1
|
Arendt-Tranholm A, Sankaranarayanan I, Payne C, Moreno MM, Mazhar K, Yap N, Chiu AP, Barry A, Patel PP, Inturi NN, Tavares Ferreira D, Amin A, Karandikar M, Jarvik JG, Turner JA, Hofstetter CP, Curatolo M, Price TJ. Single-cell characterization of the human C2 dorsal root ganglion recovered from C1-2 arthrodesis surgery: implications for neck pain. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.24.645122. [PMID: 40196625 PMCID: PMC11974819 DOI: 10.1101/2025.03.24.645122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Neurons in the dorsal root ganglion (DRG) receive and transmit sensory information from the tissues they innervate and from the external environment. Upper cervical (C1-C2) DRGs are functionally unique as they receive input from the neck, head, and occipital cranial dura, the latter two of which are also innervated by the trigeminal ganglion (TG). The C2 DRG also plays an important role in neck pain, a common and disabling disorder that is poorly understood. Advanced transcriptomic approaches have significantly improved our ability to characterize RNA expression patterns at single-cell resolution in the DRG and TG, but no previous studies have characterized the C2 DRG. Our aim was to use single-nucleus and spatial transcriptomic approaches to create a molecular map of C2 DRGs from patients undergoing arthrodesis surgery with ganglionectomy. Patients with acute (<3 months) or chronic (≥3 months) neck pain were enrolled and completed patient-reported outcomes and quantitative sensory testing prior to surgery. C2 DRGs were characterized with bulk, single nucleus, and spatial RNA sequencing technologies from 22 patients. Through a comparative analysis to published datasets of the lumbar DRG and TG, neuronal clusters identified in both TG and DRG were identified in the C2 DRG. Therefore, our study definitively characterizes the molecular composition of human C2 neurons and establishes their similarity with unique characteristics of subsets of TG neurons. We identified differentially expressed genes in endothelial, fibroblast and myelinating Schwann cells associated with chronic pain, including FGFBP2, C8orf34 and EFNA1 which have been identified in previous genome and transcriptome wide association studies (GWAS/TWAS). Our work establishes an atlas of the human C2 DRG and identifies altered gene expression patterns associated with chronic neck pain. This work establishes a foundation for the exploration of painful disorders in humans affecting the cervical spine.
Collapse
|
2
|
Tan X, Yan C, Zou G, Jing R. Neurogenic differentiation 2 promotes inflammatory activation of macrophages in doxorubicin-induced myocarditis via regulating protein kinase D. BMC Cardiovasc Disord 2025; 25:195. [PMID: 40102732 PMCID: PMC11916933 DOI: 10.1186/s12872-025-04626-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 03/03/2025] [Indexed: 03/20/2025] Open
Abstract
BACKGROUND Although it has been established that protein kinase D (PKD) plays a crucial role in various diseases, its precise role in myocarditis remains elusive. METHODS To investigate PKD's involvement in myocarditis, we established a mouse model of myocarditis using doxorubicin (DOX) to assess cardiac function, observe pathological changes, and quantify inflammatory cytokine levels in heart tissues. Additionally, macrophages were isolated from heart tissues of both control and DOX-treated groups to assess PKD expression and inflammatory cytokines in these macrophages. We explored the molecular mechanism of Neurogenic Differentiation 2 (NeuroD2) in myocarditis by utilizing NeuroD2 overexpression plasmids and NeuroD2 small interfering RNA (siRNA). Furthermore, we conducted dual-luciferase reporter and chromatin immunoprecipitation (ChIP) assays to investigate the interaction between NeuroD2 and PKD. RESULTS We observed significant upregulation of PKD in macrophages and heart tissues induced by DOX. The administration of a PKD inhibitor reduced inflammatory cytokine levels, improved cardiac function, and mitigated pathological changes in myocarditis-afflicted mice. Mechanistically, we found upregulated expression of NeuroD2 in both macrophages and heart tissues exposed to DOX. NeuroD2 could directly target PKD, enhancing the NLRP3/NF-κB signaling pathway and exacerbating macrophage inflammation. CONCLUSIONS Our study demonstrates that NeuroD2 can directly bind to the PKD promoter, potentially promoting inflammatory activation of macrophages in DOX-induced myocarditis via the NLRP3/NF-κB pathway. This suggests that the NeuroD2/PKD axis may hold promise as a potential therapeutic approach for treating DOX-induced myocarditis.
Collapse
Affiliation(s)
- Xinyu Tan
- Xiangya hospital of central south university, No. 87, Xiangya road, Changsha, China
| | - Changyu Yan
- Xiangya hospital of central south university, No. 87, Xiangya road, Changsha, China
| | - Gang Zou
- Zhong Ke Zhu Ying Intelligence Technology Academy, No. 96, Tongzipo road, Changsha, China
| | - Ran Jing
- Xiangya hospital of central south university, No. 87, Xiangya road, Changsha, China.
| |
Collapse
|
3
|
Schlett K, Oueslati Morales CO, Bencsik N, Hausser A. Getting smart - Deciphering the neuronal functions of protein kinase D. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119812. [PMID: 39147241 DOI: 10.1016/j.bbamcr.2024.119812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 08/05/2024] [Indexed: 08/17/2024]
Abstract
Protein kinase D (PKD) is a family of serine/threonine kinases that play important roles in various signalling pathways in cells, including neuronal cells. In the nervous system, PKD has been shown to be involved in learning and memory formation by regulating neurotransmitter release, neurite outgrowth and dendrite development, synapse formation and synaptic plasticity. In addition, PKD has been implicated in pain perception or neuroprotection during oxidative stress. Dysregulation of PKD expression and activity has been linked to several neurological disorders, including autism and epilepsy. In this review, we summarize the current knowledge on the function of the PKD family members in neuronal cells, including the spatial regulation of their downstream signalling pathways. We will further discuss the potential role of PKD in the pathogenesis of neurological disorders.
Collapse
Affiliation(s)
- Katalin Schlett
- Neuronal Cell Biology Group, Department of Physiology and Neurobiology, Eötvös Loránd University, Budapest, Hungary
| | - Carlos O Oueslati Morales
- Membrane Trafficking and Signalling Group, Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
| | - Norbert Bencsik
- Neuronal Cell Biology Group, Department of Physiology and Neurobiology, Eötvös Loránd University, Budapest, Hungary
| | - Angelika Hausser
- Membrane Trafficking and Signalling Group, Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany; Stuttgart Research Center Systems Biology, University of Stuttgart, Stuttgart, Germany.
| |
Collapse
|
4
|
Cluster Analysis of Short Sensory Profile Data Reveals Sensory-Based Subgroups in Autism Spectrum Disorder. Int J Mol Sci 2022; 23:ijms232113030. [PMID: 36361815 PMCID: PMC9655407 DOI: 10.3390/ijms232113030] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Accepted: 10/24/2022] [Indexed: 01/27/2023] Open
Abstract
Autism spectrum disorder is a common, heterogeneous neurodevelopmental disorder lacking targeted treatments. Additional features include restricted, repetitive patterns of behaviors and differences in sensory processing. We hypothesized that detailed sensory features including modality specific hyper- and hypo-sensitivity could be used to identify clinically recognizable subgroups with unique underlying gene variants. Participants included 378 individuals with a clinical diagnosis of autism spectrum disorder who contributed Short Sensory Profile data assessing the frequency of sensory behaviors and whole genome sequencing results to the Autism Speaks' MSSNG database. Sensory phenotypes in this cohort were not randomly distributed with 10 patterns describing 43% (162/378) of participants. Cross comparison of two independent cluster analyses on sensory responses identified six distinct sensory-based subgroups. We then characterized subgroups by calculating the percent of patients in each subgroup who had variants with a Combined Annotation Dependent Depletion (CADD) score of 15 or greater in each of 24,896 genes. Each subgroup exhibited a unique pattern of genes with a high frequency of variants. These results support the use of sensory features to identify autism spectrum disorder subgroups with shared genetic variants.
Collapse
|
5
|
Hirakawa M, Yokoyama T, Yamamoto Y, Saino T. Morphology of P2X3-immunoreactive basket-like afferent nerve endings surrounding serosal ganglia and close relationship with vesicular nucleotide transporter-immunoreactive nerve fibers in the rat gastric antrum. J Comp Neurol 2021; 529:3866-3881. [PMID: 34297862 DOI: 10.1002/cne.25219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 07/05/2021] [Accepted: 07/16/2021] [Indexed: 11/11/2022]
Abstract
We previously reported P2X3 purinoceptor (P2X3)-expressing vagal afferent nerve endings with large web-like structures in the subserosal tissue of the antral lesser curvature, suggesting that these nerve endings were one of the vagal mechanoreceptors. The present study investigated the morphological relationship between P2X3-immunoreactive nerve endings and serosal ganglia in the rat gastric antrum by immunohistochemistry of whole-mount preparations using confocal scanning laser microscopy. P2X3-immunoreactive basket-like subserosal nerve endings with new morphology were distributed laterally to the gastric sling muscles in the distal antrum of the lesser curvature. Parent axons ramified into numerous nerve fibers with pleomorphic flattened structures to form basket-like nerve endings, and the parent axons were originated from large net-like structures of vagal afferent nerve endings. Basket-like nerve endings wrapped around the whole serosal ganglia, which were characterized by neurofilament 200 kDa-immunoreactive neurons with or without neuronal nitric oxide synthase immunoreactivity and S100B-immunoreactive glial cells. Furthermore, basket-like nerve endings were localized in close apposition to dopamine beta-hydroxylase-immunoreactive sympathetic nerve fibers immunoreactive for vesicular nucleotide transporter. These results suggest that P2X3-immunoreactive basket-like nerve endings associated with serosal ganglia are the specialized ending structures of vagal subserosal mechanoreceptors in order to increase the sensitivity during antral peristalsis, and are activated by ATP from sympathetic nerve fibers and/or serosal ganglia for the regulation of mechanoreceptor function.
Collapse
Affiliation(s)
- Masato Hirakawa
- Department of Anatomy (Cell Biology), Iwate Medical University, Yahaba, Japan
| | - Takuya Yokoyama
- Department of Anatomy (Cell Biology), Iwate Medical University, Yahaba, Japan
| | - Yoshio Yamamoto
- Laboratory of Veterinary Anatomy and Cell Biology, Faculty of Agriculture, Iwate University, Morioka, Japan
| | - Tomoyuki Saino
- Department of Anatomy (Cell Biology), Iwate Medical University, Yahaba, Japan
| |
Collapse
|
6
|
Zhang X, Connelly J, Chao Y, Wang QJ. Multifaceted Functions of Protein Kinase D in Pathological Processes and Human Diseases. Biomolecules 2021; 11:biom11030483. [PMID: 33807058 PMCID: PMC8005150 DOI: 10.3390/biom11030483] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 03/13/2021] [Accepted: 03/15/2021] [Indexed: 02/06/2023] Open
Abstract
Protein kinase D (PKD) is a family of serine/threonine protein kinases operating in the signaling network of the second messenger diacylglycerol. The three family members, PKD1, PKD2, and PKD3, are activated by a variety of extracellular stimuli and transduce cell signals affecting many aspects of basic cell functions including secretion, migration, proliferation, survival, angiogenesis, and immune response. Dysregulation of PKD in expression and activity has been detected in many human diseases. Further loss- or gain-of-function studies at cellular levels and in animal models provide strong support for crucial roles of PKD in many pathological conditions, including cancer, metabolic disorders, cardiac diseases, central nervous system disorders, inflammatory diseases, and immune dysregulation. Complexity in enzymatic regulation and function is evident as PKD isoforms may act differently in different biological systems and disease models, and understanding the molecular mechanisms underlying these differences and their biological significance in vivo is essential for the development of safer and more effective PKD-targeted therapies. In this review, to provide a global understanding of PKD function, we present an overview of the PKD family in several major human diseases with more focus on cancer-associated biological processes.
Collapse
|
7
|
Pierre O, Fouchard M, Buscaglia P, Le Goux N, Leschiera R, Mignen O, Fluhr JW, Misery L, Le Garrec R. Calcium Increase and Substance P Release Induced by the Neurotoxin Brevetoxin-1 in Sensory Neurons: Involvement of PAR2 Activation through Both Cathepsin S and Canonical Signaling. Cells 2020; 9:E2704. [PMID: 33348659 PMCID: PMC7767211 DOI: 10.3390/cells9122704] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 12/14/2020] [Accepted: 12/15/2020] [Indexed: 12/17/2022] Open
Abstract
Red tides involving Karenia brevis expose humans to brevetoxins (PbTxs). Oral exposition triggers neurotoxic shellfish poisoning, whereas inhalation induces a respiratory syndrome and sensory disturbances. No curative treatment is available and the pathophysiology is not fully elucidated. Protease-activated receptor 2 (PAR2), cathepsin S (Cat-S) and substance P (SP) release are crucial mediators of the sensory effects of ciguatoxins (CTXs) which are PbTx analogs. This work explored the role of PAR2 and Cat-S in PbTx-1-induced sensory effects and deciphered the signaling pathway involved. We performed calcium imaging, PAR2 immunolocalization and SP release experiments in monocultured sensory neurons or co-cultured with keratinocytes treated with PbTx-1 or P-CTX-2. We demonstrated that PbTx-1-induced calcium increase and SP release involved Cat-S, PAR2 and transient receptor potential vanilloid 4 (TRPV4). The PbTx-1-induced signaling pathway included protein kinase A (PKA) and TRPV4, which are compatible with the PAR2 biased signaling induced by Cat-S. Internalization of PAR2 and protein kinase C (PKC), inositol triphosphate receptor and TRPV4 activation evoked by PbTx-1 are compatible with the PAR2 canonical signaling. Our results suggest that PbTx-1-induced sensory disturbances involve the PAR2-TRPV4 pathway. We identified PAR2, Cat-S, PKA, and PKC that are involved in TRPV4 sensitization induced by PbTx-1 in sensory neurons.
Collapse
Affiliation(s)
- Ophélie Pierre
- EA4685 Laboratory of Interactions Neurons-Keratinocytes (LIEN), Faculty of Medicine and Health Sciences, University Brest, F-29200 Brest, France; (M.F.); (R.L.); (J.W.F.); (L.M.); (R.L.G.)
| | - Maxime Fouchard
- EA4685 Laboratory of Interactions Neurons-Keratinocytes (LIEN), Faculty of Medicine and Health Sciences, University Brest, F-29200 Brest, France; (M.F.); (R.L.); (J.W.F.); (L.M.); (R.L.G.)
- Department of Dermatology, University Hospital of Brest, F-29200 Brest, France
| | - Paul Buscaglia
- InsermUMR1227, Lymphocytes B et Autoimmunity, University Brest, F-29200 Brest, France; (P.B.); (N.L.G.); (O.M.)
| | - Nelig Le Goux
- InsermUMR1227, Lymphocytes B et Autoimmunity, University Brest, F-29200 Brest, France; (P.B.); (N.L.G.); (O.M.)
| | - Raphaël Leschiera
- EA4685 Laboratory of Interactions Neurons-Keratinocytes (LIEN), Faculty of Medicine and Health Sciences, University Brest, F-29200 Brest, France; (M.F.); (R.L.); (J.W.F.); (L.M.); (R.L.G.)
| | - Olivier Mignen
- InsermUMR1227, Lymphocytes B et Autoimmunity, University Brest, F-29200 Brest, France; (P.B.); (N.L.G.); (O.M.)
| | - Joachim W. Fluhr
- EA4685 Laboratory of Interactions Neurons-Keratinocytes (LIEN), Faculty of Medicine and Health Sciences, University Brest, F-29200 Brest, France; (M.F.); (R.L.); (J.W.F.); (L.M.); (R.L.G.)
- Department of Dermatology, University Hospital of Brest, F-29200 Brest, France
- Department of Dermatology and Allergology, Universitaetsmedizin Charit Berlin, D-10117 Berlin, Germany
| | - Laurent Misery
- EA4685 Laboratory of Interactions Neurons-Keratinocytes (LIEN), Faculty of Medicine and Health Sciences, University Brest, F-29200 Brest, France; (M.F.); (R.L.); (J.W.F.); (L.M.); (R.L.G.)
- Department of Dermatology, University Hospital of Brest, F-29200 Brest, France
| | - Raphaële Le Garrec
- EA4685 Laboratory of Interactions Neurons-Keratinocytes (LIEN), Faculty of Medicine and Health Sciences, University Brest, F-29200 Brest, France; (M.F.); (R.L.); (J.W.F.); (L.M.); (R.L.G.)
| |
Collapse
|
8
|
Legumain Induces Oral Cancer Pain by Biased Agonism of Protease-Activated Receptor-2. J Neurosci 2020; 41:193-210. [PMID: 33172978 DOI: 10.1523/jneurosci.1211-20.2020] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Revised: 10/22/2020] [Accepted: 10/23/2020] [Indexed: 02/07/2023] Open
Abstract
Oral squamous cell carcinoma (OSCC) is one of the most painful cancers, which interferes with orofacial function including talking and eating. We report that legumain (Lgmn) cleaves protease-activated receptor-2 (PAR2) in the acidic OSCC microenvironment to cause pain. Lgmn is a cysteine protease of late endosomes and lysosomes that can be secreted; it exhibits maximal activity in acidic environments. The role of Lgmn in PAR2-dependent cancer pain is unknown. We studied Lgmn activation in human oral cancers and oral cancer mouse models. Lgmn was activated in OSCC patient tumors, compared with matched normal oral tissue. After intraplantar, facial or lingual injection, Lgmn evoked nociception in wild-type (WT) female mice but not in female mice lacking PAR2 in NaV1.8-positive neurons (Par2Nav1.8), nor in female mice treated with a Lgmn inhibitor, LI-1. Inoculation of an OSCC cell line caused mechanical and thermal hyperalgesia that was reversed by LI-1. Par2Nav1.8 and Lgmn deletion attenuated mechanical allodynia in female mice with carcinogen-induced OSCC. Lgmn caused PAR2-dependent hyperexcitability of trigeminal neurons from WT female mice. Par2 deletion, LI-1, and inhibitors of adenylyl cyclase or protein kinase A (PKA) prevented the effects of Lgmn. Under acidified conditions, Lgmn cleaved within the extracellular N terminus of PAR2 at Asn30↓Arg31, proximal to the canonical trypsin activation site. Lgmn activated PAR2 by biased mechanisms in HEK293 cells to induce Ca2+ mobilization, cAMP formation, and PKA/protein kinase D (PKD) activation, but not β-arrestin recruitment or PAR2 endocytosis. Thus, in the acidified OSCC microenvironment, Lgmn activates PAR2 by biased mechanisms that evoke cancer pain.SIGNIFICANCE STATEMENT Oral squamous cell carcinoma (OSCC) is one of the most painful cancers. We report that legumain (Lgmn), which exhibits maximal activity in acidic environments, cleaves protease-activated receptor-2 (PAR2) on neurons to produce OSCC pain. Active Lgmn was elevated in OSCC patient tumors, compared with matched normal oral tissue. Lgmn evokes pain-like behavior through PAR2 Exposure of pain-sensing neurons to Lgmn decreased the current required to generate an action potential through PAR2 Inhibitors of adenylyl cyclase and protein kinase A (PKA) prevented the effects of Lgmn. Lgmn activated PAR2 to induce calcium mobilization, cAMP formation, and activation of protein kinase D (PKD) and PKA, but not β-arrestin recruitment or PAR2 endocytosis. Thus, Lgmn is a biased agonist of PAR2 that evokes cancer pain.
Collapse
|
9
|
Zhao P, Pattison LA, Jensen DD, Jimenez-Vargas NN, Latorre R, Lieu T, Jaramillo JO, Lopez-Lopez C, Poole DP, Vanner SJ, Schmidt BL, Bunnett NW. Protein kinase D and Gβγ mediate sustained nociceptive signaling by biased agonists of protease-activated receptor-2. J Biol Chem 2019; 294:10649-10662. [PMID: 31142616 DOI: 10.1074/jbc.ra118.006935] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 05/23/2019] [Indexed: 12/20/2022] Open
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 β-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 β-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 Gβγ 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, Gβγ, and protein translation inhibited recovery of PAR2 responsiveness. PKD and Gβγ 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.
Collapse
Affiliation(s)
- Peishen Zhao
- From the Monash Institute of Pharmaceutical Sciences and Australian Research Council Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash University, Parkville, Victoria 3052, Australia
| | - Luke A Pattison
- From the Monash Institute of Pharmaceutical Sciences and Australian Research Council Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash University, Parkville, Victoria 3052, Australia
| | - Dane D Jensen
- the Departments of Surgery and Pharmacology, Columbia University Vagelos College of Physicians and Surgeons, Columbia University, New York, New York 10032
| | - Nestor N Jimenez-Vargas
- the Gastrointestinal Diseases Research Unit, Division of Gastroenterology, Queen's University, Kingston, Ontario K7L 3N6, Canada, and
| | - Rocco Latorre
- the Departments of Surgery and Pharmacology, Columbia University Vagelos College of Physicians and Surgeons, Columbia University, New York, New York 10032
| | - TinaMarie Lieu
- From the Monash Institute of Pharmaceutical Sciences and Australian Research Council Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash University, Parkville, Victoria 3052, Australia
| | - Josue O Jaramillo
- the Gastrointestinal Diseases Research Unit, Division of Gastroenterology, Queen's University, Kingston, Ontario K7L 3N6, Canada, and
| | - Cintya Lopez-Lopez
- the Gastrointestinal Diseases Research Unit, Division of Gastroenterology, Queen's University, Kingston, Ontario K7L 3N6, Canada, and
| | - Daniel P Poole
- From the Monash Institute of Pharmaceutical Sciences and Australian Research Council Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash University, Parkville, Victoria 3052, Australia
| | - Stephen J Vanner
- the Gastrointestinal Diseases Research Unit, Division of Gastroenterology, Queen's University, Kingston, Ontario K7L 3N6, Canada, and
| | - Brian L Schmidt
- the Bluestone Center for Clinical Research, New York University College of Dentistry, New York, New York 10010
| | - Nigel W Bunnett
- the Departments of Surgery and Pharmacology, Columbia University Vagelos College of Physicians and Surgeons, Columbia University, New York, New York 10032,
| |
Collapse
|
10
|
Lee YJ, Kim SJ, Kwon KW, Lee WM, Im WJ, Sohn UD. Inhibitory effect of FSLLRY-NH 2 on inflammatory responses induced by hydrogen peroxide in HepG2 cells. Arch Pharm Res 2017. [PMID: 28643288 DOI: 10.1007/s12272-017-0927-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Proteinase activated receptor 2 (PAR2), which is localized in the GI tract, the respiratory system, and the kidney tubules is a G protein-coupled receptor associated with inflammation, metabolism, and disease. The aim of this study was to explore the role of PAR2 in hydrogen peroxide (H2O2)-induced HepG2 cells by using FSLLRY-NH2 a PAR2 antagonist. H2O2 treatment resulted in induction of PAR2 in esophageal, gastric, and liver cells, with the most robust response being in HepG2 cells. Furthermore, this effect was dose-dependent in HepG2 cells. Treatment with H2O2 at concentrations above 400 μM for 24 h also reduced HepG2 cell viability. H2O2 treatment increased both the protein and mRNA levels of IL-1β, IL-8, and TNF-α, as well as those of SAPK/JNK. The increased levels of these pro-inflammatory genes and SAPK/JNK induced by H2O2 were attenuated in a dose-dependent manner when cells were co-treated with H2O2 and FSLLRY-NH2. In summary, the PAR2 antagonist peptide, FSLLRY-NH2, reduces the level of the pro-inflammatory genes IL-8, IL-1β, and TNF-α induced by H2O2, through the SAPK/JNK pathways in HepG2 cells. These data suggest that a PAR2 antagonist could be an anti-inflammatory agent in HepG2 cells.
Collapse
Affiliation(s)
- Yeon Joo Lee
- Department of Pharmacology, College of Pharmacy, Chung-Ang University, Seoul, 156-756, Republic of Korea
| | - Su Jin Kim
- Department of Pharmacology, College of Pharmacy, Chung-Ang University, Seoul, 156-756, Republic of Korea
| | - Kyoung Wan Kwon
- Department of Pharmacology, College of Pharmacy, Chung-Ang University, Seoul, 156-756, Republic of Korea
| | - Won Mo Lee
- Department of Pharmacology, College of Pharmacy, Chung-Ang University, Seoul, 156-756, Republic of Korea
| | - Wi Joon Im
- Department of Pharmacology, College of Pharmacy, Chung-Ang University, Seoul, 156-756, Republic of Korea
| | - Uy Dong Sohn
- Department of Pharmacology, College of Pharmacy, Chung-Ang University, Seoul, 156-756, Republic of Korea.
| |
Collapse
|
11
|
Mrozkova P, Spicarova D, Palecek J. Hypersensitivity Induced by Activation of Spinal Cord PAR2 Receptors Is Partially Mediated by TRPV1 Receptors. PLoS One 2016; 11:e0163991. [PMID: 27755539 PMCID: PMC5068818 DOI: 10.1371/journal.pone.0163991] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 09/19/2016] [Indexed: 12/23/2022] Open
Abstract
Protease-activated receptors 2 (PAR2) and transient receptor potential vanilloid 1 (TRPV1) receptors in the peripheral nerve endings are implicated in the development of increased sensitivity to mechanical and thermal stimuli, especially during inflammatory states. Both PAR2 and TRPV1 receptors are co-expressed in nociceptive dorsal root ganglion (DRG) neurons on their peripheral endings and also on presynaptic endings in the spinal cord dorsal horn. However, the modulation of nociceptive synaptic transmission in the superficial dorsal horn after activation of PAR2 and their functional coupling with TRPV1 is not clear. To investigate the role of spinal PAR2 activation on nociceptive modulation, intrathecal drug application was used in behavioural experiments and patch-clamp recordings of spontaneous, miniature and dorsal root stimulation-evoked excitatory postsynaptic currents (sEPSCs, mEPSCs, eEPSCs) were performed on superficial dorsal horn neurons in acute rat spinal cord slices. Intrathecal application of PAR2 activating peptide SLIGKV-NH2 induced thermal hyperalgesia, which was prevented by pretreatment with TRPV1 antagonist SB 366791 and was reduced by protein kinases inhibitor staurosporine. Patch-clamp experiments revealed robust decrease of mEPSC frequency (62.8 ± 4.9%), increase of sEPSC frequency (127.0 ± 5.9%) and eEPSC amplitude (126.9 ± 12.0%) in dorsal horn neurons after acute SLIGKV-NH2 application. All these EPSC changes, induced by PAR2 activation, were prevented by SB 366791 and staurosporine pretreatment. Our results demonstrate an important role of spinal PAR2 receptors in modulation of nociceptive transmission in the spinal cord dorsal horn at least partially mediated by activation of presynaptic TRPV1 receptors. The functional coupling between the PAR2 and TRPV1 receptors on the central branches of DRG neurons may be important especially during different pathological states when it may enhance pain perception.
Collapse
Affiliation(s)
- Petra Mrozkova
- Department of Functional Morphology, Institute of Physiology, The Czech Academy of Sciences, Prague, Czech Republic
| | - Diana Spicarova
- Department of Functional Morphology, Institute of Physiology, The Czech Academy of Sciences, Prague, Czech Republic
| | - Jiri Palecek
- Department of Functional Morphology, Institute of Physiology, The Czech Academy of Sciences, Prague, Czech Republic
- * E-mail:
| |
Collapse
|
12
|
Lieu T, Savage E, Zhao P, Edgington-Mitchell L, Barlow N, Bron R, Poole DP, McLean P, Lohman RJ, Fairlie DP, Bunnett NW. Antagonism of the proinflammatory and pronociceptive actions of canonical and biased agonists of protease-activated receptor-2. Br J Pharmacol 2016; 173:2752-65. [PMID: 27423137 PMCID: PMC4995288 DOI: 10.1111/bph.13554] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Revised: 07/05/2016] [Accepted: 07/05/2016] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND AND PURPOSE Diverse proteases cleave protease-activated receptor-2 (PAR2) on primary sensory neurons and epithelial cells to evoke pain and inflammation. Trypsin and tryptase activate PAR2 by a canonical mechanism that entails cleavage within the extracellular N-terminus revealing a tethered ligand that activates the cleaved receptor. Cathepsin-S and elastase are biased agonists that cleave PAR2 at different sites to activate distinct signalling pathways. Although PAR2 is a therapeutic target for inflammatory and painful diseases, the divergent mechanisms of proteolytic activation complicate the development of therapeutically useful antagonists. EXPERIMENTAL APPROACH We investigated whether the PAR2 antagonist GB88 inhibits protease-evoked activation of nociceptors and protease-stimulated oedema and hyperalgesia in rodents. KEY RESULTS Intraplantar injection of trypsin, cathespsin-S or elastase stimulated mechanical and thermal hyperalgesia and oedema in mice. Oral GB88 or par2 deletion inhibited the algesic and proinflammatory actions of all three proteases, but did not affect basal responses. GB88 also prevented pronociceptive and proinflammatory effects of the PAR2-selective agonists 2-furoyl-LIGRLO-NH2 and AC264613. GB88 did not affect capsaicin-evoked hyperalgesia or inflammation. Trypsin, cathepsin-S and elastase increased [Ca(2+) ]i in rat nociceptors, which expressed PAR2. GB88 inhibited this activation of nociceptors by all three proteases, but did not affect capsaicin-evoked activation of nociceptors or inhibit the catalytic activity of the three proteases. CONCLUSIONS AND IMPLICATIONS GB88 inhibits the capacity of canonical and biased protease agonists of PAR2 to cause nociception and inflammation.
Collapse
Affiliation(s)
- T Lieu
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
- Australian Research Council Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash University, Parkville, Australia
| | - E Savage
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
- Australian Research Council Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash University, Parkville, Australia
| | - P Zhao
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
- Australian Research Council Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash University, Parkville, Australia
| | - L Edgington-Mitchell
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
- Australian Research Council Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash University, Parkville, Australia
| | - N Barlow
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
- Australian Research Council Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash University, Parkville, Australia
| | - R Bron
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
- Australian Research Council Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash University, Parkville, Australia
| | - D P Poole
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
- Australian Research Council Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash University, Parkville, Australia
- Departments of Anatomy and Neuroscience, University of Melbourne, Melbourne, Australia
| | - P McLean
- Takeda Pharmaceuticals, Zurich, Switzerland
| | - R-J Lohman
- Centre for Inflammation and Disease Research and Centre for Pain Research, Institute of Molecular Bioscience, University of Queensland, Brisbane, Australia
- Australian Research Council Centre of Excellence in Advanced Molecular Imaging, University of Queensland, Brisbane, Australia
| | - D P Fairlie
- Centre for Inflammation and Disease Research and Centre for Pain Research, Institute of Molecular Bioscience, University of Queensland, Brisbane, Australia
- Australian Research Council Centre of Excellence in Advanced Molecular Imaging, University of Queensland, Brisbane, Australia
| | - N W Bunnett
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
- Australian Research Council Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash University, Parkville, Australia
- Departments of Pharmacology and Therapeutics, University of Melbourne, Melbourne, Australia
| |
Collapse
|
13
|
Jensen DD, Zhao P, Jimenez-Vargas NN, Lieu T, Gerges M, Yeatman HR, Canals M, Vanner SJ, Poole DP, Bunnett NW. Protein Kinase D and Gβγ Subunits Mediate Agonist-evoked Translocation of Protease-activated Receptor-2 from the Golgi Apparatus to the Plasma Membrane. J Biol Chem 2016; 291:11285-99. [PMID: 27030010 PMCID: PMC4900274 DOI: 10.1074/jbc.m115.710681] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Revised: 03/17/2016] [Indexed: 12/12/2022] Open
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 Gβγ 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 Gβγ, a PKD activator, to the Golgi apparatus, determined by bioluminescence resonance energy transfer between Gγ-Venus and giantin-Rluc8. Inhibitors of PKD (CRT0066101) and Gβγ (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 Gβγ in agonist-evoked mobilization of intracellular PAR2 stores that is required for sustained signaling by extracellular proteases.
Collapse
Affiliation(s)
- Dane D Jensen
- From the Monash Institute of Pharmaceutical Sciences and Australian Research Council Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash University, Parkville, Victoria 3052, Australia
| | - Peishen Zhao
- From the Monash Institute of Pharmaceutical Sciences and Australian Research Council Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash University, Parkville, Victoria 3052, Australia
| | - Nestor N Jimenez-Vargas
- the Gastrointestinal Diseases Research Unit, Division of Gastroenterology, Queen's University, Kingston, Ontario K7L 3N6, Canada
| | - TinaMarie Lieu
- From the Monash Institute of Pharmaceutical Sciences and Australian Research Council Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash University, Parkville, Victoria 3052, Australia
| | - Marina Gerges
- From the Monash Institute of Pharmaceutical Sciences and
| | | | - Meritxell Canals
- From the Monash Institute of Pharmaceutical Sciences and Australian Research Council Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash University, Parkville, Victoria 3052, Australia
| | - Stephen J Vanner
- the Gastrointestinal Diseases Research Unit, Division of Gastroenterology, Queen's University, Kingston, Ontario K7L 3N6, Canada
| | - Daniel P Poole
- From the Monash Institute of Pharmaceutical Sciences and Australian Research Council Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash University, Parkville, Victoria 3052, Australia, the Departments of Anatomy and Neuroscience and
| | - Nigel W Bunnett
- From the Monash Institute of Pharmaceutical Sciences and Australian Research Council Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash University, Parkville, Victoria 3052, Australia, Pharmacology and Therapeutics, University of Melbourne, Parkville, Victoria 3010, Australia, and
| |
Collapse
|
14
|
Mrozkova P, Palecek J, Spicarova D. The role of protease-activated receptor type 2 in nociceptive signaling and pain. Physiol Res 2016; 65:357-67. [PMID: 27070742 DOI: 10.33549/physiolres.933269] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Protease-activated receptors (PARs) belong to the G-protein-coupled receptor family, that are expressed in many body tissues especially in different epithelial cells, mast cells and also in neurons and astrocytes. PARs play different physiological roles according to the location of their expression. Increased evidence supports the importance of PARs activation during nociceptive signaling and in the development of chronic pain states. This short review focuses on the role of PAR2 receptors in nociceptive transmission with the emphasis on the modulation at the spinal cord level. PAR2 are cleaved and subsequently activated by endogenous proteases such as tryptase and trypsin. In vivo, peripheral and intrathecal administration of PAR2 agonists induces thermal and mechanical hypersensitivity that is thought to be mediated by PAR2-induced release of pronociceptive neuropeptides and modulation of different receptors. PAR2 activation leads also to sensitization of transient receptor potential channels (TRP) that are crucial for nociceptive signaling and modulation. PAR2 receptors may play an important modulatory role in the development and maintenance of different pathological pain states and could represent a potential target for new analgesic treatments.
Collapse
Affiliation(s)
- P Mrozkova
- Department of Functional Morphology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic.
| | | | | |
Collapse
|
15
|
Hermes SM, Andresen MC, Aicher SA. Localization of TRPV1 and P2X3 in unmyelinated and myelinated vagal afferents in the rat. J Chem Neuroanat 2015; 72:1-7. [PMID: 26706222 DOI: 10.1016/j.jchemneu.2015.12.003] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Revised: 11/18/2015] [Accepted: 12/09/2015] [Indexed: 01/15/2023]
Abstract
The vagus nerve is dominated by afferent fibers that convey sensory information from the viscera to the brain. Most vagal afferents are unmyelinated, slow-conducting C-fibers, while a smaller portion are myelinated, fast-conducting A-fibers. Vagal afferents terminate in the nucleus tractus solitarius (NTS) in the dorsal brainstem and regulate autonomic and respiratory reflexes, as well as ascending pathways throughout the brain. Vagal afferents form glutamatergic excitatory synapses with postsynaptic NTS neurons that are modulated by a variety of channels. The organization of vagal afferents with regard to fiber type and channels is not well understood. In the present study, we used tract tracing methods to identify distinct populations of vagal afferents to determine if key channels are selectively localized to specific groups of afferent fibers. Vagal afferents were labeled with isolectin B4 (IB4) or cholera toxin B (CTb) to detect unmyelinated and myelinated afferents, respectively. We find that TRPV1 channels are preferentially found in unmyelinated vagal afferents identified with IB4, with almost half of all IB4 fibers showing co-localization with TRPV1. These results agree with prior electrophysiological findings. In contrast, we found that the ATP-sensitive channel P2X3 is found in a subset of both myelinated and unmyelinated vagal afferent fibers. Specifically, 18% of IB4 and 23% of CTb afferents contained P2X3. The majority of CTb-ir vagal afferents contained neither channel. Since neither channel was found in all vagal afferents, there are likely further degrees of heterogeneity in the modulation of vagal afferent sensory input to the NTS beyond fiber type.
Collapse
Affiliation(s)
- Sam M Hermes
- Oregon Health & Science University, Department of Physiology and Pharmacology, 3181 Sam Jackson Park Road, Mailcode: L334, Portland, OR 97239-3098, United States
| | - Michael C Andresen
- Oregon Health & Science University, Department of Physiology and Pharmacology, 3181 Sam Jackson Park Road, Mailcode: L334, Portland, OR 97239-3098, United States
| | - Sue A Aicher
- Oregon Health & Science University, Department of Physiology and Pharmacology, 3181 Sam Jackson Park Road, Mailcode: L334, Portland, OR 97239-3098, United States.
| |
Collapse
|
16
|
Zhao P, Lieu T, Barlow N, Sostegni S, Haerteis S, Korbmacher C, Liedtke W, Jimenez-Vargas NN, Vanner SJ, Bunnett NW. Neutrophil Elastase Activates Protease-activated Receptor-2 (PAR2) and Transient Receptor Potential Vanilloid 4 (TRPV4) to Cause Inflammation and Pain. J Biol Chem 2015; 290:13875-87. [PMID: 25878251 DOI: 10.1074/jbc.m115.642736] [Citation(s) in RCA: 133] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Indexed: 12/25/2022] Open
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 β-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.
Collapse
Affiliation(s)
- Peishen Zhao
- From the Monash Institute of Pharmaceutical Sciences and
| | - TinaMarie Lieu
- From the Monash Institute of Pharmaceutical Sciences and
| | | | - Silvia Sostegni
- the Institut für Zelluläre und Molekulare Physiologie, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen 91054, Germany
| | - Silke Haerteis
- the Institut für Zelluläre und Molekulare Physiologie, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen 91054, Germany
| | - Christoph Korbmacher
- the Institut für Zelluläre und Molekulare Physiologie, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen 91054, Germany
| | - Wolfgang Liedtke
- the Department of Neurology, School of Medicine, Duke University, Durham, North Carolina 27710
| | - Nestor N Jimenez-Vargas
- the Gastrointestinal Diseases Research Unit, Division of Gastroenterology, Queen's University, Kingston, Ontario N7L 3N6, Canada, and
| | - Stephen J Vanner
- the Gastrointestinal Diseases Research Unit, Division of Gastroenterology, Queen's University, Kingston, Ontario N7L 3N6, Canada, and
| | - Nigel W Bunnett
- From the Monash Institute of Pharmaceutical Sciences and the ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash University, Parkville 3052, Australia, the Department of Pharmacology, University of Melbourne, Melbourne 3010, Australia
| |
Collapse
|
17
|
Veldhuis NA, Poole DP, Grace M, McIntyre P, Bunnett NW. The G protein-coupled receptor-transient receptor potential channel axis: molecular insights for targeting disorders of sensation and inflammation. Pharmacol Rev 2015; 67:36-73. [PMID: 25361914 DOI: 10.1124/pr.114.009555] [Citation(s) in RCA: 121] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2025] Open
Abstract
Sensory nerves are equipped with receptors and ion channels that allow them to detect and respond to diverse chemical, mechanical, and thermal stimuli. These sensory proteins include G protein-coupled receptors (GPCRs) and transient receptor potential (TRP) ion channels. A subclass of peptidergic sensory nerves express GPCRs and TRP channels that detect noxious, irritant, and inflammatory stimuli. Activation of these nerves triggers protective mechanisms that lead to withdrawal from danger (pain), removal of irritants (itch, cough), and resolution of infection (neurogenic inflammation). The GPCR-TRP axis is central to these mechanisms. Signals that emanate from the GPCR superfamily converge on the small TRP family, leading to channel sensitization and activation, which amplify pain, itch, cough, and neurogenic inflammation. Herein we discuss how GPCRs and TRP channels function independently and synergistically to excite sensory nerves that mediate noxious and irritant responses and inflammation in the skin and the gastrointestinal and respiratory systems. We discuss the signaling mechanisms that underlie the GPCR-TRP axis and evaluate how new information about the structure of GPCRs and TRP channels provides insights into their functional interactions. We propose that a deeper understanding of the GPCR-TRP axis may facilitate the development of more selective and effective therapies to treat dysregulated processes that underlie chronic pain, itch, cough, and inflammation.
Collapse
Affiliation(s)
- Nicholas A Veldhuis
- Monash Institute of Pharmaceutical Sciences, Parkville, Victoria, Australia (N.A.V., D.P.P., N.W.B); Departments of Genetics (N.A.V.), Anatomy and Neuroscience (D.P.P.), and Pharmacology (N.W.B.), The University of Melbourne, Melbourne, Victoria, Australia; School of Medical Sciences and Health Innovations Research Institute, RMIT University, Bundoora, Victoria, Australia (M.G., P.M.); and ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash University, Parkville, Victoria, Australia (N.W.B.)
| | - Daniel P Poole
- Monash Institute of Pharmaceutical Sciences, Parkville, Victoria, Australia (N.A.V., D.P.P., N.W.B); Departments of Genetics (N.A.V.), Anatomy and Neuroscience (D.P.P.), and Pharmacology (N.W.B.), The University of Melbourne, Melbourne, Victoria, Australia; School of Medical Sciences and Health Innovations Research Institute, RMIT University, Bundoora, Victoria, Australia (M.G., P.M.); and ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash University, Parkville, Victoria, Australia (N.W.B.)
| | - Megan Grace
- Monash Institute of Pharmaceutical Sciences, Parkville, Victoria, Australia (N.A.V., D.P.P., N.W.B); Departments of Genetics (N.A.V.), Anatomy and Neuroscience (D.P.P.), and Pharmacology (N.W.B.), The University of Melbourne, Melbourne, Victoria, Australia; School of Medical Sciences and Health Innovations Research Institute, RMIT University, Bundoora, Victoria, Australia (M.G., P.M.); and ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash University, Parkville, Victoria, Australia (N.W.B.)
| | - Peter McIntyre
- Monash Institute of Pharmaceutical Sciences, Parkville, Victoria, Australia (N.A.V., D.P.P., N.W.B); Departments of Genetics (N.A.V.), Anatomy and Neuroscience (D.P.P.), and Pharmacology (N.W.B.), The University of Melbourne, Melbourne, Victoria, Australia; School of Medical Sciences and Health Innovations Research Institute, RMIT University, Bundoora, Victoria, Australia (M.G., P.M.); and ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash University, Parkville, Victoria, Australia (N.W.B.)
| | - Nigel W Bunnett
- Monash Institute of Pharmaceutical Sciences, Parkville, Victoria, Australia (N.A.V., D.P.P., N.W.B); Departments of Genetics (N.A.V.), Anatomy and Neuroscience (D.P.P.), and Pharmacology (N.W.B.), The University of Melbourne, Melbourne, Victoria, Australia; School of Medical Sciences and Health Innovations Research Institute, RMIT University, Bundoora, Victoria, Australia (M.G., P.M.); and ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash University, Parkville, Victoria, Australia (N.W.B.)
| |
Collapse
|
18
|
Takanami K, Sakamoto H, Matsuda KI, Satoh K, Tanida T, Yamada S, Inoue K, Oti T, Sakamoto T, Kawata M. Distribution of gastrin-releasing peptide in the rat trigeminal and spinal somatosensory systems. J Comp Neurol 2014; 522:1858-73. [PMID: 24254931 DOI: 10.1002/cne.23506] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2013] [Revised: 08/30/2013] [Accepted: 11/15/2013] [Indexed: 12/12/2022]
Abstract
Gastrin-releasing peptide (GRP) has recently been identified as an itch-specific neuropeptide in the spinal sensory system in mice, but there are no reports of the expression and distribution of GRP in the trigeminal sensory system in mammals. We characterized and compared GRP-immunoreactive (ir) neurons in the trigeminal ganglion (TG) with those in the rat spinal dorsal root ganglion (DRG). GRP immunoreactivity was expressed in 12% of TG and 6% of DRG neurons and was restricted to the small- and medium-sized type cells. In both the TG and DRG, many GRP-ir neurons also expressed substance P and calcitonin gene-related peptide, but not isolectin B4 . The different proportions of GRP and transient receptor potential vanilloid 1 double-positive neurons in the TG and DRG imply that itch sensations via the TG and DRG pathways are transmitted through distinct mechanisms. The distribution of the axon terminals of GRP-ir primary afferents and their synaptic connectivity with the rat trigeminal sensory nuclei and spinal dorsal horn were investigated by using light and electron microscopic histochemistry. Although GRP-ir fibers were rarely observed in the trigeminal sensory nucleus principalis, oralis, and interpolaris, they were predominant in the superficial layers of the trigeminal sensory nucleus caudalis (Vc), similar to the spinal dorsal horn. Ultrastructural analysis revealed that GRP-ir terminals contained clear microvesicles and large dense-cored vesicles, and formed asymmetric synaptic contacts with a few dendrites in the Vc and spinal dorsal horn. These results suggest that GRP-dependent orofacial and spinal pruriceptive inputs are processed mainly in the superficial laminae of the Vc and spinal dorsal horn.
Collapse
Affiliation(s)
- Keiko Takanami
- Department of Anatomy and Neurobiology, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto, 602-8566, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Suen JY, Cotterell A, Lohman RJ, Lim J, Han A, Yau MK, Liu L, Cooper MA, Vesey DA, Fairlie DP. Pathway-selective antagonism of proteinase activated receptor 2. Br J Pharmacol 2014; 171:4112-24. [PMID: 24821440 DOI: 10.1111/bph.12757] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2014] [Revised: 04/04/2014] [Accepted: 04/30/2014] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND AND PURPOSE Proteinase activated receptor 2 (PAR2) is a GPCR associated with inflammation, metabolism and disease. Clues to understanding how to block PAR2 signalling associated with disease without inhibiting PAR2 activation in normal physiology could be provided by studies of biased signalling. EXPERIMENTAL APPROACH PAR2 ligand GB88 was profiled for PAR2 agonist and antagonist properties by several functional assays associated with intracellular G-protein-coupled signalling in vitro in three cell types and with PAR2-induced rat paw oedema in vivo. KEY RESULTS In HT29 cells, GB88 was a PAR2 antagonist in terms of Ca(2+) mobilization and PKC phosphorylation, but a PAR2 agonist in attenuating forskolin-induced cAMP accumulation, increasing ERK1/2 phosphorylation, RhoA activation, myosin phosphatase phosphorylation and actin filament rearrangement. In CHO-hPAR2 cells, GB88 inhibited Ca(2+) release, but activated G(i/o) and increased ERK1/2 phosphorylation. In human kidney tubule cells, GB88 inhibited cytokine secretion (IL6, IL8, GM-CSF, TNF-α) mediated by PAR2. A rat paw oedema induced by PAR2 agonists was also inhibited by orally administered GB88 and compared with effects of locally administered inhibitors of G-protein coupled pathways. CONCLUSIONS AND IMPLICATIONS GB88 is a biased antagonist of PAR2 that selectively inhibits PAR2/G(q/11)/Ca(2+)/PKC signalling, leading to anti-inflammatory activity in vivo, while being an agonist in activating three other PAR2-activated pathways (cAMP, ERK, Rho) in human cells. These findings highlight opportunities to design drugs to block specific PAR2-linked signalling pathways in disease, without blocking beneficial PAR2 signalling in normal physiology, and to dissect PAR2-associated mechanisms of disease in vivo.
Collapse
Affiliation(s)
- J Y Suen
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Qld, Australia
| | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Hegarty DM, Hermes SM, Largent-Milnes TM, Aicher SA. Capsaicin-responsive corneal afferents do not contain TRPV1 at their central terminals in trigeminal nucleus caudalis in rats. J Chem Neuroanat 2014; 61-62:1-12. [PMID: 24996127 DOI: 10.1016/j.jchemneu.2014.06.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2014] [Revised: 06/23/2014] [Accepted: 06/23/2014] [Indexed: 10/25/2022]
Abstract
We examined the substrates for ocular nociception in adult male Sprague-Dawley rats. Capsaicin application to the ocular surface in awake rats evoked nocifensive responses and suppressed spontaneous grooming responses. Thus, peripheral capsaicin was able to activate the central pathways encoding ocular nociception. Our capsaicin stimulus evoked c-Fos expression in a select population of neurons within rostral trigeminal nucleus caudalis in anesthetized rats. These activated neurons also received direct contacts from corneal afferent fibers traced with cholera toxin B from the corneal surface. However, the central terminals of the corneal afferents that contacted capsaicin-activated trigeminal neurons did not contain TRPV1. To determine if TRPV1 expression had been altered by capsaicin stimulation, we examined TRPV1 content of corneal afferents in animals that did not receive capsaicin stimulation. These studies confirmed that while TRPV1 was present in 30% of CTb-labeled corneal afferent neurons within the trigeminal ganglion, TRPV1 was only detected in 2% of the central terminals of these corneal afferents within the trigeminal nucleus caudalis. Other TRP channels were also present in low proportions of central corneal afferent terminals in unstimulated animals (TRPM8, 2%; TRPA1, 10%). These findings indicate that a pathway from the cornea to rostral trigeminal nucleus caudalis is involved in corneal nociceptive transmission, but that central TRP channel expression is unrelated to the type of stimulus transduced by the peripheral nociceptive endings.
Collapse
Affiliation(s)
- Deborah M Hegarty
- Department of Physiology and Pharmacology, Oregon Health & Science University, Mail code: L334, 3181 Sam Jackson Park Road, Portland, OR 97239-3098, United States.
| | - Sam M Hermes
- Department of Physiology and Pharmacology, Oregon Health & Science University, Mail code: L334, 3181 Sam Jackson Park Road, Portland, OR 97239-3098, United States.
| | - Tally M Largent-Milnes
- Department of Physiology and Pharmacology, Oregon Health & Science University, Mail code: L334, 3181 Sam Jackson Park Road, Portland, OR 97239-3098, United States.
| | - Sue A Aicher
- Department of Physiology and Pharmacology, Oregon Health & Science University, Mail code: L334, 3181 Sam Jackson Park Road, Portland, OR 97239-3098, United States.
| |
Collapse
|
21
|
Poole DP, Lee M, Tso P, Bunnett NW, Yo SJ, Lieu T, Shiu A, Wang JC, Nomura DK, Aponte GW. Feeding-dependent activation of enteric cells and sensory neurons by lymphatic fluid: evidence for a neurolymphocrine system. Am J Physiol Gastrointest Liver Physiol 2014; 306:G686-98. [PMID: 24578341 PMCID: PMC3989702 DOI: 10.1152/ajpgi.00433.2013] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
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.
Collapse
Affiliation(s)
- Daniel P. Poole
- 1Monash Institute of Pharmaceutical Sciences, Parkville, Victoria, Australia; ,2Department of Anatomy and Neuroscience, The University of Melbourne, Parkville, Victoria, Australia;
| | - Mike Lee
- 4Department of Pathology, Stanford University, Palo Alto, California;
| | - Patrick Tso
- 6Department of Pathobiology and Molecular Medicine, University of Cincinnati, Reading, Ohio
| | - Nigel W. Bunnett
- 1Monash Institute of Pharmaceutical Sciences, Parkville, Victoria, Australia; ,3Department of Pharmacology, The University of Melbourne, Parkville, Victoria, Australia;
| | - Sek Jin Yo
- 5Department of Nutritional Sciences and Toxicology, University of California at Berkeley, Berkeley, California;
| | - TinaMarie Lieu
- 1Monash Institute of Pharmaceutical Sciences, Parkville, Victoria, Australia;
| | - Amy Shiu
- 5Department of Nutritional Sciences and Toxicology, University of California at Berkeley, Berkeley, California;
| | - Jen-Chywan Wang
- 5Department of Nutritional Sciences and Toxicology, University of California at Berkeley, Berkeley, California;
| | - Daniel K. Nomura
- 5Department of Nutritional Sciences and Toxicology, University of California at Berkeley, Berkeley, California;
| | - Gregory W. Aponte
- 5Department of Nutritional Sciences and Toxicology, University of California at Berkeley, Berkeley, California;
| |
Collapse
|
22
|
Bao Y, Hou W, Hua B. Protease-activated receptor 2 signalling pathways: a role in pain processing. Expert Opin Ther Targets 2013; 18:15-27. [PMID: 24147628 DOI: 10.1517/14728222.2014.844792] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
INTRODUCTION Pain is a complex biological phenomenon that includes intricate neurophysiological, behavioural, psychosocial and affective components. Despite decades of pain research, many patients continue suffering from chronic pain that may be refractory to current medical regimens. Accumulating evidence has indicated an important role of protease-activated receptor 2 (PAR2) in the pathogenesis of pain, including inflammation, neuropathic and cancer pain. AREAS COVERED In this review, the role of the PAR2 signalling pathway in pain processes, basic mechanism of PAR2 activation and expression of PAR2 in the nervous system is covered. Furthermore, intracellular signalling pathways that are activated by PAR2 are also described. EXPERT OPINION The role of PAR2 in pain processing is becoming increasingly clear, and although causal implication remains to be established, PAR2 activation has been observed in several disease model systems. Since PAR2 is activated after nerve injury as well as by trypsin and related serine proteases, and PAR2 plays an important role in pain development and maintenance, exploring PAR2 and its corresponding signalling pathways will provide unfathomable knowledge in understanding the molecular basis of pain. This will also help to identify new targets for pharmacological intervention; however, in the context of potential PAR2-directed therapies, several aspects should be clarified.
Collapse
Affiliation(s)
- Yanju Bao
- Guang'anmen Hospital, China Academy of Chinese Medical Science, Department of Oncology , Beixiange 5, Xicheng District, Beijing 100053 , China +86 10 88001221 ; +86 10 88001430 ; ; ;
| | | | | |
Collapse
|
23
|
Xu P, Rosen KM, Hedstrom K, Rey O, Guha S, Hart C, Corfas G. Nerve injury induces glial cell line-derived neurotrophic factor (GDNF) expression in Schwann cells through purinergic signaling and the PKC-PKD pathway. Glia 2013; 61:1029-40. [PMID: 23553603 DOI: 10.1002/glia.22491] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2012] [Accepted: 02/06/2013] [Indexed: 12/22/2022]
Abstract
Upon peripheral nerve injury, specific molecular events, including increases in the expression of selected neurotrophic factors, are initiated to prepare the tissue for regeneration. However, the mechanisms underlying these events and the nature of the cells involved are poorly understood. We used the injury-induced upregulation of glial cell-derived neurotrophic factor (GDNF) expression as a tool to gain insights into these processes. We found that both myelinating and nonmyelinating Schwann cells are responsible for the dramatic increase in GDNF expression after injury. We also demonstrate that the GDNF upregulation is mediated by a signaling cascade involving activation of Schwann cell purinergic receptors, followed by protein kinase C signaling which activates protein kinase D (PKD), which leads to increased GDNF transcription. Given the potent effects of GDNF on survival and repair of injured peripheral neurons, we propose that targeting these pathways may yield therapeutic tools to treat peripheral nerve injury and neuropathies.
Collapse
Affiliation(s)
- Pin Xu
- F.M. Kirby Neurobiology Center, Children's Hospital Boston, Boston, MA, USA
| | | | | | | | | | | | | |
Collapse
|
24
|
Sugimoto Y, Kojima Y, Inayoshi A, Inoue K, Miura-Kusaka H, Mori K, Saku O, Ishida H, Atsumi E, Nakasato Y, Shirakura S, Toki SI, Shinoda K, Suzuki N. K-685, a TRPV1 Antagonist, Blocks PKC-Sensitized TRPV1 Activation and Improves the Inflammatory Pain in a Rat Complete Freund’s Adjuvant Model. J Pharmacol Sci 2013; 123:256-66. [DOI: 10.1254/jphs.13088fp] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
|
25
|
Guo J, Clausen DM, Beumer JH, Parise RA, Egorin MJ, Bravo-Altamirano K, Wipf P, Sharlow ER, Wang QJ, Eiseman JL. In vitro cytotoxicity, pharmacokinetics, tissue distribution, and metabolism of small-molecule protein kinase D inhibitors, kb-NB142-70 and kb-NB165-09, in mice bearing human cancer xenografts. Cancer Chemother Pharmacol 2012; 71:331-44. [PMID: 23108699 DOI: 10.1007/s00280-012-2010-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2012] [Accepted: 10/15/2012] [Indexed: 12/11/2022]
Abstract
PURPOSE Protein kinase D (PKD) mediates diverse biological responses including cell growth and survival. Therefore, PKD inhibitors may have therapeutic potential. We evaluated the in vitro cytotoxicity of two PKD inhibitors, kb-NB142-70 and its methoxy analogue, kb-NB165-09, and examined their in vivo efficacy and pharmacokinetics. METHODS The in vitro cytotoxicities of kb-NB142-70 and kb-NB165-09 were evaluated by MTT assay against PC-3, androgen-independent prostate cancer cells, and CFPAC-1 and PANC-1, pancreatic cancer cells. Efficacy studies were conducted in mice bearing either PC-3 or CPFAC-1 xenografts. Tumor-bearing mice were euthanized between 5 and 1,440 min after iv dosing, and plasma and tissue concentrations were measured by HPLC-UV. Metabolites were characterized by LC-MS/MS. RESULTS kb-NB142-70 and kb-NB165-09 inhibited cellular growth in the low-mid μM range. The compounds were inactive when administered to tumor-bearing mice. In mice treated with kb-NB142-70, the plasma C (max) was 36.9 nmol/mL, and the PC-3 tumor C (max) was 11.8 nmol/g. In mice dosed with kb-NB165-09, the plasma C (max) was 61.9 nmol/mL, while the PANC-1 tumor C (max) was 8.0 nmol/g. The plasma half-lives of kb-NB142-70 and kb-NB165-09 were 6 and 14 min, respectively. Both compounds underwent oxidation and glucuronidation. CONCLUSIONS kb-NB142-70 and kb-NB165-09 were rapidly metabolized, and concentrations in tumor were lower than those required for in vitro cytotoxicity. Replacement of the phenolic hydroxyl group with a methoxy group increased the plasma half-life of kb-NB165-09 2.3-fold over that of kb-NB142-70. Rapid metabolism in mice suggests that next-generation compounds will require further structural modifications to increase potency and/or metabolic stability.
Collapse
Affiliation(s)
- Jianxia Guo
- Molecular Therapeutics and Drug Discovery, The University of Pittsburgh Cancer Institute, Hillman Cancer Center, 5117 Centre Ave, Room G27b, Pittsburgh, PA 15213, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Amadesi S, Reni C, Katare R, Meloni M, Oikawa A, Beltrami AP, Avolio E, Cesselli D, Fortunato O, Spinetti G, Ascione R, Cangiano E, Valgimigli M, Hunt SP, Emanueli C, Madeddu P. Role for substance p-based nociceptive signaling in progenitor cell activation and angiogenesis during ischemia in mice and in human subjects. Circulation 2012; 125:1774-86, S1-19. [PMID: 22392530 DOI: 10.1161/circulationaha.111.089763] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
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.
Collapse
Affiliation(s)
- Silvia Amadesi
- Laboratories of Experimental Cardiovascular Medicine, Bristol Heart Institute, University of Bristol, Bristol, United Kingdom
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Palazzo E, Luongo L, de Novellis V, Rossi F, Marabese I, Maione S. Transient receptor potential vanilloid type 1 and pain development. Curr Opin Pharmacol 2012; 12:9-17. [DOI: 10.1016/j.coph.2011.10.022] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2011] [Revised: 10/27/2011] [Accepted: 10/28/2011] [Indexed: 02/06/2023]
|
28
|
Proteinase-activated receptors 1 and 2 regulate invasive behavior of human melanoma cells via activation of protein kinase D1. J Invest Dermatol 2011; 132:375-84. [PMID: 21993564 DOI: 10.1038/jid.2011.314] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Recent studies have indicated an important role of proteinases and proteinase-activated receptors (PARs) in tumorigenesis. Although a role for PARs has been described in various skin tumors including melanoma, the underlying cellular mechanisms have not been understood. Recent studies have suggested PAR(1) as a regulator of melanoma cell growth and metastasis by affecting angiogenic and invasive factors. Moreover, changes in the expression patterns of PAR(1) and PAR(2) correlate with skin cancer progression, and PAR(1) is overexpressed in melanoma. Therefore, we sought to elucidate the putative role of PAR(1)- and PAR(2)-mediated signal transduction pathways during melanoma progression. Activation of both PAR(1) and PAR(2) led to rapid phosphorylation of protein kinase D1 (PKD1) in cultured WM9 melanoma cells. PKD1 is known to be involved in cell migration, integrin regulation, and intracellular vesicle transport. Downregulation of PKD1 by siRNA resulted in diminished proliferation, decreased αvβ3 integrin regulation, and secretion of pro-angiogenic chemokine IL-8 in WM9 cells. In conclusion, our results show that PAR(1) and PAR(2) are involved in WM9 cell proliferation and secretion of IL-8 by activation of PKD1. Inactivation of the PKD1 pathway may be beneficial for the inhibition of PAR-induced melanoma proliferation and for maintenance of the inflammatory tumor environment.
Collapse
|
29
|
Chung MK, Jung SJ, Oh SB. Role of TRP channels in pain sensation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2011; 704:615-36. [PMID: 21290319 DOI: 10.1007/978-94-007-0265-3_33] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
It is crucial for a living organism to recognize and discern potentially harmful noxious stimuli from innocuous stimuli to avoid hazards in the environment. However, unnecessary or exaggerated nociception is at best unpleasant and often compromises the quality of life. In order to lessen the intensity of nociception or eliminate the pathological pain, it is important to understand the nature of nociception and the mechanisms of hyperalgesia or allodynia. Transient receptor potential (TRP) channels play central roles in nociception under physiological and pathological conditions including inflammation and neuropathy. In this chapter, we will highlight the enormous progress in understanding the role of TRP channels in nociception. We will mainly focus on two TRP channels (TRPV1 and TRPA1) that have been particularly implicated in transducing signals associated with pain sensation, and briefly discuss the role of TRPM8, TRPV3 and TRPV4. We will stress debatable issues that needed to be resolved and provide perspectives for the future studies.
Collapse
Affiliation(s)
- Man-Kyo Chung
- Department of Neural and Pain Sciences, University of Maryland Dental School, Baltimore, MD, USA.
| | | | | |
Collapse
|
30
|
Kojundzic SL, Puljak L, Hogan Q, Sapunar D. Depression of Ca(2+)/calmodulin-dependent protein kinase II in dorsal root ganglion neurons after spinal nerve ligation. J Comp Neurol 2010; 518:64-74. [PMID: 19882720 DOI: 10.1002/cne.22209] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The enzyme calcium/calmodulin-dependent protein kinase II (CaMKII) is associated with memory and its alpha isoform is critical for development of activity-induced synaptic changes. Therefore, we hypothesized that CaMKII is involved in altered function of dorsal root ganglion (DRG) neurons after neuronal injury. To test this hypothesis, Sprague-Dawley rats were made hyperalgesic by L5 and L6 spinal nerve ligation (SNL), and changes in total phosphorylated and unphosphorylated CaMKII (tCaMKII) and phosphorylated form of its alpha isoform (pCaMKIIalpha) were analyzed using immunochemistry in different subpopulations of DRG. SNL did not induce any changes in tCaMKII between experimental groups, while the overall percentage of pCaMKIIalpha-positive neurons in injured L5 DRG SNL (24.8%) decreased significantly when compared to control (41.7%). SNL did not change the percentage of pCaMKIIalpha/N52 colabeled neurons but decreased the percentage of N52-negative nonmyelinated neurons that expressed pCaMKIIalpha from 27% in control animals to 11% after axotomy. We also observed a significant decrease in the percentage of small nonpeptidergic neurons labeled with IB4 (37.6% in control vs. 4.0% in L5 SNL DRG), as well as a decrease in the percentage of pCaMKIIalpha/IB4 colabeled neurons in injured L5 DRGs (27% in control vs. 1% in L5 DRG of SNL group). Our results show that reduction in pCaMKIIalpha levels following peripheral injury is due to the loss of IB4-positive neurons. These results indicate that diminished afferent activity after axotomy may lead to decreased phosphorylation of CaMKIIalpha.
Collapse
Affiliation(s)
- Sanja Lovric Kojundzic
- Department of Anatomy, Histology and Embryology, University of Split School of Medicine, 21000 Split, Croatia
| | | | | | | |
Collapse
|
31
|
Cattaruzza F, Spreadbury I, Miranda-Morales M, Grady EF, Vanner S, Bunnett NW. Transient receptor potential ankyrin-1 has a major role in mediating visceral pain in mice. Am J Physiol Gastrointest Liver Physiol 2010; 298:G81-91. [PMID: 19875705 PMCID: PMC2806099 DOI: 10.1152/ajpgi.00221.2009] [Citation(s) in RCA: 99] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The excitatory ion channel transient receptor potential ankyrin-1 (TRPA1) is prominently expressed by primary afferent neurons and is a mediator of inflammatory pain. Inflammatory agents can directly activate [e.g., hydroxynonenal (HNE), prostaglandin metabolites] or indirectly sensitize [e.g., agonists of protease-activated receptor (PAR(2))] TRPA1 to induce somatic pain and hyperalgesia. However, the contribution of TRPA1 to visceral pain is unknown. We investigated the role of TRPA1 in visceral hyperalgesia by measuring abdominal visceromotor responses (VMR) to colorectal distention (CRD) after intracolonic administration of TRPA1 agonists [mustard oil (MO), HNE], sensitizing agents [PAR(2) activating peptide (PAR(2)-AP)], and the inflammatory agent trinitrobenzene sulfonic acid (TNBS) in trpa1(+/+) and trpa1(-/-) mice. Sensory neurons innervating the colon, identified by retrograde tracing, coexpressed immunoreactive TRPA1, calcitonin gene-related peptide, and substance P, expressed TRPA1 mRNA and responded to MO with depolarizing currents. Intracolonic MO and HNE increased VMR to CRD and induced immunoreactive c-fos in spinal neurons in trpa1+/+ but not in trpa1(-/-) mice. Intracolonic PAR(2)-AP induced mechanical hyperalgesia in trpa1+/+ but not in trpa1(-/-) mice. TNBS-induced colitis increased in VMR to CRD and induced c-fos in spinal neurons in trpa1(+/+) but not in trpa1(-/-) mice. Thus TRPA1 is expressed by colonic primary afferent neurons. Direct activation of TRPA1 causes visceral hyperalgesia, and TRPA1 mediates PAR(2)-induced hyperalgesia. TRPA1 deletion markedly reduces colitis-induced mechanical hyperalgesia in the colon. Our results suggest that TRPA1 has a major role in visceral nociception and may be a therapeutic target for colonic inflammatory pain.
Collapse
Affiliation(s)
| | - Ian Spreadbury
- 3Gastrointestinal Diseases Research Unit, Division of Gastroenterology, Queen's University, Kingston, Ontario, Canada
| | - Marcela Miranda-Morales
- 3Gastrointestinal Diseases Research Unit, Division of Gastroenterology, Queen's University, Kingston, Ontario, Canada
| | | | - Stephen Vanner
- 3Gastrointestinal Diseases Research Unit, Division of Gastroenterology, Queen's University, Kingston, Ontario, Canada
| | - Nigel W. Bunnett
- Departments of 1 Surgery and ,2Physiology, University of California, San Francisco, California;
| |
Collapse
|