Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
J Biol Chem ; 288(8): 5790-802, 2013 Feb 22.
Article in English | MEDLINE | ID: mdl-23288842

ABSTRACT

G protein-coupled receptors of nociceptive neurons can sensitize transient receptor potential (TRP) ion channels, which amplify neurogenic inflammation and pain. Protease-activated receptor 2 (PAR(2)), a receptor for inflammatory proteases, is a major mediator of neurogenic inflammation and pain. We investigated the signaling mechanisms by which PAR(2) regulates TRPV4 and determined the importance of tyrosine phosphorylation in this process. Human TRPV4 was expressed in HEK293 cells under control of a tetracycline-inducible promoter, allowing controlled and graded channel expression. In cells lacking TRPV4, the PAR(2) agonist stimulated a transient increase in [Ca(2+)](i). TRPV4 expression led to a markedly sustained increase in [Ca(2+)](i). Removal of extracellular Ca(2+) and treatment with the TRPV4 antagonists Ruthenium Red or HC067047 prevented the sustained response. Inhibitors of phospholipase A(2) and cytochrome P450 epoxygenase attenuated the sustained response, suggesting that PAR(2) generates arachidonic acid-derived lipid mediators, such as 5',6'-EET, that activate TRPV4. Src inhibitor 1 suppressed PAR(2)-induced activation of TRPV4, indicating the importance of tyrosine phosphorylation. The TRPV4 tyrosine mutants Y110F, Y805F, and Y110F/Y805F were expressed normally at the cell surface. However, PAR(2) was unable to activate TRPV4 with the Y110F mutation. TRPV4 antagonism suppressed PAR(2) signaling to primary nociceptive neurons, and TRPV4 deletion attenuated PAR(2)-stimulated neurogenic inflammation. Thus, PAR(2) activation generates a signal that induces sustained activation of TRPV4, which requires a key tyrosine residue (TRPV4-Tyr-110). This mechanism partly mediates the proinflammatory actions of PAR(2).


Subject(s)
Receptor, PAR-2/metabolism , TRPV Cation Channels/metabolism , Animals , Calcium/metabolism , Cytochrome P-450 CYP2J2 , Cytochrome P-450 Enzyme Inhibitors , Cytochrome P-450 Enzyme System , HEK293 Cells , Humans , Inflammation , Male , Mice , Models, Biological , Mutagenesis, Site-Directed , Pain , Phospholipase A2 Inhibitors , Phosphorylation , Rats , Rats, Sprague-Dawley , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Tyrosine/chemistry , Tyrosine/metabolism
2.
Circulation ; 125(14): 1774-86, S1-19, 2012 Apr 10.
Article in English | MEDLINE | ID: mdl-22392530

ABSTRACT

BACKGROUND: Pain triggers a homeostatic alarm reaction to injury. It remains unknown, however, whether nociceptive signaling activated by ischemia is relevant for progenitor cells (PC) release from bone marrow. To this end, we investigated the role of the neuropeptide substance P (SP) and cognate neurokinin 1 (NK1) nociceptor in PC activation and angiogenesis during ischemia in mice and in human subjects. METHODS AND RESULTS: The mouse bone marrow contains sensory fibers and PC that express SP. Moreover, SP-induced migration provides enrichment for PC that express NK1 and promote reparative angiogenesis after transplantation in a mouse model of limb ischemia. Acute myocardial infarction and limb ischemia increase SP levels in peripheral blood, decrease SP levels in bone marrow, and stimulate the mobilization of NK1-expressing PC, with these effects being abrogated by systemic administration of the opioid receptor agonist morphine. Moreover, bone marrow reconstitution with NK1-knockout cells results in depressed PC mobilization, delayed blood flow recovery, and reduced neovascularization after ischemia. We next asked whether SP is instrumental to PC mobilization and homing in patients with ischemia. Human PC express NK1, and SP-induced migration provides enrichment for proangiogenic PC. Patients with acute myocardial infarction show high circulating levels of SP and NK1-positive cells that coexpress PC antigens, such as CD34, KDR, and CXCR4. Moreover, NK1-expressing PC are abundant in infarcted hearts but not in hearts that developed an infarct after transplantation. CONCLUSIONS: Our data highlight the role of SP in reparative neovascularization. Nociceptive signaling may represent a novel target of regenerative medicine.


Subject(s)
Ischemia/physiopathology , Neovascularization, Physiologic , Nociception/physiology , Signal Transduction/physiology , Stem Cells/physiology , Substance P/physiology , Animals , Hematopoietic Stem Cell Mobilization , Humans , Male , Mice , Mice, Inbred C57BL , Receptors, Calcitonin Gene-Related Peptide/physiology , Receptors, Neurokinin-1/physiology
3.
Pharmacol Ther ; 129(1): 62-81, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20965213

ABSTRACT

Regenerative cardiovascular medicine is the frontline of 21st-century health care. Cell therapy trials using bone marrow progenitor cells documented that the approach is feasible, safe and potentially beneficial in patients with ischemic disease. However, cardiovascular prevention and rehabilitation strategies should aim to conserve the pristine healing capacity of a healthy organism as well as reactivate it under disease conditions. This requires an increased understanding of stem cell microenvironment and trafficking mechanisms. Engagement and disengagement of stem cells of the osteoblastic niche is a dynamic process, finely tuned to allow low amounts of cells move out of the bone marrow and into the circulation on a regular basis. The balance is altered under stress situations, like tissue injury or ischemia, leading to remarkably increased cell egression. Individual populations of circulating progenitor cells could give rise to mature tissue cells (e.g. endothelial cells or cardiomyocytes), while the majority may differentiate to leukocytes, affecting the environment of homing sites in a paracrine way, e.g. promoting endothelial survival, proliferation and function, as well as attenuating or enhancing inflammation. This review focuses on the dynamics of the stem cell niche in healthy and disease conditions and on therapeutic means to direct stem cell/progenitor cell mobilization and recruitment into improved tissue repair.


Subject(s)
Cardiovascular Diseases/therapy , Hematopoietic Stem Cells/physiology , Stem Cell Niche/physiology , Stem Cells/physiology , Animals , Cell Differentiation , Cell Movement , Hematopoietic Stem Cell Mobilization , Humans , Regeneration , Signal Transduction
4.
Am J Physiol Gastrointest Liver Physiol ; 299(3): G556-71, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20539005

ABSTRACT

The mechanisms of pancreatic pain, a cardinal symptom of pancreatitis, are unknown. Proinflammatory agents that activate transient receptor potential (TRP) channels in nociceptive neurons can cause neurogenic inflammation and pain. We report a major role for TRPV4, which detects osmotic pressure and arachidonic acid metabolites, and TRPA1, which responds to 4-hydroxynonenal and cyclopentenone prostaglandins, in pancreatic inflammation and pain in mice. Immunoreactive TRPV4 and TRPA1 were detected in pancreatic nerve fibers and in dorsal root ganglia neurons innervating the pancreas, which were identified by retrograde tracing. Agonists of TRPV4 and TRPA1 increased intracellular Ca(2+) concentration ([Ca(2+)](i)) in these neurons in culture, and neurons also responded to the TRPV1 agonist capsaicin and are thus nociceptors. Intraductal injection of TRPV4 and TRPA1 agonists increased c-Fos expression in spinal neurons, indicative of nociceptor activation, and intraductal TRPA1 agonists also caused pancreatic inflammation. The effects of TRPV4 and TRPA1 agonists on [Ca(2+)](i), pain and inflammation were markedly diminished or abolished in trpv4 and trpa1 knockout mice. The secretagogue cerulein induced pancreatitis, c-Fos expression in spinal neurons, and pain behavior in wild-type mice. Deletion of trpv4 or trpa1 suppressed c-Fos expression and pain behavior, and deletion of trpa1 attenuated pancreatitis. Thus TRPV4 and TRPA1 contribute to pancreatic pain, and TRPA1 also mediates pancreatic inflammation. Our results provide new information about the contributions of TRPV4 and TRPA1 to inflammatory pain and suggest that channel antagonists are an effective therapy for pancreatitis, when multiple proinflammatory agents are generated that can activate and sensitize these channels.


Subject(s)
Pain/metabolism , Pancreatitis/complications , TRPV Cation Channels/metabolism , Transient Receptor Potential Channels/metabolism , Aldehydes/toxicity , Animals , Cysteine Proteinase Inhibitors/toxicity , Female , Ganglia, Spinal/physiology , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Irritants/toxicity , Male , Mice , Mice, Knockout , Mustard Plant/toxicity , Nociceptors/physiology , Pain/etiology , Pancreas/drug effects , Pancreas/innervation , Pancreas/pathology , Pancreatitis/chemically induced , Pancreatitis/metabolism , Plant Oils/toxicity , Spinal Cord/metabolism , TRPA1 Cation Channel , TRPV Cation Channels/agonists , TRPV Cation Channels/genetics , Transient Receptor Potential Channels/agonists , Transient Receptor Potential Channels/genetics
5.
J Comp Neurol ; 516(2): 141-56, 2009 Sep 10.
Article in English | MEDLINE | ID: mdl-19575452

ABSTRACT

Serine proteases generated during injury and inflammation cleave protease-activated receptor 2 (PAR(2)) on primary sensory neurons to induce neurogenic inflammation and hyperalgesia. Hyperalgesia requires sensitization of transient receptor potential vanilloid (TRPV) ion channels by mechanisms involving phospholipase C and protein kinase C (PKC). The protein kinase D (PKD) serine/threonine kinases are activated by diacylglycerol and PKCs and can phosphorylate TRPV1. Thus, PKDs may participate in novel signal transduction pathways triggered by serine proteases during inflammation and pain. However, it is not known whether PAR(2) activates PKD, and the expression of PKD isoforms by nociceptive neurons is poorly characterized. By using HEK293 cells transfected with PKDs, we found that PAR(2) stimulation promoted plasma membrane translocation and phosphorylation of PKD1, PKD2, and PKD3, indicating activation. This effect was partially dependent on PKCepsilon. By immunofluorescence and confocal microscopy, with antibodies against PKD1/PKD2 and PKD3 and neuronal markers, we found that PKDs were expressed in rat and mouse dorsal root ganglia (DRG) neurons, including nociceptive neurons that expressed TRPV1, PAR(2), and neuropeptides. PAR(2) agonist induced phosphorylation of PKD in cultured DRG neurons, indicating PKD activation. Intraplantar injection of PAR(2) agonist also caused phosphorylation of PKD in neurons of lumbar DRG, confirming activation in vivo. Thus, PKD1, PKD2, and PKD3 are expressed in primary sensory neurons that mediate neurogenic inflammation and pain transmission, and PAR(2) agonists activate PKDs in HEK293 cells and DRG neurons in culture and in intact animals. PKD may be a novel component of a signal transduction pathway for protease-induced activation of nociceptive neurons and an important new target for antiinflammatory and analgesic therapies.


Subject(s)
Nociceptors/metabolism , Protein Kinase C/metabolism , Receptor, PAR-2/agonists , Sensory Receptor Cells/metabolism , Animals , Cell Line , Cell Membrane/physiology , Cells, Cultured , Enzyme Activation , Ganglia, Spinal/metabolism , Humans , Isoenzymes/metabolism , Male , Mice , Mice, Inbred C57BL , Neuropeptides/metabolism , Protein Kinase D2 , Protein Kinases/metabolism , Rats , Rats, Sprague-Dawley , Receptor, PAR-2/metabolism , TRPV Cation Channels/metabolism
6.
J Biol Chem ; 284(33): 22411-22425, 2009 Aug 14.
Article in English | MEDLINE | ID: mdl-19531493

ABSTRACT

Neuropeptide signaling at the cell surface is regulated by metalloendopeptidases, which degrade peptides in the extracellular fluid, and beta-arrestins, which interact with G protein-coupled receptors (GPCRs) to mediate desensitization. beta-Arrestins also recruit GPCRs and mitogen-activated protein kinases to endosomes to allow internalized receptors to continue signaling, but the mechanisms regulating endosomal signaling are unknown. We report that endothelin-converting enzyme-1 (ECE-1) degrades substance P (SP) in early endosomes of epithelial cells and neurons to destabilize the endosomal mitogen-activated protein kinase signalosome and terminate signaling. ECE-1 inhibition caused endosomal retention of the SP neurokinin 1 receptor, beta-arrestins, and Src, resulting in markedly sustained ERK2 activation in the cytosol and nucleus, whereas ECE-1 overexpression attenuated ERK2 activation. ECE-1 inhibition also enhanced SP-induced expression and phosphorylation of the nuclear death receptor Nur77, resulting in cell death. Thus, endosomal ECE-1 attenuates ERK2-mediated SP signaling in the nucleus to prevent cell death. We propose that agonist availability in endosomes, here regulated by ECE-1, controls beta-arrestin-dependent signaling of endocytosed GPCRs.


Subject(s)
Arrestins/metabolism , Aspartic Acid Endopeptidases/chemistry , Aspartic Acid Endopeptidases/physiology , Endosomes/metabolism , Metalloendopeptidases/chemistry , Metalloendopeptidases/physiology , Mitogen-Activated Protein Kinase 1/metabolism , Animals , Cell Nucleus/metabolism , Cytosol/metabolism , DNA-Binding Proteins/metabolism , Endothelin-Converting Enzymes , Humans , MAP Kinase Signaling System , Male , Mice , Mice, Inbred C57BL , Models, Biological , Nuclear Receptor Subfamily 4, Group A, Member 1 , Rats , Rats, Sprague-Dawley , Receptors, Steroid/metabolism , Signal Transduction , beta-Arrestins
7.
Proc Natl Acad Sci U S A ; 105(33): 12045-50, 2008 Aug 19.
Article in English | MEDLINE | ID: mdl-18687886

ABSTRACT

Prostaglandins (PG) are known to induce pain perception indirectly by sensitizing nociceptors. Accordingly, the analgesic action of nonsteroidal anti-inflammatory drugs (NSAIDs) results from inhibition of cyclooxygenases and blockade of PG biosynthesis. Cyclopentenone PGs, 15-d-PGJ(2), PGA(2), and PGA(1), formed by dehydration of their respective parent PGs, PGD(2), PGE(2), and PGE(1), possess a highly reactive alpha,beta-unsaturated carbonyl group that has been proposed to gate the irritant transient receptor potential A1 (TRPA1) channel. Here, by using TRPA1 wild-type (TRPA1(+/+)) or deficient (TRPA1(-/-)) mice, we show that cyclopentenone PGs produce pain by direct stimulation of nociceptors via TRPA1 activation. Cyclopentenone PGs caused a robust calcium response in dorsal root ganglion (DRG) neurons of TRPA1(+/+), but not of TRPA1(-/-) mice, and a calcium-dependent release of sensory neuropeptides from the rat dorsal spinal cord. Intraplantar injection of cyclopentenone PGs stimulated c-fos expression in spinal neurons of the dorsal horn and evoked an instantaneous, robust, and transient nociceptive response in TRPA1(+/+) but not in TRPA1(-/-) mice. The classical proalgesic PG, PGE(2), caused a slight calcium response in DRG neurons, increased c-fos expression in spinal neurons, and induced a delayed and sustained nociceptive response in both TRPA1(+/+) and TRPA1(-/-) mice. These results expand the mechanism of NSAID analgesia from blockade of indirect nociceptor sensitization by classical PGs to inhibition of direct TRPA1-dependent nociceptor activation by cyclopentenone PGs. Thus, TRPA1 antagonism may contribute to suppress pain evoked by PG metabolites without the adverse effects of inhibiting cyclooxygenases.


Subject(s)
Fatty Acids/metabolism , Pain/metabolism , Prostaglandin-Endoperoxide Synthases/metabolism , Transient Receptor Potential Channels/metabolism , Animals , Calcium/metabolism , Ganglia, Spinal/metabolism , Mice , Mice, Knockout , Neurons/metabolism , Nociceptors/metabolism , Pain/genetics , Rats , TRPA1 Cation Channel , Tissue Culture Techniques , Transient Receptor Potential Channels/deficiency , Transient Receptor Potential Channels/genetics
8.
J Clin Invest ; 118(7): 2574-82, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18568077

ABSTRACT

Cigarette smoke (CS) inhalation causes an early inflammatory response in rodent airways by stimulating capsaicin-sensitive sensory neurons that express transient receptor potential cation channel, subfamily V, member 1 (TRPV1) through an unknown mechanism that does not involve TRPV1. We hypothesized that 2 alpha,beta-unsaturated aldehydes present in CS, crotonaldehyde and acrolein, induce neurogenic inflammation by stimulating TRPA1, an excitatory ion channel coexpressed with TRPV1 on capsaicin-sensitive nociceptors. We found that CS aqueous extract (CSE), crotonaldehyde, and acrolein mobilized Ca2+ in cultured guinea pig jugular ganglia neurons and promoted contraction of isolated guinea pig bronchi. These responses were abolished by a TRPA1-selective antagonist and by the aldehyde scavenger glutathione but not by the TRPV1 antagonist capsazepine or by ROS scavengers. Treatment with CSE or aldehydes increased Ca2+ influx in TRPA1-transfected cells, but not in control HEK293 cells, and promoted neuropeptide release from isolated guinea pig airway tissue. Furthermore, the effect of CSE and aldehydes on Ca2+ influx in dorsal root ganglion neurons was abolished in TRPA1-deficient mice. These data identify alpha,beta-unsaturated aldehydes as the main causative agents in CS that via TRPA1 stimulation mediate airway neurogenic inflammation and suggest a role for TRPA1 in the pathogenesis of CS-induced diseases.


Subject(s)
Acrolein/pharmacology , Aldehydes/pharmacology , Neurogenic Inflammation/physiopathology , Nicotiana/chemistry , Smoke , Transient Receptor Potential Channels/physiology , Acrolein/analogs & derivatives , Animals , Ankyrins , Calcitonin Gene-Related Peptide/metabolism , Calcium Channels/genetics , Calcium Channels/physiology , Calcium Signaling/drug effects , Calcium Signaling/physiology , Cell Line , Ganglia, Spinal/cytology , Guinea Pigs , Humans , Lung/drug effects , Lung/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurogenic Inflammation/chemically induced , Neurogenic Inflammation/metabolism , Neurons, Afferent/drug effects , Neurons, Afferent/metabolism , Rats , Substance P/metabolism , TRPA1 Cation Channel , TRPC Cation Channels , Transient Receptor Potential Channels/agonists , Transient Receptor Potential Channels/antagonists & inhibitors , Transient Receptor Potential Channels/deficiency , Transient Receptor Potential Channels/genetics
9.
J Biol Chem ; 282(36): 26089-100, 2007 Sep 07.
Article in English | MEDLINE | ID: mdl-17623652

ABSTRACT

Although principally produced by the pancreas to degrade dietary proteins in the intestine, trypsins are also expressed in the nervous system and in epithelial tissues, where they have diverse actions that could be mediated by protease-activated receptors (PARs). We examined the biological actions of human trypsin IV (or mesotrypsin) and rat p23, inhibitor-resistant forms of trypsin. The zymogens trypsinogen IV and pro-p23 were expressed in Escherichia coli and purified to apparent homogeneity. Enteropeptidase cleaved both zymogens, liberating active trypsin IV and p23, which were resistant to soybean trypsin inhibitor and aprotinin. Trypsin IV cleaved N-terminal fragments of PAR(1), PAR(2), and PAR(4) at sites that would expose the tethered ligand (PAR(1) = PAR(4) > PAR(2)). Trypsin IV increased [Ca(2+)](i) in transfected cells expressing human PAR(1) and PAR(2) with similar potencies (PAR(1), 0.5 microm; PAR(2), 0.6 microm). p23 also cleaved fragments of PAR(1) and PAR(2) and signaled to cells expressing these receptors. Trypsin IV and p23 increased [Ca(2+)](i) in rat dorsal root ganglion neurons that responded to capsaicin and which thus mediate neurogenic inflammation and nociception. Intraplantar injection of trypsin IV and p23 in mice induced edema and granulocyte infiltration, which were not observed in PAR (-/-)(1)(trypsin IV) and PAR (-/-)(2) (trypsin IV and p23) mice. Trypsin IV and p23 caused thermal hyperalgesia and mechanical allodynia and hyperalgesia in mice, and these effects were absent in PAR (-/-)(2) mice but maintained in PAR (-/-)(1) mice. Thus, trypsin IV and p23 are inhibitor-resistant trypsins that can cleave and activate PARs, causing PAR(1)- and PAR(2)-dependent inflammation and PAR(2)-dependent hyperalgesia.


Subject(s)
Calcium Signaling , Hyperalgesia/metabolism , Receptor, PAR-1/metabolism , Receptor, PAR-2/physiology , Trypsin/metabolism , Animals , Aprotinin/chemistry , Calcium Signaling/drug effects , Capsaicin/pharmacology , Edema/chemically induced , Edema/genetics , Edema/metabolism , Edema/pathology , Enteropeptidase/chemistry , Ganglia, Spinal/metabolism , Ganglia, Spinal/pathology , Granulocytes/metabolism , Granulocytes/pathology , Humans , Hyperalgesia/chemically induced , Hyperalgesia/genetics , Hyperalgesia/pathology , Inflammation/chemically induced , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Male , Mice , Mice, Knockout , Nociceptors/metabolism , Nociceptors/pathology , Pain Measurement , Rats , Rats, Sprague-Dawley , Receptor, PAR-1/deficiency , Receptor, PAR-2/deficiency , Receptors, Proteinase-Activated/metabolism , Receptors, Thrombin/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Trypsin/chemistry , Trypsin/genetics , Trypsin/pharmacology , Trypsin Inhibitors/chemistry
10.
Gastroenterology ; 132(7): 2422-37, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17570216

ABSTRACT

BACKGROUND & AIMS: We studied the role of protease-activated receptor 2 (PAR(2)) and its activating enzymes, trypsins and tryptase, in Clostridium difficile toxin A (TxA)-induced enteritis. METHODS: We injected TxA into ileal loops in PAR(2) or dipeptidyl peptidase I (DPPI) knockout mice or in wild-type mice pretreated with tryptase inhibitors (FUT-175 or MPI-0442352) or soybean trypsin inhibitor. We examined the effect of TxA on expression and activity of PAR(2) and trypsin IV messenger RNA in the ileum and cultured colonocytes. We injected activating peptide (AP), trypsins, tryptase, and p23 in wild-type mice, some pretreated with the neurokinin 1 receptor antagonist SR140333. RESULTS: TxA increased fluid secretion, myeloperoxidase activity in fluid and tissue, and histologic damage. PAR(2) deletion decreased TxA-induced ileitis, reduced luminal fluid secretion by 20%, decreased tissue and fluid myeloperoxidase by 50%, and diminished epithelial damage, edema, and neutrophil infiltration. DPPI deletion reduced secretion by 20% and fluid myeloperoxidase by 55%. In wild-type mice, FUT-175 or MPI-0442352 inhibited secretion by 24%-28% and tissue and fluid myeloperoxidase by 31%-71%. Soybean trypsin inhibitor reduced secretion to background levels and tissue myeloperoxidase by up to 50%. TxA increased expression of PAR(2) and trypsin IV in enterocytes and colonocytes and caused a 2-fold increase in Ca(2+) responses to PAR(2) AP. AP, tryptase, and trypsin isozymes (trypsin I/II, trypsin IV, p23) caused ileitis. SR140333 prevented AP-induced ileitis. CONCLUSIONS: PAR(2) and its activators are proinflammatory in TxA-induced enteritis. TxA stimulates existing PAR(2) and up-regulates PAR(2) and activating proteases, and PAR(2) causes inflammation by neurogenic mechanisms.


Subject(s)
Bacterial Toxins , Cathepsin C/metabolism , Enteritis/chemically induced , Enterotoxins , Peptide Hydrolases/metabolism , Receptor, PAR-2/metabolism , Animals , Bacterial Toxins/pharmacology , Cathepsin C/deficiency , Cells, Cultured , Colon/cytology , Colon/metabolism , Enteritis/etiology , Enteritis/metabolism , Enteritis/pathology , Enterotoxins/pharmacology , Granulocytes/pathology , Ileitis/etiology , Ileum/metabolism , Intestinal Mucosa/metabolism , Intestines/pathology , Mice , Mice, Knockout , Nervous System/metabolism , Peroxidase/metabolism , Receptor, PAR-2/deficiency , Receptors, Neurokinin-1/metabolism , Trypsin/metabolism , Trypsin Inhibitors/pharmacology , Tryptases/pharmacology , Up-Regulation
11.
J Physiol ; 580(Pt.3): 977-91, 2007 May 01.
Article in English | MEDLINE | ID: mdl-17289784

ABSTRACT

Agonists of protease-activated receptor 2 (PAR(2)) evoke hyperexcitability of dorsal root ganglia (DRG) neurons by unknown mechanisms. We examined the cellular mechanisms underlying PAR(2)-evoked hyperexcitability of mouse colonic DRG neurons to determine their potential role in pain syndromes such as visceral hyperalgesia. Colonic DRG neurons were identified by injecting Fast Blue and DiI retrograde tracers into the mouse colon. Using immunofluorescence, we found that DiI-labelled neurons contained PAR(2) immunoreactivity, confirming the presence of receptors on colonic neurons. Whole-cell current-clamp recordings of acutely dissociated neurons demonstrated that PAR(2) activation with a brief application (3 min) of PAR(2) agonists, SLIGRL-NH(2) and trypsin, evoked sustained depolarizations (up to 60 min) which were associated with increased input resistance and a marked reduction in rheobase (50% at 30 min). In voltage clamp, SLIGRL-NH(2) markedly suppressed delayed rectifier I(K) currents (55% at 10 min), but had no effect on the transient I(A) current or TTX-resistant Na(+) currents. In whole-cell current-clamp recordings, the sustained excitability evoked by PAR(2) activation was blocked by the PKC inhibitor, calphostin, and the ERK(1/2) inhibitor PD98059. Studies of ERK(1/2) phosphorylation using confocal microscopy demonstrated that SLIGRL-NH(2) increased levels of immunoreactive pERK(1/2) in DRG neurons, particularly in proximity to the plasma membrane. Thus, activation of PAR(2) receptors on colonic nociceptive neurons causes sustained hyperexcitability that is related, at least in part, to suppression of delayed rectifier I(K) currents. Both PKC and ERK(1/2) mediate the PAR(2)-induced hyperexcitability. These studies describe a novel mechanism of sensitization of colonic nociceptive neurons that may be implicated in conditions of visceral hyperalgesia such as irritable bowel syndrome.


Subject(s)
Colon/innervation , Nociceptors/physiology , Receptor, PAR-2/physiology , Animals , Cell Membrane/enzymology , Cells, Cultured , Drug Resistance , Electric Conductivity , Enzyme Activation/physiology , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Ganglia, Spinal/cytology , Ganglia, Spinal/metabolism , Male , Mice , Mice, Inbred Strains , Neurons/metabolism , Neurons/physiology , Oligopeptides/pharmacology , Patch-Clamp Techniques , Phosphorylation/drug effects , Potassium Channels/drug effects , Potassium Channels/physiology , Protein Kinase C/metabolism , Receptor, PAR-2/agonists , Sodium Channels/drug effects , Sodium Channels/physiology , Tetrodotoxin/pharmacology , Trypsin/pharmacology
12.
J Physiol ; 578(Pt 3): 715-33, 2007 Feb 01.
Article in English | MEDLINE | ID: mdl-17124270

ABSTRACT

Exacerbated sensitivity to mechanical stimuli that are normally innocuous or mildly painful (mechanical allodynia and hyperalgesia) occurs during inflammation and underlies painful diseases. Proteases that are generated during inflammation and disease cleave protease-activated receptor 2 (PAR2) on afferent nerves to cause mechanical hyperalgesia in the skin and intestine by unknown mechanisms. We hypothesized that PAR2-mediated mechanical hyperalgesia requires sensitization of the ion channel transient receptor potential vanilloid 4 (TRPV4). Immunoreactive TRPV4 was coexpressed by rat dorsal root ganglia (DRG) neurons with PAR2, substance P (SP) and calcitonin gene-related peptide (CGRP), mediators of pain transmission. In PAR2-expressing cell lines that either naturally expressed TRPV4 (bronchial epithelial cells) or that were transfected to express TRPV4 (HEK cells), pretreatment with a PAR2 agonist enhanced Ca2+ and current responses to the TRPV4 agonists phorbol ester 4alpha-phorbol 12,13-didecanoate (4alphaPDD) and hypotonic solutions. PAR2-agonist similarly sensitized TRPV4 Ca2+ signals and currents in DRG neurons. Antagonists of phospholipase Cbeta and protein kinases A, C and D inhibited PAR2-induced sensitization of TRPV4 Ca2+ signals and currents. 4alphaPDD and hypotonic solutions stimulated SP and CGRP release from dorsal horn of rat spinal cord, and pretreatment with PAR2 agonist sensitized TRPV4-dependent peptide release. Intraplantar injection of PAR2 agonist caused mechanical hyperalgesia in mice and sensitized pain responses to the TRPV4 agonists 4alphaPDD and hypotonic solutions. Deletion of TRPV4 prevented PAR2 agonist-induced mechanical hyperalgesia and sensitization. This novel mechanism, by which PAR2 activates a second messenger to sensitize TRPV4-dependent release of nociceptive peptides and induce mechanical hyperalgesia, may underlie inflammatory hyperalgesia in diseases where proteases are activated and released.


Subject(s)
Hyperalgesia/physiopathology , Receptor, PAR-2/metabolism , TRPV Cation Channels/metabolism , Animals , Calcitonin Gene-Related Peptide/metabolism , Calcium Signaling/physiology , Cell Line , Cells, Cultured , Epithelial Cells/metabolism , Ganglia, Spinal/metabolism , Humans , Hyperalgesia/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nociceptors/physiology , Pain/physiopathology , Peptides/metabolism , Rats , Rats, Sprague-Dawley , Receptor, PAR-2/genetics , Substance P/metabolism , TRPV Cation Channels/genetics
13.
Mol Pain ; 2: 35, 2006 Nov 15.
Article in English | MEDLINE | ID: mdl-17107623

ABSTRACT

Prokineticins (PKs), consisting of PK1 and PK2, are a pair of newly identified regulatory peptides. Two closely related G-protein coupled receptors, PKR1 and PKR2, mediate the signaling of PKs. PKs/PKRs participate in the regulation of diverse biological processes, ranging from development to adult physiology. A number of studies have indicated the involvement of PKs/PKRs in nociception. Here we show that PK2 is a sensitizer for nociception. Intraplantar injection of recombinant PK2 resulted in a strong and localized hyperalgesia with reduced thresholds to nociceptive stimuli. PK2 mobilizes calcium in dissociated dorsal root ganglion (DRG) neurons. Mice lacking the PK2 gene displayed strong reduction in nociception induced by thermal and chemical stimuli, including capsaicin. However, PK2 mutant mice showed no difference in inflammatory response to capsaicin. As the majority of PK2-responsive DRG neurons also expressed transient receptor potential vanilloid (TRPV1) and exhibited sensitivity to capsaicin, TRPV1 is likely a significant downstream molecule of PK2 signaling. Taken together, these results reveal that PK2 sensitize nociception without affecting inflammation.


Subject(s)
Gastrointestinal Hormones/physiology , Hypesthesia/genetics , Hypesthesia/physiopathology , Neuropeptides/physiology , Pain Threshold/physiology , Animals , Behavior, Animal , Capsaicin , Ganglia, Spinal/cytology , Gastrointestinal Hormones/deficiency , Gastrointestinal Hormones/genetics , Gastrointestinal Hormones/pharmacology , In Situ Hybridization/methods , Inflammation/chemically induced , Inflammation/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/metabolism , Neuropeptides/deficiency , Neuropeptides/genetics , Neuropeptides/pharmacology , Pain/genetics , Pain/physiopathology , Pain Measurement/methods , Pain Threshold/drug effects , Physical Stimulation/methods , Reaction Time/drug effects , Reaction Time/physiology , Stimulation, Chemical , TRPV Cation Channels/genetics , TRPV Cation Channels/metabolism
14.
J Physiol ; 575(Pt 2): 555-71, 2006 Sep 01.
Article in English | MEDLINE | ID: mdl-16793902

ABSTRACT

Proteases that are released during inflammation and injury cleave protease-activated receptor 2 (PAR2) on primary afferent neurons to cause neurogenic inflammation and hyperalgesia. PAR2-induced thermal hyperalgesia depends on sensitization of transient receptor potential vanilloid receptor 1 (TRPV1), which is gated by capsaicin, protons and noxious heat. However, the signalling mechanisms by which PAR2 sensitizes TRPV1 are not fully characterized. Using immunofluorescence and confocal microscopy, we observed that PAR2 was colocalized with protein kinase (PK) Cepsilon and PKA in a subset of dorsal root ganglia neurons in rats, and that PAR2 agonists promoted translocation of PKCepsilon and PKA catalytic subunits from the cytosol to the plasma membrane of cultured neurons and HEK 293 cells. Subcellular fractionation and Western blotting confirmed this redistribution of kinases, which is indicative of activation. Although PAR2 couples to phospholipase Cbeta, leading to stimulation of PKC, we also observed that PAR2 agonists increased cAMP generation in neurons and HEK 293 cells, which would activate PKA. PAR2 agonists enhanced capsaicin-stimulated increases in [Ca2+]i and whole-cell currents in HEK 293 cells, indicating TRPV1 sensitization. The combined intraplantar injection of non-algesic doses of PAR2 agonist and capsaicin decreased the latency of paw withdrawal to radiant heat in mice, indicative of thermal hyperalgesia. Antagonists of PKCepsilon and PKA prevented sensitization of TRPV1 Ca2+ signals and currents in HEK 293 cells, and suppressed thermal hyperalgesia in mice. Thus, PAR2 activates PKCepsilon and PKA in sensory neurons, and thereby sensitizes TRPV1 to cause thermal hyperalgesia. These mechanisms may underlie inflammatory pain, where multiple proteases are generated and released.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/physiology , Protein Kinase C-epsilon/physiology , Receptor, PAR-2/physiology , TRPV Cation Channels/physiology , Animals , Calcium Signaling/physiology , Capsaicin/pharmacology , Cell Line , Cells, Cultured , Cyclic AMP-Dependent Protein Kinases/genetics , Cyclic AMP-Dependent Protein Kinases/metabolism , Electrophysiology , Enzyme Activation/drug effects , Enzyme Activation/physiology , Ganglia, Spinal/cytology , Ganglia, Spinal/metabolism , Gene Expression Regulation, Enzymologic/physiology , Humans , Hyperalgesia/physiopathology , Male , Mice , Mice, Inbred C57BL , Neurons, Afferent/cytology , Neurons, Afferent/metabolism , Protein Kinase C-epsilon/genetics , Protein Kinase C-epsilon/metabolism , Rats , Rats, Sprague-Dawley , Receptor, PAR-2/agonists , Receptor, PAR-2/antagonists & inhibitors
15.
J Appl Physiol (1985) ; 101(2): 506-11, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16627674

ABSTRACT

A lowered threshold to the cough response frequently accompanies chronic airway inflammatory conditions. However, the mechanism(s) that from chronic inflammation results in a lowered cough threshold is poorly understood. Irritant agents, including capsaicin, resiniferatoxin, and citric acid, elicit cough in humans and in experimental animals through the activation of the transient receptor potential vanilloid 1 (TRPV1). Protease-activated receptor-2 (PAR2) activation plays a role in inflammation and sensitizes TRPV1 in cultured sensory neurons by a PKC-dependent pathway. Here, we have investigated whether PAR2 activation exaggerates TRPV1-dependent cough in guinea pigs and whether protein kinases are involved in the PAR2-induced cough modulation. Aerosolized PAR2 agonists (PAR2-activating peptide and trypsin) did not produce any cough per se. However, they potentiated citric acid- and resiniferatoxin-induced cough, an effect that was completely prevented by the TRPV1 receptor antagonist capsazepine. In contrast, cough induced by hypertonic saline, a stimulus that provokes cough in a TRPV1-independent manner, was not modified by aerosolized PAR2 agonists. The PKC inhibitor GF-109203X, the PKA inhibitor H-89, and the cyclooxygenase inhibitor indomethacin did not affect cough induced by TRPV1 agonists, but abated the exaggeration of this response produced by PAR2 agonists. In conclusion, PAR2 stimulation exaggerates TRPV1-dependent cough by activation of diverse mechanism(s), including PKC, PKA, and prostanoid release. PAR2 activation, by sensitizing TRPV1 in primary sensory neurons, may play a role in the exaggerated cough observed in certain airways inflammatory diseases such as asthma and chronic obstructive pulmonary disease.


Subject(s)
Cough/physiopathology , Guinea Pigs/physiology , Receptor, PAR-2/physiology , TRPV Cation Channels/physiology , Animals , Capsaicin/analogs & derivatives , Capsaicin/pharmacology , Citric Acid/adverse effects , Cough/chemically induced , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic AMP-Dependent Protein Kinases/physiology , Cyclooxygenase Inhibitors/pharmacology , Diterpenes/adverse effects , Indomethacin/pharmacology , Inflammation , Isoquinolines/pharmacology , Male , Neurons, Afferent/physiology , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/physiology , Protein Kinase Inhibitors/pharmacology , Saline Solution, Hypertonic/adverse effects , Sulfonamides/pharmacology , Trypsin/pharmacology
16.
J Biol Chem ; 280(36): 31936-48, 2005 Sep 09.
Article in English | MEDLINE | ID: mdl-16027150

ABSTRACT

Tight junctions between intestinal epithelial cells prevent ingress of luminal macromolecules and bacteria and protect against inflammation and infection. During stress and inflammation, mast cells mediate increased mucosal permeability by unknown mechanisms. We hypothesized that mast cell tryptase cleaves protease-activated receptor 2 (PAR2) on colonocytes to increase paracellular permeability. Colonocytes expressed PAR2 mRNA and responded to PAR2 agonists with increased [Ca2+]i. Supernatant from degranulated mast cells increased [Ca2+]i in colonocytes, which was prevented by a tryptase inhibitor, and desensitized responses to PAR2 agonist, suggesting PAR2 cleavage. When applied to the basolateral surface of colonocytes, PAR2 agonists and mast cell supernatant decreased transepithelial resistance, increased transepithelial flux of macromolecules, and induced redistribution of tight junction ZO-1 and occludin and perijunctional F-actin. When mast cells were co-cultured with colonocytes, mast cell degranulation increased paracellular permeability of colonocytes. This was prevented by a tryptase inhibitor. We determined the role of ERK1/2 and of beta-arrestins, which recruit ERK1/2 to PAR2 in endosomes and retain ERK1/2 in the cytosol, on PAR2-mediated alterations in permeability. An ERK1/2 inhibitor abolished the effects of PAR2 agonist on permeability and redistribution of F-actin. Down-regulation of beta-arrestins with small interfering RNA inhibited PAR2-induced activation of ERK1/2 and suppressed PAR2-induced changes in permeability. Thus, mast cells signal to colonocytes in a paracrine manner by release of tryptase and activation of PAR2. PAR2 couples to beta-arrestin-dependent activation of ERK1/2, which regulates reorganization of perijunctional F-actin to increase epithelial permeability. These mechanisms may explain the increased epithelial permeability of the intestine during stress and inflammation.


Subject(s)
Arrestins/physiology , Colon/cytology , Colon/physiology , Receptor, PAR-2/physiology , Serine Endopeptidases/physiology , Tight Junctions/physiology , Actins/metabolism , Calcium/metabolism , Cell Line , Coculture Techniques , Colon/enzymology , Humans , Mast Cells/enzymology , Mitogen-Activated Protein Kinase 1/physiology , Mitogen-Activated Protein Kinase 3/physiology , Permeability , RNA, Small Interfering , Receptor, PAR-2/agonists , Receptor, PAR-2/genetics , Signal Transduction/physiology , Tight Junctions/enzymology , Tryptases , beta-Arrestins
17.
Am J Physiol Lung Cell Mol Physiol ; 289(1): L153-8, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15792966

ABSTRACT

Agonists of the dopamine receptors have been demonstrated to have bronchodilatory properties in pathologically constricted airways. The mechanism by which these agonists induce bronchodilatation is thought to involve airway sensory nerves. In this study, the expression and function of dopamine D(2) receptor were examined in sensory ganglia supplying the airways. Neuronal dopamine D(2) receptor mRNA expression was demonstrated by single-cell RT-PCR following laser-assisted microdissection. The projection of the neurons to the airways was confirmed by retrograde neuronal labeling. In functional studies, dopamine D(2) receptor agonists (AR-C65116AB and ropinirole) inhibited intraneuronal calcium mobilization in rat capsaicin-sensitive primary sensory neurons and capsaicin-induced plasma extravasation in the rat trachea. Our results provide support to the hypothesis that dopamine D(2) receptor activation inhibits neurogenic inflammation and proinflammatory reflex responses.


Subject(s)
Nodose Ganglion/metabolism , Olfactory Receptor Neurons/metabolism , Receptors, Dopamine D2/biosynthesis , Trachea/metabolism , Airway Obstruction/physiopathology , Animals , Calcium Signaling/drug effects , Calcium Signaling/physiology , Capsaicin/pharmacology , Dopamine Agonists/pharmacology , Indoles/pharmacology , Inflammation/metabolism , Male , Nodose Ganglion/cytology , Olfactory Receptor Neurons/cytology , Permeability/drug effects , Rats , Rats, Sprague-Dawley , Receptors, Dopamine D2/agonists , Receptors, Drug/metabolism
18.
Curr Opin Pharmacol ; 4(6): 551-6, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15525542

ABSTRACT

Serine proteases from the circulation, inflammatory cells, digestive glands and microorganisms can signal to cells by cleaving protease-activated receptors (PARs), a family of four G-protein-coupled receptors. Proteases cleave PARs at specific sites to expose tethered ligand domains that bind to and activate the cleaved receptors. Despite this irreversible mechanism of activation, PAR signaling is tightly regulated to prevent the uncontrolled stimulation of cells. Although PARs are found in all organ systems, protease signaling is of particular interest in the gastrointestinal tract, where proteases regulate neurotransmission, secretion, motility, epithelial permeability and intestinal inflammation, and can thus contribute to disease.


Subject(s)
Gastrointestinal Tract/physiology , Receptors, Proteinase-Activated/physiology , Serine Endopeptidases/physiology , Signal Transduction/drug effects , Animals , Gastrointestinal Tract/drug effects , Gastrointestinal Tract/microbiology , Humans , Inflammatory Bowel Diseases/metabolism , Inflammatory Bowel Diseases/physiopathology , Models, Biological , Receptors, Proteinase-Activated/drug effects , Serine Endopeptidases/pharmacology
19.
J Neurosci ; 24(18): 4300-12, 2004 May 05.
Article in English | MEDLINE | ID: mdl-15128844

ABSTRACT

Inflammatory proteases (mast cell tryptase and trypsins) cleave protease-activated receptor 2 (PAR2) on spinal afferent neurons and cause persistent inflammation and hyperalgesia by unknown mechanisms. We determined whether transient receptor potential vanilloid receptor 1 (TRPV1), a cation channel activated by capsaicin, protons, and noxious heat, mediates PAR2-induced hyperalgesia. PAR2 was coexpressed with TRPV1 in small- to medium-diameter neurons of the dorsal root ganglia (DRG), as determined by immunofluorescence. PAR2 agonists increased intracellular [Ca2+] ([Ca2+]i) in these neurons in culture, and PAR2-responsive neurons also responded to the TRPV1 agonist capsaicin, confirming coexpression of PAR2 and TRPV1. PAR2 agonists potentiated capsaicin-induced increases in [Ca2+]i in TRPV1-transfected human embryonic kidney (HEK) cells and DRG neurons and potentiated capsaicin-induced currents in DRG neurons. Inhibitors of phospholipase C and protein kinase C (PKC) suppressed PAR2-induced sensitization of TRPV1-mediated changes in [Ca2+]i and TRPV1 currents. Activation of PAR2 or PKC induced phosphorylation of TRPV1 in HEK cells, suggesting a direct regulation of the channel. Intraplantar injection of a PAR2 agonist caused persistent thermal hyperalgesia that was prevented by antagonism or deletion of TRPV1. Coinjection of nonhyperalgesic doses of PAR2 agonist and capsaicin induced hyperalgesia that was inhibited by deletion of TRPV1 or antagonism of PKC. PAR2 activation also potentiated capsaicin-induced release of substance P and calcitonin gene-related peptide from superfused segments of the dorsal horn of the spinal cord, where they mediate hyperalgesia. We have identified a novel mechanism by which proteases that activate PAR2 sensitize TRPV1 through PKC. Antagonism of PAR2, TRPV1, or PKC may abrogate protease-induced thermal hyperalgesia.


Subject(s)
Hyperalgesia/metabolism , Receptor, PAR-2/metabolism , Receptors, Drug/metabolism , Animals , Calcitonin Gene-Related Peptide/metabolism , Calcium/metabolism , Capsaicin/pharmacology , Cells, Cultured , Enzyme Activators/pharmacology , Enzyme Inhibitors/pharmacology , Humans , Hyperalgesia/chemically induced , Kidney/cytology , Kidney/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/metabolism , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptor, PAR-2/agonists , Receptor, PAR-2/genetics , Receptors, Drug/agonists , Receptors, Drug/genetics , Spinal Cord/drug effects , Spinal Cord/metabolism , Substance P/metabolism , Type C Phospholipases/antagonists & inhibitors , Type C Phospholipases/metabolism
20.
Regul Pept ; 118(3): 127-34, 2004 May 15.
Article in English | MEDLINE | ID: mdl-15003828

ABSTRACT

Although airway and pulmonary vessel tone are regulated predominantly by cholinergic and adrenergic impulses, biologically active peptides such as calcitonin gene-related peptide (CGRP) may significantly influence human smooth muscle tone in normal and pathophysiological states. In the present study, the expression of CGRP and its receptor CGRPR-1 and the biological effect of the peptide were investigated in human airways and pulmonary arteries. Immunohistochemistry revealed the presence of CGRP in human airway nerves and neuro-epithelial cells, whereas the receptor was found in epithelial cells and smooth muscle myocytes of the bronchi and in pulmonary artery endothelium. On precontracted bronchi (3-4 mm in diameter) alpha-CGRP (0.01-10 nM) caused a concentration-dependent contraction on epithelium-denuded bronchi, whereas no significant effect was recorded in bronchi with intact epithelium. In pulmonary arteries (2-6 mm in diameter), alpha-CGRP caused a concentration-dependent relaxation of endothelium intact and denuded vessels. Pre-treatment with indomethacin, but not with l-NAME, prevented the relaxation induced by alpha-CGRP in pulmonary arteries suggesting that prostaglandins but not nitric oxide (NO) are involved in the intracellular signal transduction pathway. The effects induced by alpha-CGRP in bronchi and vessels were prevented by application of the antagonist CGRP((8-37)). In summary, the present studies examined the biological function of CGRP in human airways and demonstrated a constrictory effect of CGRP only in epithelium-denuded airway smooth muscle indicating an alteration of CGRP airway effects in respiratory tract pathological states with damaged epithelium such as chronic obstructive pulmonary disease or bronchial asthma.


Subject(s)
Bronchi/physiology , Calcitonin Gene-Related Peptide/physiology , Muscle, Smooth/physiology , Pulmonary Artery/physiology , Acetylcholine/pharmacology , Bronchi/chemistry , Bronchi/drug effects , Calcitonin Gene-Related Peptide/analysis , Calcitonin Gene-Related Peptide/pharmacology , Dose-Response Relationship, Drug , Endothelium, Vascular/chemistry , Endothelium, Vascular/physiology , Epithelium/chemistry , Epithelium/physiology , Humans , Immunohistochemistry , In Vitro Techniques , Indomethacin/pharmacology , Muscle Contraction/drug effects , Muscle Relaxation/drug effects , Muscle, Smooth/chemistry , Muscle, Smooth/drug effects , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/physiology , Myocytes, Smooth Muscle/chemistry , NG-Nitroarginine Methyl Ester/pharmacology , Nerve Fibers/chemistry , Peptide Fragments/pharmacology , Phenylephrine/pharmacology , Pulmonary Artery/chemistry , Pulmonary Artery/drug effects , Receptors, Calcitonin Gene-Related Peptide/analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...