGE-324/BIIB124 versus placebo (least squares mean [standard error]: -2.31 [0.401] vs. -1.24 [0.349], P = 0.0491). The most common treatment-emergent adverse events included somnolence, dizziness, fatigue, and balance disorder. CONCLUSION: These results support further development of SAGE-324/BIIB124 for potential ET treatment. © 2024 Sage Therapeutics, Inc and The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.


Subject(s)
Essential Tremor , Humans , Essential Tremor/drug therapy , Male , Middle Aged , Female , Aged , Double-Blind Method , Adult , Aged, 80 and over , Young Adult , Adolescent , Treatment Outcome
2.
Nat Nanotechnol ; 14(12): 1150-1159, 2019 12.
Article in English | MEDLINE | ID: mdl-31686009

ABSTRACT

Nanoparticle-mediated drug delivery is especially useful for targets within endosomes because of the endosomal transport mechanisms of many nanomedicines within cells. Here, we report the design of a pH-responsive, soft polymeric nanoparticle for the targeting of acidified endosomes to precisely inhibit endosomal signalling events leading to chronic pain. In chronic pain, the substance P (SP) neurokinin 1 receptor (NK1R) redistributes from the plasma membrane to acidified endosomes, where it signals to maintain pain. Therefore, the NK1R in endosomes provides an important target for pain relief. The pH-responsive nanoparticles enter cells by clathrin- and dynamin-dependent endocytosis and accumulate in NK1R-containing endosomes. Following intrathecal injection into rodents, the nanoparticles, containing the FDA-approved NK1R antagonist aprepitant, inhibit SP-induced activation of spinal neurons and thus prevent pain transmission. Treatment with the nanoparticles leads to complete and persistent relief from nociceptive, inflammatory and neuropathic nociception and offers a much-needed non-opioid treatment option for chronic pain.


Subject(s)
Aprepitant/administration & dosage , Chronic Pain/drug therapy , Delayed-Action Preparations/metabolism , Nanoparticles/metabolism , Neurokinin-1 Receptor Antagonists/administration & dosage , Animals , Aprepitant/pharmacokinetics , Aprepitant/therapeutic use , Cell Line , Chronic Pain/metabolism , Drug Delivery Systems , Endosomes/metabolism , HEK293 Cells , Humans , Hydrogen-Ion Concentration , Male , Mice, Inbred C57BL , Neurokinin-1 Receptor Antagonists/pharmacokinetics , Neurokinin-1 Receptor Antagonists/therapeutic use , Rats , Receptors, Neurokinin-1/metabolism
3.
JCI Insight ; 4(20)2019 10 17.
Article in English | MEDLINE | ID: mdl-31536477

ABSTRACT

Itch induces scratching that removes irritants from the skin, whereas pain initiates withdrawal or avoidance of tissue damage. While pain arises from both the skin and viscera, we investigated whether pruritogenic irritant mechanisms also function within visceral pathways. We show that subsets of colon-innervating sensory neurons in mice express, either individually or in combination, the pruritogenic receptors Tgr5 and the Mas-gene-related GPCRs Mrgpra3 and Mrgprc11. Agonists of these receptors activated subsets of colonic sensory neurons and evoked colonic afferent mechanical hypersensitivity via a TRPA1-dependent mechanism. In vivo intracolonic administration of individual TGR5, MrgprA3, or MrgprC11 agonists induced pronounced visceral hypersensitivity to colorectal distension. Coadministration of these agonists as an "itch cocktail" augmented hypersensitivity to colorectal distension and changed mouse behavior. These irritant mechanisms were maintained and enhanced in a model of chronic visceral hypersensitivity relevant to irritable bowel syndrome. Neurons from human dorsal root ganglia also expressed TGR5, as well as the human ortholog MrgprX1, and showed increased responsiveness to pruritogenic agonists in pathological states. These data support the existence of an irritant-sensing system in the colon that is a visceral representation of the itch pathways found in skin, thereby contributing to sensory disturbances accompanying common intestinal disorders.


Subject(s)
Abdominal Pain/physiopathology , Colon/innervation , Intestinal Mucosa/innervation , Irritable Bowel Syndrome/physiopathology , Sensory Receptor Cells/metabolism , Abdominal Pain/etiology , Adolescent , Adult , Animals , Colon/physiopathology , Disease Models, Animal , Female , Ganglia, Spinal/cytology , Healthy Volunteers , Humans , Intestinal Mucosa/physiopathology , Irritable Bowel Syndrome/chemically induced , Irritable Bowel Syndrome/complications , Irritable Bowel Syndrome/pathology , Male , Mice , Middle Aged , Nociception/physiology , Receptors, G-Protein-Coupled/metabolism , Trinitrobenzenesulfonic Acid/toxicity , Young Adult
4.
J Biol Chem ; 294(27): 10649-10662, 2019 07 05.
Article in English | MEDLINE | ID: mdl-31142616

ABSTRACT

Proteases sustain hyperexcitability and pain by cleaving protease-activated receptor-2 (PAR2) on nociceptors through distinct mechanisms. Whereas trypsin induces PAR2 coupling to Gαq, Gαs, and ß-arrestins, cathepsin-S (CS) and neutrophil elastase (NE) cleave PAR2 at distinct sites and activate it by biased mechanisms that induce coupling to Gαs, but not to Gαq or ß-arrestins. Because proteases activate PAR2 by irreversible cleavage, and activated PAR2 is degraded in lysosomes, sustained extracellular protease-mediated signaling requires mobilization of intact PAR2 from the Golgi apparatus or de novo synthesis of new receptors by incompletely understood mechanisms. We found here that trypsin, CS, and NE stimulate PAR2-dependent activation of protein kinase D (PKD) in the Golgi of HEK293 cells, in which PKD regulates protein trafficking. The proteases stimulated translocation of the PKD activator Gßγ to the Golgi, coinciding with PAR2 mobilization from the Golgi. Proteases also induced translocation of a photoconverted PAR2-Kaede fusion protein from the Golgi to the plasma membrane of KNRK cells. After incubation of HEK293 cells and dorsal root ganglia neurons with CS, NE, or trypsin, PAR2 responsiveness initially declined, consistent with PAR2 cleavage and desensitization, and then gradually recovered. Inhibitors of PKD, Gßγ, and protein translation inhibited recovery of PAR2 responsiveness. PKD and Gßγ inhibitors also attenuated protease-evoked mechanical allodynia in mice. We conclude that proteases that activate PAR2 by canonical and biased mechanisms stimulate PKD in the Golgi; PAR2 mobilization and de novo synthesis repopulate the cell surface with intact receptors and sustain nociceptive signaling by extracellular proteases.


Subject(s)
GTP-Binding Protein beta Subunits/metabolism , GTP-Binding Protein gamma Subunits/metabolism , Protein Kinase C/metabolism , Receptor, PAR-2/metabolism , Animals , Cathepsins/metabolism , Cell Membrane/metabolism , GTP-Binding Protein beta Subunits/antagonists & inhibitors , GTP-Binding Protein gamma Subunits/antagonists & inhibitors , Ganglia, Spinal/cytology , Ganglia, Spinal/metabolism , Golgi Apparatus/metabolism , HEK293 Cells , Humans , Hyperalgesia/metabolism , Hyperalgesia/pathology , Hyperalgesia/prevention & control , Leukocyte Elastase/metabolism , Mice , Mice, Inbred C57BL , Protein Kinase C/antagonists & inhibitors , Pyrimidines/administration & dosage , Pyrimidines/pharmacology , Receptor, PAR-2/agonists , Signal Transduction/drug effects , Xanthenes/administration & dosage , Xanthenes/pharmacology
5.
Proc Natl Acad Sci U S A ; 115(31): E7438-E7447, 2018 07 31.
Article in English | MEDLINE | ID: mdl-30012612

ABSTRACT

Once activated at the surface of cells, G protein-coupled receptors (GPCRs) redistribute to endosomes, where they can continue to signal. Whether GPCRs in endosomes generate signals that contribute to human disease is unknown. We evaluated endosomal signaling of protease-activated receptor-2 (PAR2), which has been proposed to mediate pain in patients with irritable bowel syndrome (IBS). Trypsin, elastase, and cathepsin S, which are activated in the colonic mucosa of patients with IBS and in experimental animals with colitis, caused persistent PAR2-dependent hyperexcitability of nociceptors, sensitization of colonic afferent neurons to mechanical stimuli, and somatic mechanical allodynia. Inhibitors of clathrin- and dynamin-dependent endocytosis and of mitogen-activated protein kinase kinase-1 prevented trypsin-induced hyperexcitability, sensitization, and allodynia. However, they did not affect elastase- or cathepsin S-induced hyperexcitability, sensitization, or allodynia. Trypsin stimulated endocytosis of PAR2, which signaled from endosomes to activate extracellular signal-regulated kinase. Elastase and cathepsin S did not stimulate endocytosis of PAR2, which signaled from the plasma membrane to activate adenylyl cyclase. Biopsies of colonic mucosa from IBS patients released proteases that induced persistent PAR2-dependent hyperexcitability of nociceptors, and PAR2 association with ß-arrestins, which mediate endocytosis. Conjugation to cholestanol promoted delivery and retention of antagonists in endosomes containing PAR2 A cholestanol-conjugated PAR2 antagonist prevented persistent trypsin- and IBS protease-induced hyperexcitability of nociceptors. The results reveal that PAR2 signaling from endosomes underlies the persistent hyperexcitability of nociceptors that mediates chronic pain of IBS. Endosomally targeted PAR2 antagonists are potential therapies for IBS pain. GPCRs in endosomes transmit signals that contribute to human diseases.


Subject(s)
Chronic Pain/etiology , Endosomes/physiology , Irritable Bowel Syndrome/physiopathology , Receptor, PAR-2/physiology , Signal Transduction/physiology , Animals , Endocytosis , Extracellular Signal-Regulated MAP Kinases/physiology , Humans , Nociception , Nociceptors/physiology , Trypsin/pharmacology
6.
Proc Natl Acad Sci U S A ; 114(46): 12309-12314, 2017 11 14.
Article in English | MEDLINE | ID: mdl-29087309

ABSTRACT

G protein-coupled receptors (GPCRs) are considered to function primarily at the plasma membrane, where they interact with extracellular ligands and couple to G proteins that transmit intracellular signals. Consequently, therapeutic drugs are designed to target GPCRs at the plasma membrane. Activated GPCRs undergo clathrin-dependent endocytosis. Whether GPCRs in endosomes control pathophysiological processes in vivo and are therapeutic targets remains uncertain. We investigated the contribution of endosomal signaling of the calcitonin receptor-like receptor (CLR) to pain transmission. Calcitonin gene-related peptide (CGRP) stimulated CLR endocytosis and activated protein kinase C (PKC) in the cytosol and extracellular signal regulated kinase (ERK) in the cytosol and nucleus. Inhibitors of clathrin and dynamin prevented CLR endocytosis and activation of cytosolic PKC and nuclear ERK, which derive from endosomal CLR. A cholestanol-conjugated antagonist, CGRP8-37, accumulated in CLR-containing endosomes and selectively inhibited CLR signaling in endosomes. CGRP caused sustained excitation of neurons in slices of rat spinal cord. Inhibitors of dynamin, ERK, and PKC suppressed persistent neuronal excitation. CGRP8-37-cholestanol, but not unconjugated CGRP8-37, prevented sustained neuronal excitation. When injected intrathecally to mice, CGRP8-37-cholestanol inhibited nociceptive responses to intraplantar injection of capsaicin, formalin, or complete Freund's adjuvant more effectively than unconjugated CGRP8-37 Our results show that CLR signals from endosomes to control pain transmission and identify CLR in endosomes as a therapeutic target for pain. Thus, GPCRs function not only at the plasma membrane but also in endosomes to control complex processes in vivo. Endosomal GPCRs are a drug target that deserve further attention.


Subject(s)
Calcitonin Receptor-Like Protein/genetics , Endocytosis/drug effects , Endosomes/metabolism , Nociception/physiology , Pain/physiopathology , Synaptic Transmission/drug effects , Adrenergic Antagonists/pharmacology , Animals , Calcitonin Gene-Related Peptide/pharmacology , Calcitonin Receptor-Like Protein/antagonists & inhibitors , Calcitonin Receptor-Like Protein/metabolism , Capsaicin/antagonists & inhibitors , Capsaicin/pharmacology , Cholestanols/pharmacology , Clathrin/antagonists & inhibitors , Clathrin/genetics , Clathrin/metabolism , Dynamins/genetics , Dynamins/metabolism , Endosomes/drug effects , Formaldehyde/antagonists & inhibitors , Formaldehyde/pharmacology , Freund's Adjuvant/antagonists & inhibitors , Freund's Adjuvant/pharmacology , Gene Expression Regulation , Injections, Spinal , Male , Mice , Microtomy , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Nociception/drug effects , Pain/chemically induced , Pain/genetics , Pain/prevention & control , Peptide Fragments/pharmacology , Protein Kinase C/genetics , Protein Kinase C/metabolism , Rats , Spinal Cord/cytology , Spinal Cord/drug effects , Spinal Cord/metabolism , Tissue Culture Techniques
7.
Sci Transl Med ; 9(392)2017 05 31.
Article in English | MEDLINE | ID: mdl-28566424

ABSTRACT

Typically considered to be cell surface sensors of extracellular signals, heterotrimeric GTP-binding protein (G protein)-coupled receptors (GPCRs) control many pathophysiological processes and are the target of 30% of therapeutic drugs. Activated receptors redistribute to endosomes, but researchers have yet to explore whether endosomal receptors generate signals that control complex processes in vivo and are viable therapeutic targets. We report that the substance P (SP) neurokinin 1 receptor (NK1R) signals from endosomes to induce sustained excitation of spinal neurons and pain transmission and that specific antagonism of the NK1R in endosomes with membrane-anchored drug conjugates provides more effective and sustained pain relief than conventional plasma membrane-targeted antagonists. Pharmacological and genetic disruption of clathrin, dynamin, and ß-arrestin blocked SP-induced NK1R endocytosis and prevented SP-stimulated activation of cytosolic protein kinase C and nuclear extracellular signal-regulated kinase, as well as transcription. Endocytosis inhibitors prevented sustained SP-induced excitation of neurons in spinal cord slices in vitro and attenuated nociception in vivo. When conjugated to cholestanol to promote endosomal targeting, NK1R antagonists selectively inhibited endosomal signaling and sustained neuronal excitation. Cholestanol conjugation amplified and prolonged the antinociceptive actions of NK1R antagonists. These results reveal a critical role for endosomal signaling of the NK1R in the complex pathophysiology of pain and demonstrate the use of endosomally targeted GPCR antagonists.


Subject(s)
Endosomes/metabolism , Molecular Targeted Therapy , Nociception , Pain/drug therapy , Receptors, Neurokinin-1/metabolism , Signal Transduction , Animals , Cell Compartmentation , Clathrin/metabolism , Dynamins/metabolism , Endocytosis/drug effects , Endosomes/drug effects , GTP-Binding Proteins/metabolism , HEK293 Cells , Humans , Lipids/chemistry , Models, Biological , Neurokinin-1 Receptor Antagonists/pharmacology , Neurokinin-1 Receptor Antagonists/therapeutic use , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Nociception/drug effects , Pain/pathology , Protein Binding/drug effects , Rats , Signal Transduction/drug effects , Spinal Cord/pathology , Subcellular Fractions/metabolism , Substance P/metabolism , beta-Arrestins/metabolism
8.
J Immunol ; 198(9): 3565-3575, 2017 05 01.
Article in English | MEDLINE | ID: mdl-28320832

ABSTRACT

G-CSF or CSF-3, originally defined as a regulator of granulocyte lineage development via its cell surface receptor (G-CSFR), can play a role in inflammation, and hence in many pathologies, due to its effects on mature lineage populations. Given this, and because pain is an extremely important arthritis symptom, the efficacy of an anti-G-CSFR mAb for arthritic pain and disease was compared with that of a neutrophil-depleting mAb, anti-Ly6G, in both adaptive and innate immune-mediated murine models. Pain and disease were ameliorated in Ag-induced arthritis, zymosan-induced arthritis, and methylated BSA/IL-1 arthritis by both prophylactic and therapeutic anti-G-CSFR mAb treatment, whereas only prophylactic anti-Ly6G mAb treatment was effective. Efficacy for pain and disease correlated with reduced joint neutrophil numbers and, importantly, benefits were noted without necessarily the concomitant reduction in circulating neutrophils. Anti-G-CSFR mAb also suppressed zymosan-induced inflammatory pain. A new G-CSF-driven (methylated BSA/G-CSF) arthritis model was established enabling us to demonstrate that pain was blocked by a cyclooxygenase-2 inhibitor, suggesting an indirect effect on neurons. Correspondingly, dorsal root ganglion neurons cultured in G-CSF failed to respond to G-CSF in vitro, and Csf3r gene expression could not be detected in dorsal root ganglion neurons by single-cell RT-PCR. These data suggest that G-CSFR/G-CSF targeting may be a safe therapeutic strategy for arthritis and other inflammatory conditions, particularly those in which pain is important, as well as for inflammatory pain per se.


Subject(s)
Antibodies, Blocking/therapeutic use , Arthritis, Experimental/therapy , Arthritis, Rheumatoid/therapy , Immunotherapy/methods , Neurons/drug effects , Neutrophils/immunology , Receptors, Granulocyte Colony-Stimulating Factor/metabolism , Animals , Antigens, Ly/immunology , Arthritis, Experimental/chemically induced , Arthritis, Experimental/immunology , Arthritis, Rheumatoid/immunology , Cells, Cultured , Disease Models, Animal , Granulocyte Colony-Stimulating Factor/metabolism , Humans , Leukocyte Reduction Procedures , Mice , Mice, Inbred C57BL , Neurons/physiology , Neutrophils/drug effects , Neutrophils/pathology , Pain Management , Receptors, Granulocyte Colony-Stimulating Factor/genetics , Receptors, Granulocyte Colony-Stimulating Factor/immunology
9.
Nutrients ; 8(10)2016 Oct 10.
Article in English | MEDLINE | ID: mdl-27735854

ABSTRACT

TRPA1 is a ligand-activated cation channel found in the intestine and other tissues. Components of food that stimulate TRPA1 receptors (phytonutrients) include allyl isothiocyanate, cinnamaldehyde and linalool, but these may also act at other receptors. Cells lining the intestinal mucosa are immunoreactive for TRPA1 and Trpa1 mRNA occurs in mucosal extracts, suggesting that the TRPA1 receptor is the target for these agonists. However, in situ hybridisation reveals Trpa1 expression in 5-HT containing enteroendocrine cells, not enterocytes. TRPA1 agonists evoke mucosal secretion, which may be indirect (through release of 5-HT) or direct by activation of enterocytes. We investigated effects of the phytonutrients on transmucosal ion currents in mouse duodenum and colon, and the specificity of the phytonutrients in cells transfected with Trpa1, and in Trpa1-deficient mice. The phytonutrients increased currents in the duodenum with the relative potencies: allyl isothiocyanate (AITC) > cinnamaldehyde > linalool (0.1 to 300 µM). The rank order was similar in the colon, but linalool was ineffective. Responses to AITC were reduced by the TRPA1 antagonist HC-030031 (100 µM), and were greatly diminished in Trpa1-/- duodenum and colon. Responses were not reduced by tetrodotoxin, 5-HT receptor antagonists, or atropine, but inhibition of prostaglandin synthesis reduced responses. Thus, functional TRPA1 channels are expressed by enterocytes of the duodenum and colon. Activation of enterocyte TRPA1 by food components has the potential to facilitate nutrient absorption.


Subject(s)
Intestinal Mucosa/physiology , Phytochemicals/pharmacology , Transient Receptor Potential Channels/drug effects , Acrolein/analogs & derivatives , Acrolein/pharmacology , Acyclic Monoterpenes , Animals , Calcium/metabolism , Colon/physiology , Duodenum/physiology , Electrophysiological Phenomena , Enterocytes/drug effects , Enterocytes/metabolism , Food , Gene Expression , HEK293 Cells , Humans , Intestinal Mucosa/chemistry , Isothiocyanates/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Monoterpenes/pharmacology , Serotonin 5-HT3 Receptor Antagonists/pharmacology , TRPA1 Cation Channel , Transfection , Transient Receptor Potential Channels/deficiency , Transient Receptor Potential Channels/genetics
10.
Am J Physiol Gastrointest Liver Physiol ; 311(3): G548-60, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27514475

ABSTRACT

Pancreatitis is an inflammatory disease of the pancreas characterized by dysregulated activity of digestive enzymes, necrosis, immune infiltration, and pain. Repeated incidence of pancreatitis is an important risk factor for pancreatic cancer. Legumain, a lysosomal cysteine protease, has been linked to inflammatory diseases such as atherosclerosis, stroke, and cancer. Until now, legumain activation has not been studied during pancreatitis. We used a fluorescently quenched activity-based probe to assess legumain activation during caerulein-induced pancreatitis in mice. We detected activated legumain by ex vivo imaging, confocal microscopy, and gel electrophoresis. Compared with healthy controls, legumain activity in the pancreas of caerulein-treated mice was increased in a time-dependent manner. Legumain was localized to CD68(+) macrophages and was not active in pancreatic acinar cells. Using a small-molecule inhibitor of legumain, we found that this protease is not essential for the initiation of pancreatitis. However, it may serve as a biomarker of disease, since patients with chronic pancreatitis show strongly increased legumain expression in macrophages. Moreover, the occurrence of legumain-expressing macrophages in regions of acinar-to-ductal metaplasia suggests that this protease may influence reprogramming events that lead to inflammation-induced pancreatic cancer.


Subject(s)
Cysteine Endopeptidases/metabolism , Macrophages/enzymology , Pancreatitis/enzymology , Animals , Ceruletide/toxicity , Cysteine Endopeptidases/genetics , Enzyme Inhibitors/pharmacology , Female , Gene Expression Regulation, Enzymologic , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Pancreatitis/chemically induced
11.
PLoS One ; 11(5): e0155526, 2016.
Article in English | MEDLINE | ID: mdl-27213574

ABSTRACT

The effect of respiratory tract viral infection on evoked cough in guinea pigs was evaluated. Guinea pigs were inoculated intranasally with either parainfluenza type 3 (PIV3) and cough was quantified in conscious animals. The guinea pigs infected with PIV3 (day 4) coughed nearly three times more than those treated with the viral growth medium in response to capsaicin, citric acid, and bradykinin. Since capsaicin, citric acid, and bradykinin evoked coughing in guinea pigs can be inhibited by drugs that antagonize the transient receptor potential cation channel, subfamily V, member 1 (TRPV1), it was reasoned that the virally-induced hypertussive state may involve alterations in TPRV1 activity. PIV3 infection caused a phenotypic switch in tracheal nodose Aδ "cough receptors" such that nearly 50% of neurons began to express, de novo, TRPV1 mRNA. There was also an increase TRPV1 expression in jugular C-fiber neurons as determined by qPCR. It has previously been reported that tracheal-specific nodose neurons express the BDNF receptor TrkB and jugular neurons express the NGF receptor TrkA. Jugular neurons also express the artemin receptor GFRα3. All these neurotrophic factors have been associated with increases in TRPV1 expression. In an ex vivo perfused guinea pig tracheal preparation, we demonstrated that within 8 h of PIV3 infusion there was no change in NGF mRNA expression, but there was nearly a 10-fold increase in BDNF mRNA in the tissue, and a small but significant elevation in the expression of artemin mRNA. In summary, PIV3 infection leads to elevations in TRPV1 expression in the two key cough evoking nerve subtypes in the guinea pig trachea, and this is associated with a hypertussive state with respect to various TRPV1 activating stimuli.


Subject(s)
Cough/virology , Parainfluenza Virus 3, Human/physiology , Paramyxoviridae Infections/complications , Respiratory Hypersensitivity/virology , Animals , Cough/genetics , Cough/immunology , Gene Expression , Guinea Pigs , Male , Nerve Fibers, Unmyelinated/physiology , Nerve Fibers, Unmyelinated/virology , Nerve Growth Factors/genetics , Nerve Growth Factors/metabolism , Nodose Ganglion/immunology , Nodose Ganglion/virology , Paramyxoviridae Infections/genetics , Paramyxoviridae Infections/immunology , Paramyxoviridae Infections/virology , Respiratory Hypersensitivity/complications , Respiratory Hypersensitivity/genetics , Respiratory Hypersensitivity/immunology , TRPV Cation Channels/genetics , TRPV Cation Channels/metabolism , Trachea/immunology , Trachea/innervation , Trachea/metabolism , Trachea/virology
12.
J Biol Chem ; 291(21): 11285-99, 2016 May 20.
Article in English | MEDLINE | ID: mdl-27030010

ABSTRACT

Agonist-evoked endocytosis of G protein-coupled receptors has been extensively studied. The mechanisms by which agonists stimulate mobilization and plasma membrane translocation of G protein-coupled receptors from intracellular stores are unexplored. Protease-activated receptor-2 (PAR2) traffics to lysosomes, and sustained protease signaling requires mobilization and plasma membrane trafficking of PAR2 from Golgi stores. We evaluated the contribution of protein kinase D (PKD) and Gßγ to this process. In HEK293 and KNRK cells, the PAR2 agonists trypsin and 2-furoyl-LIGRLO-NH2 activated PKD in the Golgi apparatus, where PKD regulates protein trafficking. PAR2 activation induced translocation of Gßγ, a PKD activator, to the Golgi apparatus, determined by bioluminescence resonance energy transfer between Gγ-Venus and giantin-Rluc8. Inhibitors of PKD (CRT0066101) and Gßγ (gallein) prevented PAR2-stimulated activation of PKD. CRT0066101, PKD1 siRNA, and gallein all inhibited recovery of PAR2-evoked Ca(2+) signaling. PAR2 with a photoconvertible Kaede tag was expressed in KNRK cells to examine receptor translocation from the Golgi apparatus to the plasma membrane. Irradiation of the Golgi region (405 nm) induced green-red photo-conversion of PAR2-Kaede. Trypsin depleted PAR2-Kaede from the Golgi apparatus and repleted PAR2-Kaede at the plasma membrane. CRT0066101 inhibited PAR2-Kaede translocation to the plasma membrane. CRT0066101 also inhibited sustained protease signaling to colonocytes and nociceptive neurons that naturally express PAR2 and mediate protease-evoked inflammation and nociception. Our results reveal a major role for PKD and Gßγ in agonist-evoked mobilization of intracellular PAR2 stores that is required for sustained signaling by extracellular proteases.


Subject(s)
GTP-Binding Protein beta Subunits/metabolism , GTP-Binding Protein gamma Subunits/metabolism , Protein Kinase C/metabolism , Receptor, PAR-2/agonists , Receptor, PAR-2/metabolism , Animals , Biological Transport, Active/drug effects , Calcium Signaling , Cell Line , Cell Membrane/metabolism , Endosomes/metabolism , GTP-Binding Protein beta Subunits/antagonists & inhibitors , GTP-Binding Protein gamma Subunits/antagonists & inhibitors , Golgi Apparatus/metabolism , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Protein Kinase C/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , Rats , Xanthenes/pharmacology
13.
Hepatology ; 63(4): 1287-98, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26426865

ABSTRACT

UNLABELLED: A challenge in obstetrics is to distinguish pathological symptoms from those associated with normal changes of pregnancy, typified by the need to differentiate whether gestational pruritus of the skin is an early symptom of intrahepatic cholestasis of pregnancy (ICP) or due to benign pruritus gravidarum. ICP is characterized by raised serum bile acids and complicated by spontaneous preterm labor and stillbirth. A biomarker for ICP would be invaluable for early diagnosis and treatment and to enable its differentiation from other maternal diseases. Three progesterone sulfate compounds, whose concentrations have not previously been studied, were newly synthesized and assayed in the serum of three groups of ICP patients and found to be significantly higher in ICP at 9-15 weeks of gestation and prior to symptom onset (group 1 cases/samples: ICP n = 35/80, uncomplicated pregnancy = 29/100), demonstrating that all three progesterone sulfates are prognostic for ICP. Concentrations of progesterone sulfates were associated with itch severity and, in combination with autotaxin, distinguished pregnant women with itch that would subsequently develop ICP from pruritus gravidarum (group 2: ICP n = 41, pruritus gravidarum n = 14). In a third group of first-trimester samples all progesterone sulfates were significantly elevated in serum from low-risk asymptomatic women who subsequently developed ICP (ICP/uncomplicated pregnancy n = 54/51). Finally, we show mechanistically that progesterone sulfates mediate itch by evoking a Tgr5-dependent scratch response in mice. CONCLUSION: Our discovery that sulfated progesterone metabolites are a prognostic indicator for ICP will help predict onset of ICP and distinguish it from benign pruritus gravidarum, enabling targeted obstetric care to a high-risk population. Delineation of a progesterone sulfate-TGR5 pruritus axis identifies a therapeutic target for itch management in ICP.


Subject(s)
Bile Acids and Salts/blood , Cholestasis, Intrahepatic/diagnosis , Pregnancy Complications/diagnosis , Pregnancy Outcome , Pregnancy, Animal , Progesterone/metabolism , Pruritus/diagnosis , Adult , Animals , Behavior, Animal , Case-Control Studies , Cholestasis, Intrahepatic/blood , Chromatography, High Pressure Liquid/methods , Female , Humans , Odds Ratio , Predictive Value of Tests , Pregnancy , Pregnancy Complications/blood , Pruritus/metabolism , ROC Curve , Severity of Illness Index , Tandem Mass Spectrometry/methods , United Kingdom
14.
Neuron ; 88(4): 635-49, 2015 Nov 18.
Article in English | MEDLINE | ID: mdl-26590341

ABSTRACT

G protein-coupled receptors (GPCRs) are the major class of sensory proteins and a primary therapeutic target in the pathways to pain and itch. GPCRs are complex signaling machines. Their association with ligands, other receptors, and signaling and regulatory partners induces GPCRs to adopt distinct conformations and to traffic to microdomains within plasma and endosomal membranes. This conformational and positional dynamism controls GPCR signaling in time and space and defines the outcome of receptor activation. An understanding of the dynamic nature of GPCRs within primary sensory neurons and neighboring cells brings new insights into their contributions to the physiology and pathophysiology of pain and itch and provides novel opportunities for therapeutic intervention.


Subject(s)
Nociceptors/metabolism , Pain/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Humans , Nociceptors/physiology , Pruritus/metabolism , Receptors, G-Protein-Coupled/physiology , Sensory Receptor Cells/metabolism , Sensory Receptor Cells/physiology , Signal Transduction
15.
Am J Physiol Gastrointest Liver Physiol ; 309(4): G248-59, 2015 Aug 15.
Article in English | MEDLINE | ID: mdl-26138465

ABSTRACT

Activated G protein-coupled receptors traffic to endosomes and are sorted to recycling or degradative pathways. Endosomes are also a site of receptor signaling of sustained and pathophysiologically important processes, including inflammation. However, the mechanisms of endosomal sorting of receptors and the impact of disease on trafficking have not been fully defined. We examined the effects of inflammation on the subcellular distribution and trafficking of the substance P (SP) neurokinin 1 receptor (NK1R) in enteric neurons. We studied NK1R trafficking in enteric neurons of the mouse colon using immunofluorescence and confocal microscopy. The impact of inflammation was studied in IL10(-/-)-piroxicam and trinitrobenzenesulfonic acid colitis models. NK1R was localized to the plasma membrane of myenteric and submucosal neurons of the uninflamed colon. SP evoked NK1R endocytosis and recycling. Deletion of ß-arrestin2, which associates with the activated NK1R, accelerated recycling. Inhibition of endothelin-converting enzyme-1 (ECE-1), which degrades endosomal SP, prevented recycling. Inflammation was associated with NK1R endocytosis in myenteric but not submucosal neurons. Whereas the NK1R in uninflamed neurons recycled within 60 min, NK1R recycling in inflamed neurons was delayed for >120 min, suggesting defective recycling machinery. Inflammation was associated with ß-arrestin2 upregulation and ECE-1 downregulation, which may contribute to the defective NK1R recycling. We conclude that inflammation evokes redistribution of NK1R from the plasma membrane to endosomes of myenteric neurons through enhanced SP release and defective NK1R recycling. Defective recycling may be secondary to upregulation of ß-arrestin2 and downregulation of ECE-1. Internalized NK1R may generate sustained proinflammatory signals that disrupt normal neuronal functions.


Subject(s)
Colitis, Ulcerative/metabolism , Endosomes/metabolism , Enteric Nervous System/metabolism , Receptors, Neurokinin-1/metabolism , Animals , Arrestins/genetics , Arrestins/metabolism , Aspartic Acid Endopeptidases/genetics , Aspartic Acid Endopeptidases/metabolism , Endocytosis , Endothelin-Converting Enzymes , Inflammation/metabolism , Metalloendopeptidases/genetics , Metalloendopeptidases/metabolism , Mice , Mice, Inbred C57BL , Protein Transport , beta-Arrestins
16.
J Biol Chem ; 290(22): 13875-87, 2015 May 29.
Article in English | MEDLINE | ID: mdl-25878251

ABSTRACT

Proteases that cleave protease-activated receptor-2 (PAR(2)) at Arg(36)↓Ser(37) reveal a tethered ligand that binds to the cleaved receptor. PAR(2) activates transient receptor potential (TRP) channels of nociceptive neurons to induce neurogenic inflammation and pain. Although proteases that cleave PAR(2) at non-canonical sites can trigger distinct signaling cascades, the functional importance of the PAR(2)-biased agonism is uncertain. We investigated whether neutrophil elastase, a biased agonist of PAR(2), causes inflammation and pain by activating PAR2 and TRP vanilloid 4 (TRPV4). Elastase cleaved human PAR(2) at Ala(66)↓Ser(67) and Ser(67)↓Val(68). Elastase stimulated PAR(2)-dependent cAMP accumulation and ERK1/2 activation, but not Ca(2+) mobilization, in KNRK cells. Elastase induced PAR(2) coupling to Gαs but not Gαq in HEK293 cells. Although elastase did not promote recruitment of G protein-coupled receptor kinase-2 (GRK(2)) or ß-arrestin to PAR(2), consistent with its inability to promote receptor endocytosis, elastase did stimulate GRK6 recruitment. Elastase caused PAR(2)-dependent sensitization of TRPV4 currents in Xenopus laevis oocytes by adenylyl cyclase- and protein kinase A (PKA)-dependent mechanisms. Elastase stimulated PAR(2)-dependent cAMP formation and ERK1/2 phosphorylation, and a PAR(2)- and TRPV4-mediated influx of extracellular Ca(2+) in mouse nociceptors. Adenylyl cyclase and PKA-mediated elastase-induced activation of TRPV4 and hyperexcitability of nociceptors. Intraplantar injection of elastase to mice caused edema and mechanical hyperalgesia by PAR(2)- and TRPV4-mediated mechanisms. Thus, the elastase-biased agonism of PAR(2) causes Gαs-dependent activation of adenylyl cyclase and PKA, which activates TRPV4 and sensitizes nociceptors to cause inflammation and pain. Our results identify a novel mechanism of elastase-induced activation of TRPV4 and expand the role of PAR(2) as a mediator of protease-driven inflammation and pain.


Subject(s)
Inflammation/metabolism , Leukocyte Elastase/metabolism , Pain/metabolism , Receptor, PAR-2/metabolism , TRPV Cation Channels/metabolism , Animals , Calcium/metabolism , Cell Membrane/metabolism , Cyclic AMP/metabolism , Edema/metabolism , Edema/pathology , GTP-Binding Proteins/metabolism , Ganglia, Spinal/metabolism , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Neurons/metabolism , Nociception , Oocytes/cytology , Oocytes/metabolism , Patch-Clamp Techniques , Peptide Hydrolases/metabolism , Protein Structure, Tertiary , Signal Transduction , Xenopus laevis/metabolism
17.
Gastroenterology ; 147(6): 1417-28, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25194674

ABSTRACT

BACKGROUND & AIMS: Patients with cholestatic disease have increased systemic concentrations of bile acids (BAs) and profound pruritus. The G-protein-coupled BA receptor 1 TGR5 (encoded by GPBAR1) is expressed by primary sensory neurons; its activation induces neuronal hyperexcitability and scratching by unknown mechanisms. We investigated whether the transient receptor potential ankyrin 1 (TRPA1) is involved in BA-evoked, TGR5-dependent pruritus in mice. METHODS: Co-expression of TGR5 and TRPA1 in cutaneous afferent neurons isolated from mice was analyzed by immunofluorescence, in situ hybridization, and single-cell polymerase chain reaction. TGR5-induced activation of TRPA1 was studied in in HEK293 cells, Xenopus laevis oocytes, and primary sensory neurons by measuring Ca(2+) signals. The contribution of TRPA1 to TGR5-induced release of pruritogenic neuropeptides, activation of spinal neurons, and scratching behavior were studied using TRPA1 antagonists or Trpa1(-/-) mice. RESULTS: TGR5 and TRPA1 protein and messenger RNA were expressed by cutaneous afferent neurons. In HEK cells, oocytes, and neurons co-expressing TGR5 and TRPA1, BAs caused TGR5-dependent activation and sensitization of TRPA1 by mechanisms that required Gßγ, protein kinase C, and Ca(2+). Antagonists or deletion of TRPA1 prevented BA-stimulated release of the pruritogenic neuropeptides gastrin-releasing peptide and atrial natriuretic peptide B in the spinal cord. Disruption of Trpa1 in mice blocked BA-induced expression of Fos in spinal neurons and prevented BA-stimulated scratching. Spontaneous scratching was exacerbated in transgenic mice that overexpressed TRG5. Administration of a TRPA1 antagonist or the BA sequestrant colestipol, which lowered circulating levels of BAs, prevented exacerbated spontaneous scratching in TGR5 overexpressing mice. CONCLUSIONS: BAs induce pruritus in mice by co-activation of TGR5 and TRPA1. Antagonists of TGR5 and TRPA1, or inhibitors of the signaling mechanism by which TGR5 activates TRPA1, might be developed for treatment of cholestatic pruritus.


Subject(s)
Bile Acids and Salts/metabolism , Cholestasis/metabolism , Pruritus/metabolism , Receptors, G-Protein-Coupled/metabolism , Transient Receptor Potential Channels/metabolism , Animals , Cholestasis/complications , Disease Models, Animal , Ganglia, Spinal/cytology , Ganglia, Spinal/metabolism , Gastrin-Releasing Peptide/metabolism , HEK293 Cells , Humans , Mice, Knockout , Natriuretic Peptides/metabolism , Neurons, Afferent/cytology , Neurons, Afferent/metabolism , Nociceptors/metabolism , Oocytes/cytology , Oocytes/metabolism , Primary Cell Culture , Pruritus/etiology , Receptors, G-Protein-Coupled/genetics , TRPA1 Cation Channel , Transient Receptor Potential Channels/genetics , Xenopus laevis
18.
J Biol Chem ; 289(39): 27215-27234, 2014 Sep 26.
Article in English | MEDLINE | ID: mdl-25118282

ABSTRACT

Serine proteases such as trypsin and mast cell tryptase cleave protease-activated receptor-2 (PAR2) at R(36)↓S(37) and reveal a tethered ligand that excites nociceptors, causing neurogenic inflammation and pain. Whether proteases that cleave PAR2 at distinct sites are biased agonists that also induce inflammation and pain is unexplored. Cathepsin S (Cat-S) is a lysosomal cysteine protease of antigen-presenting cells that is secreted during inflammation and which retains activity at extracellular pH. We observed that Cat-S cleaved PAR2 at E(56)↓T(57), which removed the canonical tethered ligand and prevented trypsin activation. In HEK and KNRK cell lines and in nociceptive neurons of mouse dorsal root ganglia, Cat-S and a decapeptide mimicking the Cat-S-revealed tethered ligand-stimulated PAR2 coupling to Gαs and formation of cAMP. In contrast to trypsin, Cat-S did not mobilize intracellular Ca(2+), activate ERK1/2, recruit ß-arrestins, or induce PAR2 endocytosis. Cat-S caused PAR2-dependent activation of transient receptor potential vanilloid 4 (TRPV4) in Xenopus laevis oocytes, HEK cells and nociceptive neurons, and stimulated neuronal hyperexcitability by adenylyl cyclase and protein kinase A-dependent mechanisms. Intraplantar injection of Cat-S caused inflammation and hyperalgesia in mice that was attenuated by PAR2 or TRPV4 deletion and adenylyl cyclase inhibition. Cat-S and PAR2 antagonists suppressed formalin-induced inflammation and pain, which implicates endogenous Cat-S and PAR2 in inflammatory pain. Our results identify Cat-S as a biased agonist of PAR2 that causes PAR2- and TRPV4-dependent inflammation and pain. They expand the role of PAR2 as a mediator of protease-driven inflammatory pain.


Subject(s)
Cathepsins/metabolism , Pain , Receptor, PAR-2 , TRPV Cation Channels , Adenylyl Cyclases/genetics , Adenylyl Cyclases/metabolism , Animals , Cathepsins/genetics , HEK293 Cells , Humans , Hyperalgesia/genetics , Hyperalgesia/metabolism , Hyperalgesia/pathology , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Mice , Mice, Knockout , Pain/genetics , Pain/metabolism , Pain/pathology , Receptor, PAR-2/agonists , Receptor, PAR-2/genetics , Receptor, PAR-2/metabolism , TRPV Cation Channels/agonists , TRPV Cation Channels/genetics , TRPV Cation Channels/metabolism , Xenopus laevis
19.
J Biol Chem ; 289(29): 20283-94, 2014 Jul 18.
Article in English | MEDLINE | ID: mdl-24898255

ABSTRACT

Although the intracellular trafficking of G protein-coupled receptors controls specific signaling events, it is unclear how the spatiotemporal control of signaling contributes to complex pathophysiological processes such as inflammation. By using bioluminescence resonance energy transfer and superresolution microscopy, we found that substance P (SP) induces the association of the neurokinin 1 receptor (NK1R) with two classes of proteins that regulate SP signaling from plasma and endosomal membranes: the scaffolding proteins ß-arrestin (ßARRs) 1 and 2 and the transmembrane metallopeptidases ECE-1c and ECE-1d. In HEK293 cells and non-transformed human colonocytes, we observed that G protein-coupled receptor kinase 2 and ßARR1/2 terminate plasma membrane Ca(2+) signaling and initiate receptor trafficking to endosomes that is necessary for sustained activation of ERKs in the nucleus. ßARRs deliver the SP-NK1R endosomes, where ECE-1 associates with the complex, degrades SP, and allows the NK1R, freed from ßARRs, to recycle. Thus, both ECE-1 and ßARRs mediate the resensitization of NK1R Ca(2+) signaling at the plasma membrane. Sustained exposure of colonocytes to SP activates NF-κB and stimulates IL-8 secretion. This proinflammatory signaling is unaffected by inhibition of the endosomal ERK pathway but is suppressed by ECE-1 inhibition or ßARR2 knockdown. Inhibition of protein phosphatase 2A, which also contributes to sustained NK1R signaling at the plasma membrane, similarly attenuates IL-8 secretion. Thus, the primary function of ßARRs and ECE-1 in SP-dependent inflammatory signaling is to promote resensitization, which allows the sustained NK1R signaling from the plasma membrane that drives inflammation.


Subject(s)
Arrestins/metabolism , Aspartic Acid Endopeptidases/metabolism , Metalloendopeptidases/metabolism , Receptors, Neurokinin-1/metabolism , Substance P/metabolism , Arrestins/antagonists & inhibitors , Arrestins/genetics , Aspartic Acid Endopeptidases/genetics , Cell Line , Cell Membrane/metabolism , Endosomes/metabolism , Endothelin-Converting Enzymes , Fluorescence Resonance Energy Transfer , Gene Knockdown Techniques , HEK293 Cells , Humans , Inflammation Mediators/metabolism , MAP Kinase Signaling System , Metalloendopeptidases/genetics , RNA, Small Interfering/genetics , Receptors, G-Protein-Coupled/metabolism , Receptors, Neurokinin-1/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction , beta-Arrestins
20.
Am J Physiol Gastrointest Liver Physiol ; 306(8): G686-98, 2014 Apr 15.
Article in English | MEDLINE | ID: mdl-24578341

ABSTRACT

Lymphatic fluid is a plasma filtrate that can be viewed as having biological activity through the passive accumulation of molecules from the interstitial fluid. The possibility that lymphatic fluid is part of an active self-contained signaling process that parallels the endocrine system, through the activation of G-protein coupled receptors (GPCR), has remained unexplored. We show that the GPCR lysophosphatidic acid 5 (LPA5) is found in sensory nerve fibers expressing calcitonin gene-related peptide (CGRP) that innervate the lumen of lymphatic lacteals and enteric nerves. Using LPA5 as a model for nutrient-responsive GPCRs present on sensory nerves, we demonstrate that dietary protein hydrolysate (peptone) can induce c-Fos expression in enterocytes and nerves that express LPA5. Mesenteric lymphatic fluid (MLF) mobilizes intracellular calcium in cell models expressing LPA5 upon feeding in a time- and dose-dependent manner. Primary cultured neurons of the dorsal root ganglia expressing CGRP are activated by MLF, which is enhanced upon LPA5 overexpression. Activation is independent of the known LPA5 agonists, lysophosphatidic acid and farnesyl pyrophosphate. These data bring forth a pathway for the direct stimulation of sensory nerves by luminal contents and interstitial fluid. Thus, by activating LPA5 on sensory nerves, MLF provides a means for known and yet to be identified constituents of the interstitial fluid to act as signals to comprise a "neurolymphocrine" system.


Subject(s)
Enterocytes/physiology , Lymph/physiology , Receptors, Lysophosphatidic Acid/metabolism , Sensory Receptor Cells/physiology , Animals , Calcitonin Gene-Related Peptide/metabolism , Cell Physiological Phenomena , Dietary Proteins/metabolism , Mice , Mice, Inbred C57BL , Peptones/metabolism , Rats , Rats, Sprague-Dawley , Receptors, G-Protein-Coupled/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...