1
|
Brum EDS, Fialho MFP, Becker G, Nogueira CW, Oliveira SM. Involvement of peripheral mast cells in a fibromyalgia model in mice. Eur J Pharmacol 2024; 967:176385. [PMID: 38311276 DOI: 10.1016/j.ejphar.2024.176385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 01/19/2024] [Accepted: 02/01/2024] [Indexed: 02/10/2024]
Abstract
Fibromyalgia is a painful disorder of unknown aetiology that presents activation and recruitment of innate immune cells, including mast cells. Efforts have been made to understand its pathogenesis to manage it better. Thus, we explored the involvement of peripheral mast cells in an experimental model of fibromyalgia induced by reserpine. Reserpine (1 mg/kg) was subcutaneously (s.c.) injected once daily in the back of male Swiss mice for three consecutive days. We analysed mechanical and cold allodynia, muscle fatigue and number of mast cell in plantar tissue. The fibromyalgia induction produced mast cell infiltration (i.e., mastocytosis) in the mice's plantar tissue. The depletion of mast cell mediators with the compound 48/80 (0.5-4 mg/kg, intraperitoneal (i.p.)) or the mast cell membrane stabilizer ketotifen fumarate (10 mg/kg, oral route (p.o.) widely (80-90 %) and extensively (from 1 up to 10 days) prevented reserpine-induced mechanical and cold allodynia and muscle fatigue. Compound 48/80 also prevented the reserpine-induced mastocytosis. Finally, we demonstrated that PAR-2, 5-HT2A, 5-HT3, H1, NK1 and MrgprB2 receptors, expressed in neuronal or mast cells, seem crucial to mediate fibromyalgia-related cardinal symptoms since antagonists or inhibitors of these receptors (gabexate (10 mg/kg, s.c.), ENMD-1068 (10 mg/kg, i.p.), ketanserin (1 mg/kg, i.p.), ondansetron (1 mg/kg, p.o.), promethazine (1 mg/kg, i.p.), and L733,060 (5 mg/kg, s.c.), respectively) transiently reversed the reserpine-induced allodynia and fatigue. The results indicate that mast cells mediate painful and fatigue behaviours in this fibromyalgia model, representing potential therapy targets to treat fibromyalgia syndrome.
Collapse
Affiliation(s)
- Evelyne da Silva Brum
- Graduate Program in Biological Sciences: Toxicological Biochemistry, Centre of Natural and Exact Sciences, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Maria Fernanda Pessano Fialho
- Graduate Program in Biological Sciences: Toxicological Biochemistry, Centre of Natural and Exact Sciences, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Gabriela Becker
- Graduate Program in Biological Sciences: Toxicological Biochemistry, Centre of Natural and Exact Sciences, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Cristina Wayne Nogueira
- Graduate Program in Biological Sciences: Toxicological Biochemistry, Centre of Natural and Exact Sciences, Federal University of Santa Maria, Santa Maria, RS, Brazil; Department of Biochemistry and Molecular Biology, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Sara Marchesan Oliveira
- Graduate Program in Biological Sciences: Toxicological Biochemistry, Centre of Natural and Exact Sciences, Federal University of Santa Maria, Santa Maria, RS, Brazil; Department of Biochemistry and Molecular Biology, Federal University of Santa Maria, Santa Maria, RS, Brazil.
| |
Collapse
|
2
|
Rondeau LE, Da Luz BB, Santiago A, Bermudez-Brito M, Hann A, De Palma G, Jury J, Wang X, Verdu EF, Galipeau HJ, Rolland C, Deraison C, Ruf W, Bercik P, Vergnolle N, Caminero A. Proteolytic bacteria expansion during colitis amplifies inflammation through cleavage of the external domain of PAR2. Gut Microbes 2024; 16:2387857. [PMID: 39171684 PMCID: PMC11346554 DOI: 10.1080/19490976.2024.2387857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 07/03/2024] [Accepted: 07/23/2024] [Indexed: 08/23/2024] Open
Abstract
Imbalances in proteolytic activity have been linked to the development of inflammatory bowel diseases (IBD) and experimental colitis. Proteases in the intestine play important roles in maintaining homeostasis, but exposure of mucosal tissues to excess proteolytic activity can promote pathology through protease-activated receptors (PARs). Previous research implicates microbial proteases in IBD, but the underlying pathways and specific interactions between microbes and PARs remain unclear. In this study, we investigated the role of microbial proteolytic activation of the external domain of PAR2 in intestinal injury using mice expressing PAR2 with a mutated N-terminal external domain that is resistant to canonical activation by proteolytic cleavage. Our findings demonstrate the key role of proteolytic cleavage of the PAR2 external domain in promoting intestinal permeability and inflammation during colitis. In wild-type mice expressing protease-sensitive PAR2, excessive inflammation leads to the expansion of bacterial taxa that cleave the external domain of PAR2, exacerbating colitis severity. In contrast, mice expressing mutated protease-resistant PAR2 exhibit attenuated colitis severity and do not experience the same proteolytic bacterial expansion. Colonization of wild-type mice with proteolytic PAR2-activating Enterococcus and Staphylococcus worsens colitis severity. Our study identifies a previously unknown interaction between proteolytic bacterial communities, which are shaped by inflammation, and the external domain of PAR2 in colitis. The findings should encourage new therapeutic developments for IBD by targeting excessive PAR2 cleavage by bacterial proteases.
Collapse
Affiliation(s)
- Liam Emile Rondeau
- Department of Medicine, Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON, Canada
| | - Bruna Barbosa Da Luz
- Department of Medicine, Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON, Canada
| | - Alba Santiago
- Department of Medicine, Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON, Canada
| | - Miriam Bermudez-Brito
- Department of Medicine, Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON, Canada
| | - Amber Hann
- Department of Medicine, Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON, Canada
| | - Giada De Palma
- Department of Medicine, Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON, Canada
| | - Jennifer Jury
- Department of Medicine, Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON, Canada
| | - Xuanyu Wang
- Department of Medicine, Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON, Canada
| | - Elena Francisca Verdu
- Department of Medicine, Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON, Canada
| | - Heather Jean Galipeau
- Department of Medicine, Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON, Canada
| | - Corinne Rolland
- IRSD, Université de Toulouse, INSERM, INRA, ENVT, UPS, Toulouse, France
| | - Celine Deraison
- IRSD, Université de Toulouse, INSERM, INRA, ENVT, UPS, Toulouse, France
| | - Wolfram Ruf
- Center for Thrombosis and Hemostasis, Johannes Gutenberg University Medical Center, Mainz, Germany
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA
| | - Premysl Bercik
- Department of Medicine, Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON, Canada
| | | | - Alberto Caminero
- Department of Medicine, Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
3
|
Barrett TJ. Targeting PAR4 to Reduce Atherosclerosis. Arterioscler Thromb Vasc Biol 2023; 43:2179-2182. [PMID: 37767705 PMCID: PMC10772896 DOI: 10.1161/atvbaha.123.320046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/29/2023]
Affiliation(s)
- Tessa J Barrett
- New York University Grossman School of Medicine, Department of Medicine, Department of Pathology, New York
| |
Collapse
|
4
|
Bagang N, Gupta K, Singh G, Kanuri SH, Mehan S. Protease-activated receptors in kidney diseases: A comprehensive review of pathological roles, therapeutic outcomes and challenges. Chem Biol Interact 2023; 377:110470. [PMID: 37011708 DOI: 10.1016/j.cbi.2023.110470] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 03/21/2023] [Accepted: 03/31/2023] [Indexed: 04/03/2023]
Abstract
Studies have demonstrated that protease-activated receptors (PARs) with four subtypes (PAR1-4) are mainly expressed in the renal epithelial, endothelial, and podocyte cells. Some endogenous and urinary proteases, namely thrombin, trypsin, urokinase, and kallikrein released during diseased conditions, are responsible for activating different subtypes of PARs. Each PAR receptor subtype is involved in kidney disease of distinct aetiology. PAR1 and PAR2 have shown differential therapeutic outcomes in rodent models of type-1 and type-2 diabetic kidney diseases due to the distinct etiological basis of each disease type, however such findings need to be confirmed in other diabetic renal injury models. PAR1 and PAR2 blockers have been observed to abolish drug-induced nephrotoxicity in rodents by suppressing tubular inflammation and fibrosis and preventing mitochondrial dysfunction. Notably, PAR2 inhibition improved autophagy and prevented fibrosis, inflammation, and remodeling in the urethral obstruction model. Only the PAR1/4 subtypes have emerged as a therapeutic target for treating experimentally induced nephrotic syndrome, where their respective antibodies attenuated the podocyte apoptosis induced upon thrombin activation. Strikingly PAR2 and PAR4 subtypes involvement has been tested in sepsis-induced acute kidney injury (AKI) and renal ischemia-reperfusion injury models. Thus, more studies are required to delineate the role of other subtypes in the sepsis-AKI model. Evidence suggests that PARs regulate oxidative, inflammatory stress, immune cell activation, fibrosis, autophagic flux, and apoptosis during kidney diseases.
Collapse
|
5
|
Berger M, Guiraud L, Dumas A, Sagnat D, Payros G, Rolland C, Vergnolle N, Deraison C, Cenac N, Racaud-Sultan C. Prenatal stress induces changes in PAR2- and M3-dependent regulation of colon primitive cells. Am J Physiol Gastrointest Liver Physiol 2022; 323:G609-G626. [PMID: 36283083 PMCID: PMC9722261 DOI: 10.1152/ajpgi.00061.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Prenatal stress is associated with a high risk of developing adult intestinal pathologies, such as irritable bowel syndrome, chronic inflammation, and cancer. Although epithelial stem cells and progenitors have been implicated in intestinal pathophysiology, how prenatal stress could impact their functions is still unknown. We have investigated the proliferative and differentiation capacities of primitive cells using epithelial crypts isolated from colons of adult male and female mice whose mothers have been stressed during late gestation. Our results show that stem cell/progenitor proliferation and differentiation in vitro are negatively impacted by prenatal stress in male progeny. This is promoted by a reinforcement of the negative proliferative/differentiation control by the protease-activated receptor 2 (PAR2) and the muscarinic receptor 3 (M3), two G protein-coupled receptors present in the crypt. Conversely, prenatal stress does not change in vitro proliferation of colon primitive cells in female progeny. Importantly, this maintenance is associated with a functional switch in the M3 negative control of colonoid growth, becoming proliferative after prenatal stress. In addition, the proliferative role of PAR2 specific to females is maintained under prenatal stress, even though PAR2-targeted stress signals Dusp6 and activated GSK3β are increased, reaching the levels of males. An epithelial serine protease could play a critical role in the activation of the survival kinase GSK3β in colonoids from prenatally stressed female progeny. Altogether, our results show that following prenatal stress, colon primitive cells cope with stress through sexually dimorphic mechanisms that could pave the way to dysregulated crypt regeneration and intestinal pathologies.NEW & NOTEWORTHY Primitive cells isolated from mouse colon following prenatal stress and exposed to additional stress conditions such as in vitro culture, present sexually dimorphic mechanisms based on PAR2- and M3-dependent regulation of proliferation and differentiation. Whereas prenatal stress reinforces the physiological negative control exerted by PAR2 and M3 in crypts from males, in females, it induces a switch in M3- and PAR2-dependent regulation leading to a resistant and proliferative phenotype of progenitor.
Collapse
Affiliation(s)
- Mathieu Berger
- 1Institut de Recherche en Santé Digestive, INSERM U1220, Institut
National de Recherche pour l’Agriculture, l’Alimentation et
l’Environnement, Ecole Nationale Vétérinaire de Toulouse, University of Toulouse, Toulouse, France
| | - Laura Guiraud
- 1Institut de Recherche en Santé Digestive, INSERM U1220, Institut
National de Recherche pour l’Agriculture, l’Alimentation et
l’Environnement, Ecole Nationale Vétérinaire de Toulouse, University of Toulouse, Toulouse, France
| | - Alexia Dumas
- 1Institut de Recherche en Santé Digestive, INSERM U1220, Institut
National de Recherche pour l’Agriculture, l’Alimentation et
l’Environnement, Ecole Nationale Vétérinaire de Toulouse, University of Toulouse, Toulouse, France
| | - David Sagnat
- 1Institut de Recherche en Santé Digestive, INSERM U1220, Institut
National de Recherche pour l’Agriculture, l’Alimentation et
l’Environnement, Ecole Nationale Vétérinaire de Toulouse, University of Toulouse, Toulouse, France
| | - Gaëlle Payros
- 1Institut de Recherche en Santé Digestive, INSERM U1220, Institut
National de Recherche pour l’Agriculture, l’Alimentation et
l’Environnement, Ecole Nationale Vétérinaire de Toulouse, University of Toulouse, Toulouse, France
| | - Corinne Rolland
- 1Institut de Recherche en Santé Digestive, INSERM U1220, Institut
National de Recherche pour l’Agriculture, l’Alimentation et
l’Environnement, Ecole Nationale Vétérinaire de Toulouse, University of Toulouse, Toulouse, France
| | - Nathalie Vergnolle
- 1Institut de Recherche en Santé Digestive, INSERM U1220, Institut
National de Recherche pour l’Agriculture, l’Alimentation et
l’Environnement, Ecole Nationale Vétérinaire de Toulouse, University of Toulouse, Toulouse, France,2Department of Physiology and Pharmacology, Cumming School of
Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Céline Deraison
- 1Institut de Recherche en Santé Digestive, INSERM U1220, Institut
National de Recherche pour l’Agriculture, l’Alimentation et
l’Environnement, Ecole Nationale Vétérinaire de Toulouse, University of Toulouse, Toulouse, France
| | - Nicolas Cenac
- 1Institut de Recherche en Santé Digestive, INSERM U1220, Institut
National de Recherche pour l’Agriculture, l’Alimentation et
l’Environnement, Ecole Nationale Vétérinaire de Toulouse, University of Toulouse, Toulouse, France
| | - Claire Racaud-Sultan
- 1Institut de Recherche en Santé Digestive, INSERM U1220, Institut
National de Recherche pour l’Agriculture, l’Alimentation et
l’Environnement, Ecole Nationale Vétérinaire de Toulouse, University of Toulouse, Toulouse, France
| |
Collapse
|
6
|
Electroacupuncture Regulates TRPV1 through PAR2/PKC Pathway to Alleviate Visceral Hypersensitivity in FD Rats. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:1975228. [PMID: 34880917 PMCID: PMC8648456 DOI: 10.1155/2021/1975228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 08/17/2021] [Accepted: 10/28/2021] [Indexed: 11/22/2022]
Abstract
Visceral hypersensitivity (VH) is the predominant pathogenesis of functional dyspepsia (FD). Duodenal hypersensitivity along with nausea further reduces the comfort level in gastric balloon dilatation and inhibits gastric receptive relaxation. The potential mechanism behind electroacupuncture- (EA-) mediated alleviation of VH has not been elucidated. In an FD rat model with tail clamping stress, iodine acetamide (IA) induced VH. The rats were treated with EA with or without PAR2 antagonist FSLLRY-NH2, and the body weight, gastric sensitivity, compliance, and gastrointestinal motility were determined. Mast cells and activated degranulation were stained with toluidine blue (TB) staining and visualized under a transmission electron microscope (TEM). Immunofluorescence was used to detect the expression of PAR2, PKC, and TRPV1 in the duodenum and dorsal root ganglion (DRG) and that of CGRP, SP in DRG, and c-fos in the spinal cord. EA alone and EA + antagonist enhanced the gastrointestinal motility but diminished the expression of TRPV1, CGRP, SP, and c-fos-downstream of PAR2/PKC pathway and alleviated VH in FD rats. However, there was no obvious superposition effect between the antagonists and EA + antagonists. The effect of EA alone was better than that of antagonists and EA + antagonists 2 alone. EA-induced amelioration of VH in FD rats was mediated by TRPV1 regulation through PAR2/PKC pathway. This protective mechanism involved several pathways and included several targets.
Collapse
|
7
|
Guo ZB, Tang L, Wang LP, Wu HH, Huang CL, Zhan MX, Shi ZM, Yang CL, Chen GZ, Zou YQ, Yang F, Wu XZ. The analgesic effects of ulinastatin either as a single agent or in combination with sufentanil: A novel therapeutic potential for postoperative pain. Eur J Pharmacol 2021; 907:174267. [PMID: 34146590 DOI: 10.1016/j.ejphar.2021.174267] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Revised: 06/07/2021] [Accepted: 06/14/2021] [Indexed: 01/11/2023]
Abstract
Ulinastatin is a broad-spectrum protease inhibitor widely used for the treatment of various inflammation-related diseases owing to its recognized excellent anti-inflammatory and cytoprotective properties. However, whether ulinastatin can relieve postoperative pain remains unclear. In this study, we evaluated the analgesic effects of ulinastatin administered either as a single agent or in combination with sufentanil in a validated preclinical rat model of postoperative pain induced by plantar incision. We found that incisional surgery on the hind paw of these rats induced sustained ipsilateral mechanical pain hypersensitivity that lasted for at least 10 days. A single intraperitoneal (i.p.) injection of ulinastatin prevented the development and reversed the maintenance of incision-induced mechanical pain hypersensitivity in a dose-dependent manner. However, ulinastatin had no effect on the baseline nociceptive threshold. Moreover, repeated i.p. injections of ulinastatin persistently attenuated incision-induced mechanical pain hypersensitivity and promoted recovery from the surgery. The rats did not develop any analgesic tolerance over the course of repeated injections of ulinastatin. A single i.p. injection of ulinastatin was also sufficient to inhibit the initiation and maintenance of incision-induced hyperalgesic priming when the rats were subsequently challenged with an ipsilateral intraplantar prostaglandin E2 injection. Furthermore, the combined administration of ulinastatin and sufentanil significantly enhanced the analgesic effect of sufentanil on postoperative pain, which involved mechanisms other than a direct influence on opioid receptors. These findings demonstrated that ulinastatin had a significant analgesic effect on postoperative pain and might be a novel pharmacotherapeutic agent for managing postoperative pain either alone or as an adjuvant.
Collapse
Affiliation(s)
- Zhi-Bin Guo
- Department of Anesthesiology and Perioperative Medicine, 900 Hospital of the Joint Logistic Support Force / Fuzong Clinical Medical College, Fujian Medical University, Fuzhou, 350025, Fujian, PR China; Department of Anesthesiology and Perioperative Medicine, Dongfang Hospital, Xiamen University, Fuzhou, 350025, Fujian, PR China
| | - Li Tang
- Department of Stomatology, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, PR China
| | - Li-Ping Wang
- Department of Anesthesiology and Perioperative Medicine, 900 Hospital of the Joint Logistic Support Force / Fuzong Clinical Medical College, Fujian Medical University, Fuzhou, 350025, Fujian, PR China; Department of Anesthesiology and Perioperative Medicine, Dongfang Hospital, Xiamen University, Fuzhou, 350025, Fujian, PR China
| | - Huang-Hui Wu
- Department of Anesthesiology and Perioperative Medicine, 900 Hospital of the Joint Logistic Support Force / Fuzong Clinical Medical College, Fujian Medical University, Fuzhou, 350025, Fujian, PR China
| | - Chang-Lu Huang
- Department of Anesthesiology and Perioperative Medicine, 900 Hospital of the Joint Logistic Support Force / Fuzong Clinical Medical College, Fujian Medical University, Fuzhou, 350025, Fujian, PR China
| | - Mei-Xiang Zhan
- Department of Anesthesiology and Perioperative Medicine, 900 Hospital of the Joint Logistic Support Force / Fuzong Clinical Medical College, Fujian Medical University, Fuzhou, 350025, Fujian, PR China
| | - Zhong-Mou Shi
- Department of Anesthesiology and Perioperative Medicine, 900 Hospital of the Joint Logistic Support Force / Fuzong Clinical Medical College, Fujian Medical University, Fuzhou, 350025, Fujian, PR China
| | - Chen-Long Yang
- Department of Anesthesiology and Perioperative Medicine, 900 Hospital of the Joint Logistic Support Force / Fuzong Clinical Medical College, Fujian Medical University, Fuzhou, 350025, Fujian, PR China
| | - Guo-Zhong Chen
- Department of Anesthesiology and Perioperative Medicine, 900 Hospital of the Joint Logistic Support Force / Fuzong Clinical Medical College, Fujian Medical University, Fuzhou, 350025, Fujian, PR China; Department of Anesthesiology and Perioperative Medicine, Dongfang Hospital, Xiamen University, Fuzhou, 350025, Fujian, PR China
| | - Yi-Qing Zou
- Department of Anesthesiology and Perioperative Medicine, 900 Hospital of the Joint Logistic Support Force / Fuzong Clinical Medical College, Fujian Medical University, Fuzhou, 350025, Fujian, PR China; Department of Anesthesiology and Perioperative Medicine, Dongfang Hospital, Xiamen University, Fuzhou, 350025, Fujian, PR China.
| | - Fei Yang
- Department of Anesthesiology and Perioperative Medicine, 900 Hospital of the Joint Logistic Support Force / Fuzong Clinical Medical College, Fujian Medical University, Fuzhou, 350025, Fujian, PR China; Department of Anesthesiology and Perioperative Medicine, Dongfang Hospital, Xiamen University, Fuzhou, 350025, Fujian, PR China; Pain Research Laboratory, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350122, Fujian, PR China.
| | - Xiao-Zhi Wu
- Department of Anesthesiology and Perioperative Medicine, 900 Hospital of the Joint Logistic Support Force / Fuzong Clinical Medical College, Fujian Medical University, Fuzhou, 350025, Fujian, PR China; Department of Anesthesiology and Perioperative Medicine, Dongfang Hospital, Xiamen University, Fuzhou, 350025, Fujian, PR China.
| |
Collapse
|
8
|
Kuwahara A, Matsuda K, Kuwahara Y, Asano S, Inui T, Marunaka Y. Microbiota-gut-brain axis: enteroendocrine cells and the enteric nervous system form an interface between the microbiota and the central nervous system. Biomed Res 2021; 41:199-216. [PMID: 33071256 DOI: 10.2220/biomedres.41.199] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The microbiota-gut-brain axis transmits bidirectional communication between the gut and the central nervous system and links the emotional and cognitive centers of the brain with peripheral gut functions. This communication occurs along the axis via local, paracrine, and endocrine mechanisms involving a variety of gut-derived peptide/amine produced by enteroendocrine cells. Neural networks, such as the enteric nervous system, and the central nervous system, including the autonomic nervous system, also transmit information through the microbiota-gut-brain axis. Recent advances in research have described the importance of the gut microbiota in influencing normal physiology and contributing to disease. We are only beginning to understand this bidirectional communication system. In this review, we summarize the available data supporting the existence of these interactions, highlighting data related to the contribution of enteroendocrine cells and the enteric nervous system as an interface between the gut microbiota and brain.
Collapse
Affiliation(s)
- Atsukazu Kuwahara
- Research Unit for Epithelial Physiology and Research Center for Drug Discovery and Pharmaceutical Development Science, Research Organization of Science and Technology, Ritsumeikan University
| | - Kyoko Matsuda
- Department of Molecular Physiology, College of Pharmaceutical Sciences, Ritsumeikan University
| | - Yuko Kuwahara
- Research Unit for Epithelial Physiology and Research Center for Drug Discovery and Pharmaceutical Development Science, Research Organization of Science and Technology, Ritsumeikan University
| | - Shinji Asano
- Department of Molecular Physiology, College of Pharmaceutical Sciences, Ritsumeikan University
| | | | - Yoshinori Marunaka
- Research Unit for Epithelial Physiology and Research Center for Drug Discovery and Pharmaceutical Development Science, Research Organization of Science and Technology, Ritsumeikan University.,Department of Molecular Cell Physiology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine.,Research Institute for Clinical Physiology, Kyoto Industrial Health Association
| |
Collapse
|
9
|
Mwirigi J, Kume M, Hassler SN, Ahmad A, Ray PR, Jiang C, Chamessian A, Mseeh N, Ludwig BP, Rivera BD, Nieman MT, Van de Ven T, Ji RR, Dussor G, Boitano S, Vagner J, Price TJ. A Role for Protease Activated Receptor Type 3 (PAR3) in Nociception Demonstrated Through Development of a Novel Peptide Agonist. THE JOURNAL OF PAIN 2021; 22:692-706. [PMID: 33429107 PMCID: PMC8197731 DOI: 10.1016/j.jpain.2020.12.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 12/02/2020] [Accepted: 12/20/2020] [Indexed: 02/07/2023]
Abstract
The protease activated receptor (PAR) family is a group of G-protein coupled receptors (GPCRs) activated by proteolytic cleavage of the extracellular domain. PARs are expressed in a variety of cell types with crucial roles in homeostasis, immune responses, inflammation, and pain. PAR3 is the least researched of the four PARs, with little known about its expression and function. We sought to better understand its potential function in the peripheral sensory nervous system. Mouse single-cell RNA sequencing data demonstrates that PAR3 is widely expressed in dorsal root ganglion (DRG) neurons. Co-expression of PAR3 mRNA with other PARs was identified in various DRG neuron subpopulations, consistent with its proposed role as a coreceptor of other PARs. We developed a lipid tethered PAR3 agonist, C660, that selectively activates PAR3 by eliciting a Ca2+ response in DRG and trigeminal neurons. In vivo, C660 induces mechanical hypersensitivity and facial grimacing in WT but not PAR3-/- mice. We characterized other nociceptive phenotypes in PAR3-/- mice and found a loss of hyperalgesic priming in response to IL-6, carrageenan, and a PAR2 agonist, suggesting that PAR3 contributes to long-lasting nociceptor plasticity in some contexts. To examine the potential role of PAR3 in regulating the activity of other PARs in sensory neurons, we administered PAR1, PAR2, and PAR4 agonists and assessed mechanical and affective pain behaviors in WT and PAR3-/- mice. We observed that the nociceptive effects of PAR1 agonists were potentiated in the absence of PAR3. Our findings suggest a complex role of PAR3 in the physiology and plasticity of nociceptors. PERSPECTIVE: We evaluated the role of PAR3, a G-protein coupled receptor, in nociception by developing a selective peptide agonist. Our findings suggest that PAR3 contributes to nociception in various contexts and plays a role in modulating the activity of other PARs.
Collapse
Affiliation(s)
- Juliet Mwirigi
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, Richardson, Texas
| | - Moeno Kume
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, Richardson, Texas
| | - Shayne N Hassler
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, Richardson, Texas
| | - Ayesha Ahmad
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, Richardson, Texas
| | - Pradipta R Ray
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, Richardson, Texas
| | - Changyu Jiang
- Duke University School of Medicine, Department of Anesthesiology, Pharmacology, and Cancer Biology, Durham, North Carolina
| | - Alexander Chamessian
- Duke University School of Medicine, Department of Anesthesiology, Pharmacology, and Cancer Biology, Durham, North Carolina
| | - Nakleh Mseeh
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, Richardson, Texas
| | - Breya P Ludwig
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, Richardson, Texas
| | - Benjamin D Rivera
- Department of Physiology, University of Arizona, Asthma and Airway Disease Research Center, Tucson, Arizona
| | - Marvin T Nieman
- Case Western Reserve University School of Medicine, Department of Pharmacology, Cleveland, Ohio
| | - Thomas Van de Ven
- Duke University School of Medicine, Department of Anesthesiology, Pharmacology, and Cancer Biology, Durham, North Carolina
| | - Ru-Rong Ji
- Duke University School of Medicine, Department of Anesthesiology, Pharmacology, and Cancer Biology, Durham, North Carolina
| | - Gregory Dussor
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, Richardson, Texas
| | - Scott Boitano
- Department of Physiology, University of Arizona, Asthma and Airway Disease Research Center, Tucson, Arizona
| | - Josef Vagner
- University of Arizona, Bio5 Research Institute, Tucson, Arizona
| | - Theodore J Price
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, Richardson, Texas.
| |
Collapse
|
10
|
Brizuela M, Castro J, Harrington AM, Brierley SM. Pruritogenic mechanisms and gut sensation: putting the "irritant" into irritable bowel syndrome. Am J Physiol Gastrointest Liver Physiol 2021; 320:G1131-G1141. [PMID: 33949199 DOI: 10.1152/ajpgi.00331.2020] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Chronic abdominal pain is a common clinical condition experienced by patients with irritable bowel syndrome (IBS). A general lack of suitable treatment options for the management of visceral pain is the major contributing factor to the debilitating nature of the disease. Understanding the underlying causes of chronic visceral pain is pivotal to identifying new effective therapies for IBS. This review provides the current evidence, demonstrating that mediators and receptors that induce itch in the skin also act as "gut irritants" in the gastrointestinal tract. Activation of these receptors triggers specific changes in the neuronal excitability of sensory pathways responsible for the transmission of nociceptive information from the periphery to the central nervous system leading to visceral hypersensitivity and visceral pain. Accumulating evidence points to significant roles of irritant mediators and their receptors in visceral hypersensitivity and thus constitutes potential targets for the development of more effective therapeutic options for IBS.
Collapse
Affiliation(s)
- Mariana Brizuela
- Visceral Pain Research Group, College of Medicine and Public Health, Flinders Health and Medical Research Institute, Flinders University, Adelaide, South Australia, Australia.,Hopwood Centre for Neurobiology, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| | - Joel Castro
- Visceral Pain Research Group, College of Medicine and Public Health, Flinders Health and Medical Research Institute, Flinders University, Adelaide, South Australia, Australia.,Hopwood Centre for Neurobiology, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| | - Andrea M Harrington
- Visceral Pain Research Group, College of Medicine and Public Health, Flinders Health and Medical Research Institute, Flinders University, Adelaide, South Australia, Australia.,Hopwood Centre for Neurobiology, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| | - Stuart M Brierley
- Visceral Pain Research Group, College of Medicine and Public Health, Flinders Health and Medical Research Institute, Flinders University, Adelaide, South Australia, Australia.,Hopwood Centre for Neurobiology, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia.,Discipline of Medicine, University of Adelaide, Adelaide, South Australia, Australia
| |
Collapse
|
11
|
Zhan MX, Tang L, Lu YF, Wu HH, Guo ZB, Shi ZM, Yang CL, Zou YQ, Yang F, Chen GZ. Ulinastatin Exhibits Antinociception in Rat Models of Acute Somatic and Visceral Pain Through Inhibiting the Local and Central Inflammation. J Pain Res 2021; 14:1201-1214. [PMID: 33976570 PMCID: PMC8106509 DOI: 10.2147/jpr.s303595] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 03/31/2021] [Indexed: 12/28/2022] Open
Abstract
Introduction Ulinastatin, a broad-spectrum serine protease inhibitor, has been widely used to treat various diseases clinically. However, so far, the antinociceptive effect of ulinastatin remains less studied experimentally and the underlying mechanisms of ulinastatin for pain relief remain unclear. This study aimed to find evidence of the analgesic effect of ulinastatin on acute somatic and visceral pain. Methods The analgesic effect of ulinastatin on acute somatic and visceral pain was evaluated by using formalin and acetic acid-induced writhing test. The analgesic mechanism of ulinastatin was verified by detecting the peripheral inflammatory cell infiltration and spinal glial activation with hematoxylin-eosin (H&E) and immunohistochemistry staining. Results We found that both of intraperitoneal (i.p.) pre-administration and post-administration of ulinastatin could reduce the total number of flinching and the licking duration following intraplantar formalin injection in a dose-related manner. However, the inhibitory effect of ulinastatin existed only in the second phase (Phase 2) of formalin-induced spontaneous pain response, with no effect in the first phase (Phase 1). The formalin-induced edema and ulcer were also improved by i.p. administration of ulinastatin. Moreover, i.p. administration of ulinastatin was also able to delay the occurrence of acetic acid-induced writhing and reduced the total number of writhes dose-dependently. We further demonstrated that ulinastatin significantly decreased the local inflammatory cell infiltration in injured paw and peritoneum tissue under formalin and acetic acid test separately. The microglial and astrocytic activation in the spinal dorsal horn induced by intraplantar formalin and i.p. acetic acid injection were also dramatically inhibited by i.p. administration of ulinastatin. Conclusion Our results for the first time provided a new line of evidence showing that ulinastatin could attenuate acute somatic and visceral pain by inhibiting the peripheral and spinal inflammatory reaction.
Collapse
Affiliation(s)
- Mei-Xiang Zhan
- Department of Anesthesiology and Perioperative Medicine, Clinical Medical College, (900 Hospital of the Joint Logistic Support Force), Fujian Medical University, Fuzhou, Fujian, 350025, People's Republic of China.,Department of Anesthesiology and Perioperative Medicine, Dongfang Hospital, Xiamen University, Fuzhou, Fujian, 350025, People's Republic of China
| | - Li Tang
- Department of Stomatology, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, People's Republic of China
| | - Yun-Fei Lu
- Department of Anesthesiology, Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, People's Republic of China
| | - Huang-Hui Wu
- Department of Anesthesiology and Perioperative Medicine, Clinical Medical College, (900 Hospital of the Joint Logistic Support Force), Fujian Medical University, Fuzhou, Fujian, 350025, People's Republic of China
| | - Zhi-Bin Guo
- Department of Anesthesiology and Perioperative Medicine, Clinical Medical College, (900 Hospital of the Joint Logistic Support Force), Fujian Medical University, Fuzhou, Fujian, 350025, People's Republic of China
| | - Zhong-Mou Shi
- Department of Anesthesiology and Perioperative Medicine, Clinical Medical College, (900 Hospital of the Joint Logistic Support Force), Fujian Medical University, Fuzhou, Fujian, 350025, People's Republic of China
| | - Chen-Long Yang
- Department of Anesthesiology and Perioperative Medicine, Clinical Medical College, (900 Hospital of the Joint Logistic Support Force), Fujian Medical University, Fuzhou, Fujian, 350025, People's Republic of China
| | - Yi-Qing Zou
- Department of Anesthesiology and Perioperative Medicine, Clinical Medical College, (900 Hospital of the Joint Logistic Support Force), Fujian Medical University, Fuzhou, Fujian, 350025, People's Republic of China.,Department of Anesthesiology and Perioperative Medicine, Dongfang Hospital, Xiamen University, Fuzhou, Fujian, 350025, People's Republic of China
| | - Fei Yang
- Department of Anesthesiology and Perioperative Medicine, Clinical Medical College, (900 Hospital of the Joint Logistic Support Force), Fujian Medical University, Fuzhou, Fujian, 350025, People's Republic of China.,Department of Anesthesiology and Perioperative Medicine, Dongfang Hospital, Xiamen University, Fuzhou, Fujian, 350025, People's Republic of China.,Laboratory of Pain Research, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian, 350122, People's Republic of China
| | - Guo-Zhong Chen
- Department of Anesthesiology and Perioperative Medicine, Clinical Medical College, (900 Hospital of the Joint Logistic Support Force), Fujian Medical University, Fuzhou, Fujian, 350025, People's Republic of China.,Department of Anesthesiology and Perioperative Medicine, Dongfang Hospital, Xiamen University, Fuzhou, Fujian, 350025, People's Republic of China
| |
Collapse
|
12
|
Host Components That Modulate the Disease Caused by hMPV. Viruses 2021; 13:v13030519. [PMID: 33809875 PMCID: PMC8004172 DOI: 10.3390/v13030519] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 02/27/2021] [Accepted: 03/01/2021] [Indexed: 12/13/2022] Open
Abstract
Human metapneumovirus (hMPV) is one of the main pathogens responsible for acute respiratory infections in children up to 5 years of age, contributing substantially to health burden. The worldwide economic and social impact of this virus is significant and must be addressed. The structural components of hMPV (either proteins or genetic material) can be detected by several receptors expressed by host cells through the engagement of pattern recognition receptors. The recognition of the structural components of hMPV can promote the signaling of the immune response to clear the infection, leading to the activation of several pathways, such as those related to the interferon response. Even so, several intrinsic factors are capable of modulating the immune response or directly inhibiting the replication of hMPV. This article will discuss the current knowledge regarding the innate and adaptive immune response during hMPV infections. Accordingly, the host intrinsic components capable of modulating the immune response and the elements capable of restricting viral replication during hMPV infections will be examined.
Collapse
|
13
|
Kido K, Katagiri N, Kawana H, Sugino S, Yamauchi M, Masaki E. Nociceptive Sensitization by Activation of Protease-Activated Receptor 2 in a Rat Model of Incisional Pain. Brain Sci 2021; 11:brainsci11020144. [PMID: 33499207 PMCID: PMC7911446 DOI: 10.3390/brainsci11020144] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 01/13/2021] [Accepted: 01/20/2021] [Indexed: 12/20/2022] Open
Abstract
Postoperative pain and consequent inflammatory responses after tissue incision adversely affects many surgical patients due to complicated mechanisms. In this study, we examined whether activation of protease-activated receptor 2 (PAR-2), which is stimulated by tryptase from mast cells, elicits nociception and whether the PAR-2 antagonist could reduce incisional nociceptive responses in vivo and in vitro. The effects of a selective PAR-2 antagonist, N3-methylbutyryl-N-6-aminohexanoyl-piperazine (ENMD-1068), pretreatment on pain behaviors were assessed after plantar incision in rats. The effects of a PAR-2 agonist, SLIGRL-NH2, on nociception was assessed after the injection into the hind paw. Furthermore, the responses of C-mechanosensitive nociceptors to the PAR-2 agonist were observed using an in vitro skin–nerve preparation as well. Intraplantar injection of SLIGRL-NH2 elicited spontaneous nociceptive behavior and hyperalgesia. Local administration of ENMD-1068 suppressed guarding behaviors, mechanical and heat hyperalgesia only within the first few hours after incision. SLIGRL-NH2 caused ongoing activity in 47% of C-mechanonociceptors in vitro. This study suggests that PAR-2 may support early nociception after incision by direct or indirect sensitization of C-fibers in rats. Moreover, PAR-2 may play a regulatory role in the early period of postoperative pain together with other co-factors to that contribute to postoperative pain.
Collapse
Affiliation(s)
- Kanta Kido
- Department of Anesthesiology, Kanagawa Dental University Hospital, Yokosuka, Kanagawa 2388570, Japan;
- Correspondence:
| | - Norika Katagiri
- Department of Anesthesiology, Kanagawa Dental University Hospital, Yokosuka, Kanagawa 2388570, Japan;
| | - Hiromasa Kawana
- Department of Oral and Maxillofacial Implantology, Kanagawa Dental University Hospital, Yokosuka, Kanagawa 2388570, Japan;
| | - Shigekazu Sugino
- Department of Anesthesiology and Perioperative Medicine, Tohoku University School of Medicine, Sendai, Miyagi 9808575, Japan; (S.S.); (M.Y.)
| | - Masanori Yamauchi
- Department of Anesthesiology and Perioperative Medicine, Tohoku University School of Medicine, Sendai, Miyagi 9808575, Japan; (S.S.); (M.Y.)
| | - Eiji Masaki
- Department of Anesthesiology, International University of Health and Welfare Hospital, Nasushiobara, Tochigi 3292763, Japan;
| |
Collapse
|
14
|
A Phase II, Double-Blind, Randomized, Placebo-Controlled, Multicenter Study Evaluating the Efficacy and Safety of Alpha-1 Antitrypsin (AAT) (Glassia ®) in the Treatment of Recent-Onset Type 1 Diabetes. Int J Mol Sci 2019; 20:ijms20236032. [PMID: 31795482 PMCID: PMC6928874 DOI: 10.3390/ijms20236032] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Revised: 11/25/2019] [Accepted: 11/27/2019] [Indexed: 12/20/2022] Open
Abstract
Our aim was to assess the efficacy, safety, and tolerability of alpha-1 antitrypsin (AAT) as a therapeutic modality for β-cell preservation in patients with recent-onset type 1 diabetes. Seventy type 1 diabetes patients (37 males; mean age 13.1 ± 4.1years) were randomized to treatment with 22 infusions of AAT (Glassia®) (60 or 120 mg/kg) or placebo. The primary outcome was the area under the curve (AUC) of C-peptide from a 2-h mixed-meal tolerance test after 52 weeks. At week 52, C-peptide was 0.9, 0.45, and 0.48 pmol/mL in the AAT-120, AAT-60, and placebo groups (p = 0.170 and p = 0.866 vs. placebo, respectively). The declines in C-peptide glycated hemoglobin (HbA1c) and the total insulin dose (U/kg) were similar across groups. Within the predefined 12–18-years subgroup, the C-peptide AUC decreased significantly in the placebo and AAT-60 groups (−0.34 and −0.54 pmol/mL, respectively, p < 0.01), with a borderline decrease in the AAT-120 group (−0.29 pmol/mL, p = 0.047). The mean HbA1c level was significantly lower in the AAT-120 group compared to the placebo (6.7% ± 0.9% vs. 8.2 ± 1.4%, p = 0.05), and a higher percentage of patients attained HbA1c ≤ 7% (75% vs. 25%, p = 0.05). AAT was tolerated well, with a similar safety profile between groups. The AAT intervention showed promise in the subgroup of adolescents with recent-onset type 1 diabetes. Further studies are warranted to determine the impact and proposed mechanism of action of AAT in β-cell preservation.
Collapse
|
15
|
Cryan JF, O'Riordan KJ, Cowan CSM, Sandhu KV, Bastiaanssen TFS, Boehme M, Codagnone MG, Cussotto S, Fulling C, Golubeva AV, Guzzetta KE, Jaggar M, Long-Smith CM, Lyte JM, Martin JA, Molinero-Perez A, Moloney G, Morelli E, Morillas E, O'Connor R, Cruz-Pereira JS, Peterson VL, Rea K, Ritz NL, Sherwin E, Spichak S, Teichman EM, van de Wouw M, Ventura-Silva AP, Wallace-Fitzsimons SE, Hyland N, Clarke G, Dinan TG. The Microbiota-Gut-Brain Axis. Physiol Rev 2019; 99:1877-2013. [PMID: 31460832 DOI: 10.1152/physrev.00018.2018] [Citation(s) in RCA: 2425] [Impact Index Per Article: 404.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The importance of the gut-brain axis in maintaining homeostasis has long been appreciated. However, the past 15 yr have seen the emergence of the microbiota (the trillions of microorganisms within and on our bodies) as one of the key regulators of gut-brain function and has led to the appreciation of the importance of a distinct microbiota-gut-brain axis. This axis is gaining ever more traction in fields investigating the biological and physiological basis of psychiatric, neurodevelopmental, age-related, and neurodegenerative disorders. The microbiota and the brain communicate with each other via various routes including the immune system, tryptophan metabolism, the vagus nerve and the enteric nervous system, involving microbial metabolites such as short-chain fatty acids, branched chain amino acids, and peptidoglycans. Many factors can influence microbiota composition in early life, including infection, mode of birth delivery, use of antibiotic medications, the nature of nutritional provision, environmental stressors, and host genetics. At the other extreme of life, microbial diversity diminishes with aging. Stress, in particular, can significantly impact the microbiota-gut-brain axis at all stages of life. Much recent work has implicated the gut microbiota in many conditions including autism, anxiety, obesity, schizophrenia, Parkinson’s disease, and Alzheimer’s disease. Animal models have been paramount in linking the regulation of fundamental neural processes, such as neurogenesis and myelination, to microbiome activation of microglia. Moreover, translational human studies are ongoing and will greatly enhance the field. Future studies will focus on understanding the mechanisms underlying the microbiota-gut-brain axis and attempt to elucidate microbial-based intervention and therapeutic strategies for neuropsychiatric disorders.
Collapse
Affiliation(s)
- John F. Cryan
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Kenneth J. O'Riordan
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Caitlin S. M. Cowan
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Kiran V. Sandhu
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Thomaz F. S. Bastiaanssen
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Marcus Boehme
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Martin G. Codagnone
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Sofia Cussotto
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Christine Fulling
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Anna V. Golubeva
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Katherine E. Guzzetta
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Minal Jaggar
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Caitriona M. Long-Smith
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Joshua M. Lyte
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Jason A. Martin
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Alicia Molinero-Perez
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Gerard Moloney
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Emanuela Morelli
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Enrique Morillas
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Rory O'Connor
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Joana S. Cruz-Pereira
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Veronica L. Peterson
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Kieran Rea
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Nathaniel L. Ritz
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Eoin Sherwin
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Simon Spichak
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Emily M. Teichman
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Marcel van de Wouw
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Ana Paula Ventura-Silva
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Shauna E. Wallace-Fitzsimons
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Niall Hyland
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Gerard Clarke
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Timothy G. Dinan
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| |
Collapse
|
16
|
Lomax AE, Pradhananga S, Sessenwein JL, O'Malley D. Bacterial modulation of visceral sensation: mediators and mechanisms. Am J Physiol Gastrointest Liver Physiol 2019; 317:G363-G372. [PMID: 31290688 DOI: 10.1152/ajpgi.00052.2019] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The potential role of the intestinal microbiota in modulating visceral pain has received increasing attention during recent years. This has led to the identification of signaling pathways that have been implicated in communication between gut bacteria and peripheral pain pathways. In addition to the well-characterized impact of the microbiota on the immune system, which in turn affects nociceptor excitability, bacteria can modulate visceral afferent pathways by effects on enterocytes, enteroendocrine cells, and the neurons themselves. Proteases produced by bacteria, or by host cells in response to bacteria, can increase or decrease the excitability of nociceptive dorsal root ganglion (DRG) neurons depending on the receptor activated. Short-chain fatty acids generated by colonic bacteria are involved in gut-brain communication, and intracolonic short-chain fatty acids have pronociceptive effects in rodents but may be antinociceptive in humans. Gut bacteria modulate the synthesis and release of enteroendocrine cell mediators, including serotonin and glucagon-like peptide-1, which activate extrinsic afferent neurons. Deciphering the complex interactions between visceral afferent neurons and the gut microbiota may lead to the development of improved probiotic therapies for visceral pain.
Collapse
Affiliation(s)
- Alan E Lomax
- Gastrointestinal Diseases Research Unit, Queen's University, Kingston, Ontario, Canada
| | - Sabindra Pradhananga
- Gastrointestinal Diseases Research Unit, Queen's University, Kingston, Ontario, Canada
| | - Jessica L Sessenwein
- Gastrointestinal Diseases Research Unit, Queen's University, Kingston, Ontario, Canada
| | - Dervla O'Malley
- APC Microbiome Ireland, University College Cork, Cork, Ireland.,Department of Physiology, University College Cork, Cork, Ireland
| |
Collapse
|
17
|
Du Q, Liao Q, Chen C, Yang X, Xie R, Xu J. The Role of Transient Receptor Potential Vanilloid 1 in Common Diseases of the Digestive Tract and the Cardiovascular and Respiratory System. Front Physiol 2019; 10:1064. [PMID: 31496955 PMCID: PMC6712094 DOI: 10.3389/fphys.2019.01064] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Accepted: 08/02/2019] [Indexed: 01/30/2023] Open
Abstract
Transient receptor potential vanilloid subtype 1 (TRPV1), a member of the transient receptor potential vanilloid (TRPV) channel family, is a nonselective cation channel that is widely expressed in sensory nerve fibers and nonneuronal cells, including certain vascular endothelial cells and smooth muscle cells. The activation of TRPV1 may be involved in the regulation of various physiological functions, such as the release of inflammatory mediators in the body, gastrointestinal motility function, and temperature regulation. In recent years, a large number of studies have revealed that TRPV1 plays an important role in the physiological and pathological conditions of the digestive system, cardiovascular system, and respiratory system, but there is no systematic report on TRPV1. The objective of this review is to explain the function and effects of TRPV1 on specific diseases, such as irritable bowel syndrome, hypertension, and asthma, and to further investigate the intrinsic relationship between the expression and function of TRPV1 in those diseases to find new therapeutic targets for the cure of related diseases.
Collapse
Affiliation(s)
| | | | | | | | - Rui Xie
- Department of Gastroenterology, Affiliated Hospital to Zunyi Medical University, Zunyi, China
| | - Jingyu Xu
- Department of Gastroenterology, Affiliated Hospital to Zunyi Medical University, Zunyi, China
| |
Collapse
|
18
|
Desormeaux C, Bautzova T, Garcia-Caraballo S, Rolland C, Barbaro MR, Brierley SM, Barbara G, Vergnolle N, Cenac N. Protease-activated receptor 1 is implicated in irritable bowel syndrome mediators-induced signaling to thoracic human sensory neurons. Pain 2019; 159:1257-1267. [PMID: 29554016 DOI: 10.1097/j.pain.0000000000001208] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Proteases and protease-activated receptors (PARs) are major mediators involved in irritable bowel syndrome (IBS). Our objectives were to decipher the expression and functionality (calcium signaling) of PARs in human dorsal root ganglia (DRG) neurons and to define mechanisms involved in human sensory neuron signaling by IBS patient mediators. Human thoracic DRG were obtained from the national disease resource interchange. Expression of PAR1, PAR2, and PAR4 was assessed by immunohistochemistry and quantitative reverse transcription PCR (RT-qPCR) in whole DRG or in primary cultures of isolated neurons. Calcium signaling in response to PAR agonist peptides (PAR-AP), their inactive peptides (PAR-IP), thrombin (10 U/mL), supernatants from colonic biopsies of patients with IBS, or healthy controls, with or without PAR1 or PAR4 antagonist were studied in cultured human DRG neurons. PAR1, PAR2, and PAR4 were all expressed in human DRG, respectively, in 20%, 40%, and 40% of the sensory neurons. PAR1-AP increased intracellular calcium concentration in a dose-dependent manner. This increase was inhibited by PAR1 antagonism. By contrast, PAR2-AP, PAR4-AP, and PAR-IP did not cause calcium mobilization. PAR1-AP-induced calcium flux was significantly reduced by preincubation with PAR4-AP, but not with PAR2-AP. Thrombin increased calcium flux, which was inhibited by a PAR1 antagonist and increased by a PAR4 antagonist. Supernatants from colonic biopsies of patients with IBS induced calcium flux in human sensory neurons compared with healthy controls, and this induction was reversed by a PAR1 antagonist. Taken together, our results highlight that PAR1 antagonism should be investigated as a new therapeutic target for IBS symptoms.
Collapse
Affiliation(s)
- Cleo Desormeaux
- IRSD, Université de Toulouse, INSERM, INRA, ENVT, UPS, Toulouse, France
| | - Tereza Bautzova
- IRSD, Université de Toulouse, INSERM, INRA, ENVT, UPS, Toulouse, France
| | - Sonia Garcia-Caraballo
- Visceral Pain Group, Human Physiology, Flinders University, Bedford Park, South Australia, Australia.,Centre for Nutrition and Gastrointestinal Diseases, Discipline of Medicine, University of Adelaide, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, South Australia, Australia
| | - Corinne Rolland
- IRSD, Université de Toulouse, INSERM, INRA, ENVT, UPS, Toulouse, France
| | | | - Stuart M Brierley
- Visceral Pain Group, Human Physiology, Flinders University, Bedford Park, South Australia, Australia.,Centre for Nutrition and Gastrointestinal Diseases, Discipline of Medicine, University of Adelaide, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, South Australia, Australia
| | - Giovanni Barbara
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Nathalie Vergnolle
- IRSD, Université de Toulouse, INSERM, INRA, ENVT, UPS, Toulouse, France.,Department of Physiology and Pharmacology, Faculty of Medicine, University of Calgary, Calgary, Canada
| | - Nicolas Cenac
- IRSD, Université de Toulouse, INSERM, INRA, ENVT, UPS, Toulouse, France
| |
Collapse
|
19
|
Tikoo S, Barki N, Jain R, Zulkhernain NS, Buhner S, Schemann M, Weninger W. Imaging of mast cells. Immunol Rev 2019; 282:58-72. [PMID: 29431206 DOI: 10.1111/imr.12631] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Mast cells are a part of the innate immune system implicated in allergic reactions and the regulation of host-pathogen interactions. The distribution, morphology and biochemical composition of mast cells has been studied in detail in vitro and on tissue sections both at the light microscopic and ultrastructural level. More recently, the development of fluorescent reporter strains and intravital imaging modalities has enabled first glimpses of the real-time behavior of mast cells in situ. In this review, we describe commonly used imaging approaches to study mast cells in cell culture as well as within normal and diseased tissues. We further describe the interrogation of mast cell function via imaging by providing a detailed description of mast cell-nerve plexus interactions in the intestinal tract. Together, visualizing mast cells has expanded our view of these cells in health and disease.
Collapse
Affiliation(s)
- Shweta Tikoo
- The Centenary Institute, Newtown, NSW, Australia.,Discipline of Dermatology, Sydney Medical School, Sydney, NSW, Australia
| | - Natasja Barki
- LS Human Biology, Technical University München, München, Germany
| | - Rohit Jain
- The Centenary Institute, Newtown, NSW, Australia.,Discipline of Dermatology, Sydney Medical School, Sydney, NSW, Australia
| | | | - Sabine Buhner
- LS Human Biology, Technical University München, München, Germany
| | - Michael Schemann
- LS Human Biology, Technical University München, München, Germany
| | - Wolfgang Weninger
- The Centenary Institute, Newtown, NSW, Australia.,Discipline of Dermatology, Sydney Medical School, Sydney, NSW, Australia.,Department of Dermatology, Royal Prince Alfred Hospital, Camperdown, NSW, Australia
| |
Collapse
|
20
|
Abstract
Proteases drive the life cycle of all proteins, ensuring the transportation and activation of newly minted, would-be proteins into their functional form while recycling spent or unneeded proteins. Far from their image as engines of protein digestion, proteases play fundamental roles in basic physiology and regulation at multiple levels of systems biology. Proteases are intimately associated with disease and modulation of proteolytic activity is the presumed target for successful therapeutics. "Proteases: Pivot Points in Functional Proteomics" examines the crucial roles of proteolysis across a wide range of physiological processes and diseases. The existing and potential impacts of proteolysis-related activity on drug and biomarker development are presented in detail. All told the decisive roles of proteases in four major categories comprising 23 separate subcategories are addressed. Within this construct, 15 sets of subject-specific, tabulated data are presented that include identification of proteases, protease inhibitors, substrates, and their actions. Said data are derived from and confirmed by over 300 references. Cross comparison of datasets indicates that proteases, their inhibitors/promoters and substrates intersect over a range of physiological processes and diseases, both chronic and pathogenic. Indeed, "Proteases: Pivot Points …" closes by dramatizing this very point through association of (pro)Thrombin and Fibrin(ogen) with: hemostasis, innate immunity, cardiovascular and metabolic disease, cancer, neurodegeneration, and bacterial self-defense.
Collapse
Affiliation(s)
- Ingrid M Verhamme
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN, USA.
| | - Sarah E Leonard
- Chemical and Biomolecular Engineering, University of Illinois Champaign-Urbana School of Chemical Sciences, Champaign, IL, USA
| | - Ray C Perkins
- New Liberty Proteomics Corporation, New Liberty, KY, USA.
| |
Collapse
|
21
|
Rigg RA, Healy LD, Chu TT, Ngo ATP, Mitrugno A, Zilberman-Rudenko J, Aslan JE, Hinds MT, Vecchiarelli LD, Morgan TK, Gruber A, Temple KJ, Lindsley CW, Duvernay MT, Hamm HE, McCarty OJT. Protease-activated receptor 4 activity promotes platelet granule release and platelet-leukocyte interactions. Platelets 2018; 30:126-135. [PMID: 30560697 DOI: 10.1080/09537104.2017.1406076] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Human platelets express two protease-activated receptors (PARs), PAR1 (F2R) and PAR4 (F2RL3), which are activated by a number of serine proteases that are generated during pathological events and cause platelet activation. Recent interest has focused on PAR4 as a therapeutic target, given PAR4 seems to promote experimental thrombosis and procoagulant microparticle formation, without a broadly apparent role in hemostasis. However, it is not yet known whether PAR4 activity plays a role in platelet-leukocyte interactions, which are thought to contribute to both thrombosis and acute or chronic thrombo-inflammatory processes. We sought to determine whether PAR4 activity contributes to granule secretion from activated platelets and platelet-leukocyte interactions. We performed in vitro and ex vivo studies of platelet granule release and platelet-leukocyte interactions in the presence of PAR4 agonists including PAR4 activating peptide, thrombin, cathepsin G, and plasmin in combination with small-molecule PAR4 antagonists. Activation of human platelets with thrombin, cathepsin G, or plasmin potentiated platelet dense granule secretion that was specifically impaired by PAR4 inhibitors. Platelet-leukocyte interactions and platelet P-selectin exposure the following stimulation with PAR4 agonists were also impaired by activated PAR4 inhibition in either a purified system or in whole blood. These results indicate PAR4-specific promotion of platelet granule release and platelet-leukocyte aggregate formation and suggest that pharmacological control of PAR4 activity could potentially attenuate platelet granule release or platelet-leukocyte interaction-mediated pathological processes.
Collapse
Affiliation(s)
- Rachel A Rigg
- a Department of Biomedical Engineering , School of Medicine, Oregon Health & Science University , Portland , OR , USA
| | - Laura D Healy
- b Department of Cell, Developmental & Cancer Biology , School of Medicine, Oregon Health & Science University , Portland , OR , USA
| | - Tiffany T Chu
- a Department of Biomedical Engineering , School of Medicine, Oregon Health & Science University , Portland , OR , USA
| | - Anh T P Ngo
- a Department of Biomedical Engineering , School of Medicine, Oregon Health & Science University , Portland , OR , USA
| | - Annachiara Mitrugno
- a Department of Biomedical Engineering , School of Medicine, Oregon Health & Science University , Portland , OR , USA
| | - Jevgenia Zilberman-Rudenko
- a Department of Biomedical Engineering , School of Medicine, Oregon Health & Science University , Portland , OR , USA
| | - Joseph E Aslan
- d Department of Biochemistry and Molecular Biology , School of Medicine, Oregon Health & Science University , Portland , OR , USA.,e Knight Cardiovascular Institute , School of Medicine, Oregon Health & Science University , Portland , OR , USA
| | - Monica T Hinds
- a Department of Biomedical Engineering , School of Medicine, Oregon Health & Science University , Portland , OR , USA
| | - Lisa Dirling Vecchiarelli
- f Department of Pathology , School of Medicine, Oregon Health & Science University , Portland , OR , USA
| | - Terry K Morgan
- f Department of Pathology , School of Medicine, Oregon Health & Science University , Portland , OR , USA
| | - András Gruber
- a Department of Biomedical Engineering , School of Medicine, Oregon Health & Science University , Portland , OR , USA.,c Division of Hematology & Medical Oncology , School of Medicine, Oregon Health & Science University , Portland , OR , USA
| | - Kayla J Temple
- g Department of Pharmacology , Vanderbilt University School of Medicine , Nashville , TN , USA.,h Vanderbilt Center for Neuroscience Drug Discovery , Nashville , TN , USA
| | - Craig W Lindsley
- g Department of Pharmacology , Vanderbilt University School of Medicine , Nashville , TN , USA.,h Vanderbilt Center for Neuroscience Drug Discovery , Nashville , TN , USA
| | - Matthew T Duvernay
- g Department of Pharmacology , Vanderbilt University School of Medicine , Nashville , TN , USA
| | - Heidi E Hamm
- g Department of Pharmacology , Vanderbilt University School of Medicine , Nashville , TN , USA
| | - Owen J T McCarty
- a Department of Biomedical Engineering , School of Medicine, Oregon Health & Science University , Portland , OR , USA.,b Department of Cell, Developmental & Cancer Biology , School of Medicine, Oregon Health & Science University , Portland , OR , USA.,c Division of Hematology & Medical Oncology , School of Medicine, Oregon Health & Science University , Portland , OR , USA
| |
Collapse
|
22
|
George T, Chakraborty M, Giembycz MA, Newton R. A bronchoprotective role for Rgs2 in a murine model of lipopolysaccharide-induced airways inflammation. Allergy Asthma Clin Immunol 2018; 14:40. [PMID: 30305828 PMCID: PMC6166284 DOI: 10.1186/s13223-018-0266-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Accepted: 05/22/2018] [Indexed: 02/07/2023] Open
Abstract
Background Asthma exacerbations are associated with the recruitment of neutrophils to the lungs. These cells release proteases and mediators, many of which act at G protein-coupled receptors (GPCRs) that couple via Gq to promote bronchoconstriction and inflammation. Common asthma therapeutics up-regulate expression of the regulator of G protein signalling (RGS), RGS2. As RGS2 reduces signaling from Gq-coupled GPCRs, we have defined role(s) for this GTPase-activating protein in an acute neutrophilic model of lung inflammation. Methods Wild type and Rgs2−/− C57Bl6 mice were exposed to nebulized lipopolysaccharide (LPS). Lung function (respiratory system resistance and compliance) was measured using a SCIREQ flexivent small animal ventilator. Lung inflammation was assessed by histochemistry, cell counting and by cytokine and chemokine expression in bronchoalveolar lavage (BAL) fluid. Results Lipopolysaccharide inhalation induced transient airways hyperreactivity (AHR) and neutrophilic lung inflammation. While AHR and inflammation was greatest 3 h post-LPS exposure, BAL neutrophils persisted for 24 h. At 3 h post-LPS inhalation, multiple inflammatory cytokines (CSF2, CSF3, IL6, TNF) and chemokines (CCL3, CCL4, CXCL1, CXCL2) were highly expressed in the BAL fluid, prior to declining by 24 h. Compared to wild type counterparts, Rgs2−/− mice developed significantly greater airflow resistance in response to inhaled methacholine (MCh) at 3 h post-LPS exposure. At 24 h post-LPS exposure, when lung function was recovering in the wild type animals, MCh-induced resistance was increased, and compliance decreased, in Rgs2−/− mice. Thus, Rgs2−/− mice show AHR and stiffer lungs 24 h post-LPS exposure. Histological markers of inflammation, total and differential cell counts, and major cytokine and chemokine expression in BAL fluid were similar between wild type and Rgs2−/− mice. However, 3 and 24 h post-LPS exposure, IL12B expression was significantly elevated in BAL fluid from Rgs2−/− mice compared to wild type animals. Conclusions While Rgs2 is bronchoprotective in acute neutrophilic inflammation, no clear anti-inflammatory effect was apparent. Nevertheless, elevated IL12B expression in Rgs2−/− animals raises the possibility that RGS2 could dampen Th1 responses. These findings indicate that up-regulation of RGS2, as occurs in response to inhaled corticosteroids and long-acting β2-adrenoceptor agonists, may be beneficial in acute neutrophilic exacerbations of airway disease, including asthma. Electronic supplementary material The online version of this article (10.1186/s13223-018-0266-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Tresa George
- 1Airways Inflammation Research Group, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB T2N 4Z6 Canada
| | - Mainak Chakraborty
- 2Immunology Research Group, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB T2N 4Z6 Canada
| | - Mark A Giembycz
- 1Airways Inflammation Research Group, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB T2N 4Z6 Canada
| | - Robert Newton
- 1Airways Inflammation Research Group, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB T2N 4Z6 Canada
| |
Collapse
|
23
|
Sébert M, Denadai-Souza A, Quaranta M, Racaud-Sultan C, Chabot S, Lluel P, Monjotin N, Alric L, Portier G, Kirzin S, Bonnet D, Ferrand A, Vergnolle N. Thrombin modifies growth, proliferation and apoptosis of human colon organoids: a protease-activated receptor 1- and protease-activated receptor 4-dependent mechanism. Br J Pharmacol 2018; 175:3656-3668. [PMID: 29959891 PMCID: PMC6109216 DOI: 10.1111/bph.14430] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 04/24/2018] [Accepted: 06/08/2018] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND AND PURPOSE Thrombin is massively released upon tissue damage associated with bleeding or chronic inflammation. The effects of this thrombin on tissue regrowth and repair has been scarcely addressed and only in cancer cell lines. Hence, the purpose of the present study was to determine thrombin's pharmacological effects on human intestinal epithelium growth, proliferation and apoptosis, using three-dimensional cultures of human colon organoids. EXPERIMENTAL APPROACH Crypts were isolated from human colonic resections and cultured for 6 days, forming human colon organoids. Cultured organoids were exposed to 10 and 50 mU·mL-1 of thrombin, in the presence or not of protease-activated receptor (PAR) antagonists. Organoid morphology, metabolism, proliferation and apoptosis were followed. KEY RESULTS Thrombin favoured organoid maturation leading to a decreased number of immature cystic structures and a concomitant increased number of larger structures releasing cell debris and apoptotic cells. The size of budding structures, metabolic activity and proliferation were significantly reduced in organoid cultures exposed to thrombin, while apoptosis was dramatically increased. Both PAR1 and PAR4 antagonists inhibited apoptosis regardless of thrombin doses. Thrombin-induced inhibition of proliferation and metabolic activity were reversed by PAR4 antagonist for thrombin's lowest dose and by PAR1 antagonist for thrombin's highest dose. CONCLUSIONS AND IMPLICATIONS Overall, our data suggest that the presence of thrombin in the vicinity of human colon epithelial cells favours their maturation at the expense of their regenerative capacities. Our data point to thrombin and its two receptors PAR1 and PAR4 as potential molecular targets for epithelial repair therapies.
Collapse
Affiliation(s)
- Morgane Sébert
- IRSD, Université de Toulouse, INSERM, INRA, ENVT, UPS, Toulouse, France
| | | | - Muriel Quaranta
- IRSD, Université de Toulouse, INSERM, INRA, ENVT, UPS, Toulouse, France
| | | | | | | | | | - Laurent Alric
- Department of Internal Medicine and Digestive Diseases, CHU Purpan, Toulouse, France
| | - Guillaume Portier
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
| | - Sylvain Kirzin
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
| | - Delphine Bonnet
- IRSD, Université de Toulouse, INSERM, INRA, ENVT, UPS, Toulouse, France
| | - Audrey Ferrand
- IRSD, Université de Toulouse, INSERM, INRA, ENVT, UPS, Toulouse, France
| | - Nathalie Vergnolle
- IRSD, Université de Toulouse, INSERM, INRA, ENVT, UPS, Toulouse, France.,Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
24
|
Kumar H, Lee SH, Kim KT, Zeng X, Han I. TRPV4: a Sensor for Homeostasis and Pathological Events in the CNS. Mol Neurobiol 2018; 55:8695-8708. [PMID: 29582401 DOI: 10.1007/s12035-018-0998-8] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2017] [Accepted: 03/07/2018] [Indexed: 01/22/2023]
Abstract
Transient receptor potential vanilloid type 4 (TRPV4) was originally described as a calcium-permeable nonselective cation channel. TRPV4 is now recognized as a polymodal ionotropic receptor: it is a broadly expressed, nonselective cation channel (permeable to calcium, potassium, magnesium, and sodium) that plays an important role in a multitude of physiological processes. TRPV4 is involved in maintaining homeostasis, serves as an osmosensor and thermosensor, can be activated directly by endogenous or exogenous chemical stimuli, and can be activated or sensitized indirectly via intracellular signaling pathways. Additionally, TRPV4 is upregulated in a variety of pathological conditions. In this review, we focus on the role of TRPV4 in mediating homeostasis and pathological events in the central nervous system (CNS). This review is composed of three parts. Section 1 describes the role of TRPV4 in maintaining homeostatic processes, including the volume of body water, ionic concentrations, volume, and the temperature. Section 2 describes the effects of activation and inhibition of TRPV4 in the CNS. Section 3 focuses on the role of TRPV4 during pathological events in CNS.
Collapse
Affiliation(s)
- Hemant Kumar
- Department of Neurosurgery, CHA University, CHA Bundang Medical Center, Seongnam-si, Gyeonggi-do, 13496, Republic of Korea
| | - Soo-Hong Lee
- Department of Biomedical Science, CHA University, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Kyoung-Tae Kim
- Department of Neurosurgery, Kyungpook National University Hospital, 130, Dongdeok-ro, Jung-gu, Daegu, 41944, Republic of Korea
| | - Xiang Zeng
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China.
| | - Inbo Han
- Department of Neurosurgery, CHA University, CHA Bundang Medical Center, Seongnam-si, Gyeonggi-do, 13496, Republic of Korea.
| |
Collapse
|
25
|
Meegan JE, Yang X, Coleman DC, Jannaway M, Yuan SY. Neutrophil-mediated vascular barrier injury: Role of neutrophil extracellular traps. Microcirculation 2018; 24. [PMID: 28120468 DOI: 10.1111/micc.12352] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Accepted: 01/12/2017] [Indexed: 12/19/2022]
Abstract
Neutrophils play an essential role in host defense against infection or injury. While neutrophil activation is necessary for pathogen clearance and tissue repair, a hyperactive response can lead to tissue damage and microcirculatory disorders, a process involving complex neutrophil-endothelium cross talk. This review highlights recent research findings about neutrophil-mediated signaling and structural changes, including those induced by neutrophil extracellular traps, which ultimately lead to vascular barrier injury.
Collapse
Affiliation(s)
- Jamie E Meegan
- Department of Molecular Pharmacology and Physiology, University of South Florida College of Medicine, Tampa, FL, USA
| | - Xiaoyuan Yang
- Department of Molecular Pharmacology and Physiology, University of South Florida College of Medicine, Tampa, FL, USA
| | - Danielle C Coleman
- Department of Molecular Pharmacology and Physiology, University of South Florida College of Medicine, Tampa, FL, USA
| | - Melanie Jannaway
- Department of Molecular Pharmacology and Physiology, University of South Florida College of Medicine, Tampa, FL, USA
| | - Sarah Y Yuan
- Department of Molecular Pharmacology and Physiology, University of South Florida College of Medicine, Tampa, FL, USA
| |
Collapse
|
26
|
Oikonomopoulou K, Diamandis EP, Hollenberg MD, Chandran V. Proteinases and their receptors in inflammatory arthritis: an overview. Nat Rev Rheumatol 2018; 14:170-180. [PMID: 29416136 DOI: 10.1038/nrrheum.2018.17] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Proteinases are enzymes with established roles in physiological and pathological processes such as digestion and the homeostasis, destruction and repair of tissues. Over the past few years, the hormone-like properties of circulating proteinases have become increasingly appreciated. Some proteolytic enzymes trigger cell signalling via proteinase-activated receptors, a family of G protein-coupled receptors that have been implicated in inflammation and pain in inflammatory arthritis. Proteinases can also regulate ion flux owing to the cross-sensitization of transient receptor potential cation channel subfamily V members 1 and 4, which are associated with mechanosensing and pain. In this Review, the idea that proteinases have the potential to orchestrate inflammatory signals by interacting with receptors on cells within the synovial microenvironment of an inflamed joint is revisited in three arthritic diseases: osteoarthritis, spondyloarthritis and rheumatoid arthritis. Unanswered questions are highlighted and the therapeutic potential of modulating this proteinase-receptor axis for the management of disease in patients with these types of arthritis is also discussed.
Collapse
Affiliation(s)
- Katerina Oikonomopoulou
- Centre for Prognosis Studies in Rheumatic Diseases, Krembil Research Institute, Toronto Western Hospital, University Health Network, Toronto, Ontario, Canada
| | - Eleftherios P Diamandis
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada.,Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, Ontario, Canada.,Department of Clinical Biochemistry, University Health Network, Toronto, Ontario, Canada
| | - Morley D Hollenberg
- Department of Physiology & Pharmacology, University of Calgary Cumming School of Medicine, Calgary, Alberta, Canada.,Department of Medicine, University of Calgary Cumming School of Medicine, Calgary, Alberta, Canada
| | - Vinod Chandran
- Centre for Prognosis Studies in Rheumatic Diseases, Krembil Research Institute, Toronto Western Hospital, University Health Network, Toronto, Ontario, Canada.,Division of Rheumatology, Department of Medicine, University of Toronto, Toronto, Ontario, Canada.,Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
27
|
Lê VB, Riteau B, Alessi MC, Couture C, Jandrot-Perrus M, Rhéaume C, Hamelin MÈ, Boivin G. Protease-activated receptor 1 inhibition protects mice against thrombin-dependent respiratory syncytial virus and human metapneumovirus infections. Br J Pharmacol 2017; 175:388-403. [PMID: 29105740 DOI: 10.1111/bph.14084] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 10/11/2017] [Accepted: 10/13/2017] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND AND PURPOSE Protease-activated receptor 1 (PAR1) has been demonstrated to be involved in the pathogenesis of viral diseases. However, its role remains controversial. The goal of our study was to investigate the contribution of PAR1 to respiratory syncytial virus (RSV) and human metapneumovirus (hMPV) infections. EXPERIMENTAL APPROACH Pharmacological approaches were used to investigate the role of PAR1 during RSV and hMPV infection, in vitro using epithelial A549 cells and in vivo using a mouse model of virus infection. KEY RESULTS In vitro, the PAR1 antagonist RWJ-56110 reduced the replication of RSV and hMPV in A549 cells. In agreement with these results, RWJ-56110-treated mice were protected against RSV and hMPV infections, as indicated by less weight loss and mortality. This protective effect in mice correlated with decreased lung viral replication and inflammation. In contrast, hMPV-infected mice treated with the PAR1 agonist TFLLR-NH2 showed increased mortality, as compared to infected mice, which were left untreated. Thrombin generation was shown to occur downstream of PAR1 activation in infected mice via tissue factor exposure as part of the inflammatory response, and thrombin inhibition by argatroban reduced the pathogenicity of the infection with no additive effect to that induced by PAR1 inhibition. CONCLUSION AND IMPLICATIONS These data show that PAR1 plays a detrimental role during RSV and hMPV infections in mice via, at least, a thrombin-dependent mechanism. Thus, the use of PAR1 antagonists and thrombin inhibitors may have potential as a novel approach for the treatment of RSV and hMPV infections.
Collapse
Affiliation(s)
- Vuong Ba Lê
- Infectious Disease Research Centre, Laval University, Quebec City, Quebec, Canada
| | - Béatrice Riteau
- UMR INSERM U1062/INRA 1260/AMU, Aix Marseille University, Marseille, France
| | | | - Christian Couture
- Department of Anatomy-Pathology, Laval University Institute of Cardiology and Pneumology, Quebec City, Quebec, Canada
| | | | - Chantal Rhéaume
- Infectious Disease Research Centre, Laval University, Quebec City, Quebec, Canada
| | - Marie-Ève Hamelin
- Infectious Disease Research Centre, Laval University, Quebec City, Quebec, Canada
| | - Guy Boivin
- Infectious Disease Research Centre, Laval University, Quebec City, Quebec, Canada
| |
Collapse
|
28
|
Opportunities for therapeutic antibodies directed at G-protein-coupled receptors. Nat Rev Drug Discov 2017; 16:787-810. [PMID: 28706220 DOI: 10.1038/nrd.2017.91] [Citation(s) in RCA: 108] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
G-protein-coupled receptors (GPCRs) are activated by a diverse range of ligands, from large proteins and proteases to small peptides, metabolites, neurotransmitters and ions. They are expressed on all cells in the body and have key roles in physiology and homeostasis. As such, GPCRs are one of the most important target classes for therapeutic drug discovery. The development of drugs targeting GPCRs has therapeutic value across a wide range of diseases, including cancer, immune and inflammatory disorders as well as neurological and metabolic diseases. The progress made by targeting GPCRs with antibody-based therapeutics, as well as technical hurdles to overcome, are presented and discussed in this Review. Antibody therapeutics targeting C-C chemokine receptor type 4 (CCR4), CCR5 and calcitonin gene-related peptide (CGRP) are used as illustrative clinical case studies.
Collapse
|
29
|
Rovai ES, Holzhausen M. The Role of Proteinase-Activated Receptors 1 and 2 in the Regulation of Periodontal Tissue Metabolism and Disease. J Immunol Res 2017; 2017:5193572. [PMID: 28503577 PMCID: PMC5414592 DOI: 10.1155/2017/5193572] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Revised: 01/13/2017] [Accepted: 03/05/2017] [Indexed: 01/13/2023] Open
Abstract
Proteinase-activated receptors 1 (PAR1) and 2 (PAR2) are the most highly expressed members of the PAR family in the periodontium. These receptors regulate periodontal inflammatory and repair processes through their activation by endogenous and bacterial enzymes. PAR1 is expressed by the periodontal cells such as human gingival fibroblasts, gingival epithelial cells, periodontal ligament cells, osteoblasts, and monocytic cells and can be activated by thrombin, matrix metalloproteinase 1 (MMP-1), MMP-13, fibrin, and gingipains from Porphyromonas gingivalis. PAR2 is expressed by neutrophils, osteoblasts, oral epithelial cells, and human gingival fibroblasts, and its possible activators in the periodontium are gingipains, neutrophil proteinase 3, and mast cell tryptase. The mechanisms through which PARs can respond to periodontal enzymes and result in appropriate immune responses have until recently been poorly understood. This review discusses recent findings that are beginning to identify a cardinal role for PAR1 and PAR2 on periodontal tissue metabolism.
Collapse
Affiliation(s)
- E. S. Rovai
- Division of Periodontics, Department of Stomatology, School of Dentistry, University of São Paulo, São Paulo, SP, Brazil
| | - M. Holzhausen
- Division of Periodontics, Department of Stomatology, School of Dentistry, University of São Paulo, São Paulo, SP, Brazil
| |
Collapse
|
30
|
Van Spaendonk H, Ceuleers H, Witters L, Patteet E, Joossens J, Augustyns K, Lambeir AM, De Meester I, De Man JG, De Winter BY. Regulation of intestinal permeability: The role of proteases. World J Gastroenterol 2017; 23:2106-2123. [PMID: 28405139 PMCID: PMC5374123 DOI: 10.3748/wjg.v23.i12.2106] [Citation(s) in RCA: 120] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Revised: 01/20/2017] [Accepted: 03/02/2017] [Indexed: 02/06/2023] Open
Abstract
The gastrointestinal barrier is - with approximately 400 m2 - the human body’s largest surface separating the external environment from the internal milieu. This barrier serves a dual function: permitting the absorption of nutrients, water and electrolytes on the one hand, while limiting host contact with noxious luminal antigens on the other hand. To maintain this selective barrier, junction protein complexes seal the intercellular space between adjacent epithelial cells and regulate the paracellular transport. Increased intestinal permeability is associated with and suggested as a player in the pathophysiology of various gastrointestinal and extra-intestinal diseases such as inflammatory bowel disease, celiac disease and type 1 diabetes. The gastrointestinal tract is exposed to high levels of endogenous and exogenous proteases, both in the lumen and in the mucosa. There is increasing evidence to suggest that a dysregulation of the protease/antiprotease balance in the gut contributes to epithelial damage and increased permeability. Excessive proteolysis leads to direct cleavage of intercellular junction proteins, or to opening of the junction proteins via activation of protease activated receptors. In addition, proteases regulate the activity and availability of cytokines and growth factors, which are also known modulators of intestinal permeability. This review aims at outlining the mechanisms by which proteases alter the intestinal permeability. More knowledge on the role of proteases in mucosal homeostasis and gastrointestinal barrier function will definitely contribute to the identification of new therapeutic targets for permeability-related diseases.
Collapse
|
31
|
Coleman JLJ, Ngo T, Smith NJ. The G protein-coupled receptor N-terminus and receptor signalling: N-tering a new era. Cell Signal 2017; 33:1-9. [PMID: 28188824 DOI: 10.1016/j.cellsig.2017.02.004] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Accepted: 02/06/2017] [Indexed: 01/22/2023]
Abstract
G protein-coupled receptors (GPCRs) are a vast family of membrane-traversing proteins, essential to the ability of eukaryotic life to detect, and mount an intracellular response to, a diverse range of extracellular stimuli. GPCRs have evolved with archetypal features including an extracellular N-terminus and intracellular C-terminus that flank a transmembrane structure of seven sequential helices joined by intracellular and extracellular loops. These structural domains contribute to the ability of a GPCR to be correctly synthesised and inserted into the cell membrane, to interact with its cognate ligand(s) and to couple with signal-transducing heterotrimeric G proteins, allowing the activated receptor to selectively modulate a number of signalling cascades. Whilst well known for its importance in receptor translation and trafficking, the GPCR N-terminus is underexplored as a participant in receptor signalling. This review aims to discuss and integrate recent advances in knowledge of the vital roles of the GPCR N-terminus in receptor signalling.
Collapse
Affiliation(s)
- James L J Coleman
- Molecular Pharmacology Group, Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia; St Vincent's Clinical School, University of New South Wales, Darlinghurst, NSW 2010, Australia.
| | - Tony Ngo
- Molecular Pharmacology Group, Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia; St Vincent's Clinical School, University of New South Wales, Darlinghurst, NSW 2010, Australia
| | - Nicola J Smith
- Molecular Pharmacology Group, Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia; St Vincent's Clinical School, University of New South Wales, Darlinghurst, NSW 2010, Australia.
| |
Collapse
|
32
|
Rea K, O'Mahony SM, Dinan TG, Cryan JF. The Role of the Gastrointestinal Microbiota in Visceral Pain. Handb Exp Pharmacol 2017; 239:269-287. [PMID: 28035535 DOI: 10.1007/164_2016_115] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
A growing body of preclinical and clinical evidence supports a relationship between the complexity and diversity of the microorganisms that inhabit our gut (human gastrointestinal microbiota) and health status. Under normal homeostatic conditions this microbial population helps maintain intestinal peristalsis, mucosal integrity, pH balance, immune priming and protection against invading pathogens. Furthermore, these microbes can influence centrally regulated emotional behaviour through mechanisms including microbially derived bioactive molecules (amino acid metabolites, short-chain fatty acids, neuropeptides and neurotransmitters), mucosal immune and enteroendocrine cell activation, as well as vagal nerve stimulation.The microbiota-gut-brain axis comprises a dynamic matrix of tissues and organs including the brain, autonomic nervous system, glands, gut, immune cells and gastrointestinal microbiota that communicate in a complex multidirectional manner to maintain homeostasis and resist perturbation to the system. Changes to the microbial environment, as a consequence of illness, stress or injury, can lead to a broad spectrum of physiological and behavioural effects locally including a decrease in gut barrier integrity, altered gut motility, inflammatory mediator release as well as nociceptive and distension receptor sensitisation. Centrally mediated events including hypothalamic-pituitary-adrenal (HPA) axis, neuroinflammatory events and neurotransmitter systems are concomitantly altered. Thus, both central and peripheral pathways associated with pain manifestation and perception are altered as a consequence of the microbiota-gut-brain axis imbalance.In this chapter the involvement of the gastrointestinal microbiota in visceral pain is reviewed. We focus on the anatomical and physiological nodes whereby microbiota may be mediating pain response, and address the potential for manipulating gastrointestinal microbiota as a therapeutic target for visceral pain.
Collapse
Affiliation(s)
- Kieran Rea
- APC Microbiome Institute, University College Cork, Cork, Ireland
| | - Siobhain M O'Mahony
- APC Microbiome Institute, University College Cork, Cork, Ireland
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - Timothy G Dinan
- APC Microbiome Institute, University College Cork, Cork, Ireland
- Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland
| | - John F Cryan
- APC Microbiome Institute, University College Cork, Cork, Ireland.
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland.
| |
Collapse
|
33
|
Tang W, Huang B, Wang J, An L, Zhong H, Yang H, Li P, Chen J. A label-free screening approach targeted protease-activated receptor 1 based on dynamic mass redistribution in living cells. RSC Adv 2017. [DOI: 10.1039/c7ra07927c] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Protease-activated receptor 1 (PAR-1) antagonists strongly inhibit thrombin-induced platelet aggregation and are proved to be effective as anti-thrombotic drugs.
Collapse
Affiliation(s)
- Weiwei Tang
- State Key Laboratory of Natural Medicines
- China Pharmaceutical University
- Nanjing 210009
- P. R. China
| | - Bixia Huang
- State Key Laboratory of Natural Medicines
- China Pharmaceutical University
- Nanjing 210009
- P. R. China
| | - Jiancheng Wang
- State Key Laboratory of Natural Medicines
- China Pharmaceutical University
- Nanjing 210009
- P. R. China
| | - Lin An
- State Key Laboratory of Natural Medicines
- China Pharmaceutical University
- Nanjing 210009
- P. R. China
| | | | - Hua Yang
- State Key Laboratory of Natural Medicines
- China Pharmaceutical University
- Nanjing 210009
- P. R. China
| | - Ping Li
- State Key Laboratory of Natural Medicines
- China Pharmaceutical University
- Nanjing 210009
- P. R. China
| | - Jun Chen
- State Key Laboratory of Natural Medicines
- China Pharmaceutical University
- Nanjing 210009
- P. R. China
| |
Collapse
|
34
|
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
|
35
|
Perazzo J, Lopes-Ferreira M, Sá Santos S, Serrano I, Pinto A, Lima C, Bardaji E, Tavares I, Heras M, Conceição K, Castanho MARB. Endothelium-Mediated Action of Analogues of the Endogenous Neuropeptide Kyotorphin (Tyrosil-Arginine): Mechanistic Insights from Permeation and Effects on Microcirculation. ACS Chem Neurosci 2016; 7:1130-40. [PMID: 27244291 DOI: 10.1021/acschemneuro.6b00099] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Kyotorphin (KTP) is an endogenous peptide with analgesic properties when administered into the central nervous system (CNS). Its amidated form (l-Tyr-l-Arg-NH2; KTP-NH2) has improved analgesic efficacy after systemic administration, suggesting blood-brain barrier (BBB) crossing. KTP-NH2 also has anti-inflammatory action impacting on microcirculation. In this work, selected derivatives of KTP-NH2 were synthesized to improve lipophilicity and resistance to enzymatic degradation while introducing only minor changes in the chemical structure: N-terminal methylation and/or use of d amino acid residues. Intravital microscopy data show that KTP-NH2 having a d-Tyr residue, KTP-NH2-DL, efficiently decreases the number of leukocyte rolling in a murine model of inflammation induced by bacterial lipopolysaccharide (LPS): down to 46% after 30 min with 96 μM KTP-NH2-DL. The same molecule has lower ability to permeate membranes (relative permeability of 0.38) and no significant activity in a behavioral test which evaluates thermal nociception (hot-plate test). On the contrary, methylated isomers at 96 μM increase leukocyte rolling up to nearly 5-fold after 30 min, suggesting a proinflammatory activity. They have maximal ability to permeate membranes (relative permeability of 0.8) and induce long-lasting antinociception.
Collapse
Affiliation(s)
- Juliana Perazzo
- Instituto de Medicina
Molecular, Faculdade de Medicina da Universidade de Lisboa, Av. Professor Egas Moniz, 1649-028 Lisboa, Portugal
| | - Mônica Lopes-Ferreira
- Unidade
de Imunorregulação, Laboratório Especial de Toxinologia
Aplicada, Instituto Butantan, Av. Vital Brasil, 1500 São Paulo, Brazil
| | - Sónia Sá Santos
- Instituto de Medicina
Molecular, Faculdade de Medicina da Universidade de Lisboa, Av. Professor Egas Moniz, 1649-028 Lisboa, Portugal
| | - Isa Serrano
- Instituto de Medicina
Molecular, Faculdade de Medicina da Universidade de Lisboa, Av. Professor Egas Moniz, 1649-028 Lisboa, Portugal
| | - Antónia Pinto
- Instituto de Medicina
Molecular, Faculdade de Medicina da Universidade de Lisboa, Av. Professor Egas Moniz, 1649-028 Lisboa, Portugal
| | - Carla Lima
- Unidade
de Imunorregulação, Laboratório Especial de Toxinologia
Aplicada, Instituto Butantan, Av. Vital Brasil, 1500 São Paulo, Brazil
| | - Eduard Bardaji
- Laboratori
d’Innovació en processos i Productes de Síntesi
Orgànica (LIPPSO), Department de Química, Universitat de Girona, Campus Montilivi, 17071 Girona, Spain
| | - Isaura Tavares
- Instituto de Biologia Molecular e Celular, Rua do Campo Alegre, 4150-180 Porto, Portugal
- i3S
- Instituto de Inovação e Investigação
em Saúde, and Departamento de Biologia Experimental, Faculdade
de Medicina, Universidade do Porto, 4099-002 Porto, Portugal
| | - Montserrat Heras
- Laboratori
d’Innovació en processos i Productes de Síntesi
Orgànica (LIPPSO), Department de Química, Universitat de Girona, Campus Montilivi, 17071 Girona, Spain
| | - Katia Conceição
- Departamento
de Ciência e Tecnologia, Universidade Federal de São Paulo, UNIFESP, Rua Talim, 330, 04021-001 São José dos Campos, Brazil
| | - Miguel A. R. B. Castanho
- Instituto de Medicina
Molecular, Faculdade de Medicina da Universidade de Lisboa, Av. Professor Egas Moniz, 1649-028 Lisboa, Portugal
| |
Collapse
|
36
|
Nasri I, Bonnet D, Zwarycz B, d'Aldebert E, Khou S, Mezghani-Jarraya R, Quaranta M, Rolland C, Bonnart C, Mas E, Ferrand A, Cenac N, Magness S, Van Landeghem L, Vergnolle N, Racaud-Sultan C. PAR2-dependent activation of GSK3β regulates the survival of colon stem/progenitor cells. Am J Physiol Gastrointest Liver Physiol 2016; 311:G221-36. [PMID: 27313176 PMCID: PMC5007290 DOI: 10.1152/ajpgi.00328.2015] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Accepted: 05/16/2016] [Indexed: 01/31/2023]
Abstract
Protease-activated receptors PAR1 and PAR2 play an important role in the control of epithelial cell proliferation and migration. However, the survival of normal and tumor intestinal stem/progenitor cells promoted by proinflammatory mediators may be critical in oncogenesis. The glycogen synthase kinase-3β (GSK3β) pathway is overactivated in colon cancer cells and promotes their survival and drug resistance. We thus aimed to determine PAR1 and PAR2 effects on normal and tumor intestinal stem/progenitor cells and whether they involved GSK3β. First, PAR1 and PAR2 were identified in colon stem/progenitor cells by immunofluorescence. In three-dimensional cultures of murine crypt units or single tumor Caco-2 cells, PAR2 activation decreased numbers and size of normal or cancerous spheroids, and PAR2-deficient spheroids showed increased proliferation, indicating that PAR2 represses proliferation. PAR2-stimulated normal cells were more resistant to stress (serum starvation or spheroid passaging), suggesting prosurvival effects of PAR2 Accordingly, active caspase-3 was strongly increased in PAR2-deficient normal spheroids. PAR2 but not PAR1 triggered GSK3β activation through serine-9 dephosphorylation in normal and tumor cells. The PAR2-triggered GSK3β activation implicates an arrestin/PP2A/GSK3β complex that is dependent on the Rho kinase activity. Loss of PAR2 was associated with high levels of GSK3β nonactive form, strengthening the role of PAR2 in GSK3β activation. GSK3 pharmacological inhibition impaired the survival of PAR2-stimulated spheroids and serum-starved cells. Altogether our data identify PAR2/GSK3β as a novel pathway that plays a critical role in the regulation of stem/progenitor cell survival and proliferation in normal colon crypts and colon cancer.
Collapse
Affiliation(s)
- Imen Nasri
- 1Institut de Recherche en Santé Digestive, Université de Toulouse, Institut National de la Santé et de la Recherche Médicale, Institut National de la Recherche Agronomique, Ecole Nationale Vétérinaire de Toulouse, Université Paul Sabatier, Toulouse, France; ,2Laboratoire de Chimie des Substances Naturelles, Faculté des Sciences de Sfax, Université de Sfax, Sfax, Tunisia;
| | - Delphine Bonnet
- 1Institut de Recherche en Santé Digestive, Université de Toulouse, Institut National de la Santé et de la Recherche Médicale, Institut National de la Recherche Agronomique, Ecole Nationale Vétérinaire de Toulouse, Université Paul Sabatier, Toulouse, France; ,3Service de Médecine Interne, Fédération Digestive, Centre Hospitalier Universitaire Purpan, Toulouse, France;
| | - Bailey Zwarycz
- 4Departments of Medicine and Cell Biology & Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina;
| | - Emilie d'Aldebert
- 1Institut de Recherche en Santé Digestive, Université de Toulouse, Institut National de la Santé et de la Recherche Médicale, Institut National de la Recherche Agronomique, Ecole Nationale Vétérinaire de Toulouse, Université Paul Sabatier, Toulouse, France;
| | - Sokchea Khou
- 1Institut de Recherche en Santé Digestive, Université de Toulouse, Institut National de la Santé et de la Recherche Médicale, Institut National de la Recherche Agronomique, Ecole Nationale Vétérinaire de Toulouse, Université Paul Sabatier, Toulouse, France;
| | - Raoudha Mezghani-Jarraya
- 2Laboratoire de Chimie des Substances Naturelles, Faculté des Sciences de Sfax, Université de Sfax, Sfax, Tunisia;
| | - Muriel Quaranta
- 1Institut de Recherche en Santé Digestive, Université de Toulouse, Institut National de la Santé et de la Recherche Médicale, Institut National de la Recherche Agronomique, Ecole Nationale Vétérinaire de Toulouse, Université Paul Sabatier, Toulouse, France;
| | - Corinne Rolland
- 1Institut de Recherche en Santé Digestive, Université de Toulouse, Institut National de la Santé et de la Recherche Médicale, Institut National de la Recherche Agronomique, Ecole Nationale Vétérinaire de Toulouse, Université Paul Sabatier, Toulouse, France;
| | - Chrystelle Bonnart
- 1Institut de Recherche en Santé Digestive, Université de Toulouse, Institut National de la Santé et de la Recherche Médicale, Institut National de la Recherche Agronomique, Ecole Nationale Vétérinaire de Toulouse, Université Paul Sabatier, Toulouse, France;
| | - Emmanuel Mas
- 1Institut de Recherche en Santé Digestive, Université de Toulouse, Institut National de la Santé et de la Recherche Médicale, Institut National de la Recherche Agronomique, Ecole Nationale Vétérinaire de Toulouse, Université Paul Sabatier, Toulouse, France; ,5Service de Gastroentérologie, Hépatologie et Nutrition, Hôpital des Enfants, Toulouse, France; and
| | - Audrey Ferrand
- 1Institut de Recherche en Santé Digestive, Université de Toulouse, Institut National de la Santé et de la Recherche Médicale, Institut National de la Recherche Agronomique, Ecole Nationale Vétérinaire de Toulouse, Université Paul Sabatier, Toulouse, France;
| | - Nicolas Cenac
- 1Institut de Recherche en Santé Digestive, Université de Toulouse, Institut National de la Santé et de la Recherche Médicale, Institut National de la Recherche Agronomique, Ecole Nationale Vétérinaire de Toulouse, Université Paul Sabatier, Toulouse, France;
| | - Scott Magness
- 4Departments of Medicine and Cell Biology & Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina;
| | - Laurianne Van Landeghem
- 6Institut National de la Santé et de la Recherche Médicale U913, Université de Nantes, Institut des Maladies de l'Appareil Digestif, Nantes, France
| | - Nathalie Vergnolle
- 1Institut de Recherche en Santé Digestive, Université de Toulouse, Institut National de la Santé et de la Recherche Médicale, Institut National de la Recherche Agronomique, Ecole Nationale Vétérinaire de Toulouse, Université Paul Sabatier, Toulouse, France;
| | - Claire Racaud-Sultan
- Institut de Recherche en Santé Digestive, Université de Toulouse, Institut National de la Santé et de la Recherche Médicale, Institut National de la Recherche Agronomique, Ecole Nationale Vétérinaire de Toulouse, Université Paul Sabatier, Toulouse, France;
| |
Collapse
|
37
|
Neuroimmunomodulation in the Gut: Focus on Inflammatory Bowel Disease. Mediators Inflamm 2016; 2016:1363818. [PMID: 27471349 PMCID: PMC4947661 DOI: 10.1155/2016/1363818] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Accepted: 06/07/2016] [Indexed: 12/17/2022] Open
Abstract
Intestinal immunity is finely regulated by several concomitant and overlapping mechanisms, in order to efficiently sense external stimuli and mount an adequate response of either tolerance or defense. In this context, a complex interplay between immune and nonimmune cells is responsible for the maintenance of normal homeostasis. However, in certain conditions, the disruption of such an intricate network may result in intestinal inflammation, including inflammatory bowel disease (IBD). IBD is believed to result from a combination of genetic and environmental factors acting in concert with an inappropriate immune response, which in turn interacts with nonimmune cells, including nervous system components. Currently, evidence shows that the interaction between the immune and the nervous system is bidirectional and plays a critical role in the regulation of intestinal inflammation. Recently, the maintenance of intestinal homeostasis has been shown to be under the reciprocal control of the microbiota by immune mechanisms, whereas intestinal microorganisms can modulate mucosal immunity. Therefore, in addition to presenting the mechanisms underlying the interaction between immune and nervous systems in the gut, here we discuss the role of the microbiota also in the regulation of neuroimmune crosstalk involved in intestinal homeostasis and inflammation, with potential implications to IBD pathogenesis.
Collapse
|
38
|
Abstract
The GI tract is the most exposed organ to proteases, both in physiological and pathophysiological conditions. For digestive purposes, the lumen of the upper GI tract contains large amounts of pancreatic proteases, but studies have also demonstrated increased proteolytic activity into mucosal tissues (both in the upper and lower GI tract), associated with pathological conditions. This review aims at outlining the evidences for dysregulated proteolytic homeostasis in GI diseases and the pathogenic mechanisms of increased proteolytic activity. The therapeutic potential of protease inhibition in GI diseases is discussed, with a particular focus on IBDs, functional GI disorders and colorectal cancer.
Collapse
Affiliation(s)
- Nathalie Vergnolle
- Inserm, U1220, Toulouse, France,Université de Toulouse, Université Paul Sabatier, Institut de Recherche en Santé Digestive (IRSD), Toulouse, France,Inra, U1416, Toulouse, France,Ecole Nationale Vétérinaire de Toulouse (ENVT), France,Department of Pharmacology and Physiology, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
39
|
Monjotin N, Gillespie J, Farrié M, Le Grand B, Junquero D, Vergnolle N. F16357, a novel protease-activated receptor 1 antagonist, improves urodynamic parameters in a rat model of interstitial cystitis. Br J Pharmacol 2016; 173:2224-36. [PMID: 27111354 PMCID: PMC4919585 DOI: 10.1111/bph.13501] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Revised: 03/17/2016] [Accepted: 04/10/2016] [Indexed: 01/15/2023] Open
Abstract
Background and Purpose The aims of the present study were to characterize the role of PAR1 in rat bladder under inflammatory conditions and determine whether a selective PAR1 antagonist, F16357, can prevent the pathophysiological symptoms of cyclophosphamide‐induced interstitial cystitis (IC). Experimental Approach Immunohistochemistry, contractile activity in isolated bladder and urodynamics were determined before and after cyclophosphamide treatment. F16357 was administered intravesically during the acute phase of inflammation, and effects on PAR1 and PAR1‐related bladder contraction evaluated 24 h after cyclophosphamide injection. Urodynamics and associated voided volumes were recorded 7 and 24 h after cyclophosphamide. Key Results In control conditions, PAR1 was present only in some umbrella cells. Cyclophosphamide disrupted the urothelium and expression of PAR1 by all remaining urothelial cells. After F16357 treatment, urothelial damage was absent and PAR1 immunoreactivity similar to control tissues. Thrombin and TFLLR‐NH2 induced bladder contractions. These were increased in inflammatory conditions and antagonized by F16357 in a concentration‐dependent manner. In telemetric experiments, furosemide increased urine production and voiding frequency for 60 min, 7 h after cyclophosphamide injection. Intravesical administration of F16357 blocked these changes with a return to a physiological profile; 24 h after cyclophosphamide, the volume of micturition was still lower with no increase in number of micturitions. F16357 30 μM reduced the number of micturitions and improved bladder capacity, but did not affect diuresis. Under similar experimental conditions, lidocaine 2% induced comparable effects. Conclusions and Implications PAR1 is expressed in rat bladder, overactivated in inflammatory conditions and involved in bladder function and sensation. F16357 could represent an interesting candidate for IC treatment.
Collapse
Affiliation(s)
- N Monjotin
- Institut de Recherche Pierre Fabre, Castres, France
| | - J Gillespie
- Newcastle University, Newcastle upon Tyne, England
| | - M Farrié
- Institut de Recherche Pierre Fabre, Castres, France
| | - B Le Grand
- Institut de Recherche Pierre Fabre, Castres, France
| | - D Junquero
- Institut de Recherche Pierre Fabre, Castres, France
| | - N Vergnolle
- IRSD, Université de Toulouse, INSERM, INRA, INP-ENVT, Université de Toulouse-3 Paul Sabatier, Toulouse, France
| |
Collapse
|
40
|
Tulic MK, Vivinus-Nébot M, Rekima A, Rabelo Medeiros S, Bonnart C, Shi H, Walker A, Dainese R, Boyer J, Vergnolle N, Piche T, Verhasselt V. Presence of commensal house dust mite allergen in human gastrointestinal tract: a potential contributor to intestinal barrier dysfunction. Gut 2016; 65:757-66. [PMID: 26646935 DOI: 10.1136/gutjnl-2015-310523] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Accepted: 11/05/2015] [Indexed: 12/15/2022]
Abstract
BACKGROUND Abnormal gut barrier function is the basis of gut inflammatory disease. It is known that house dust mite (HDM) aero-allergens induce inflammation in respiratory mucosa. We have recently reported allergen from Dermatophagoides pteronyssinus (Der p1) to be present in rodent gut. OBJECTIVE To examine whether Der p1 is present in human gut and to assess its effect on gut barrier function and inflammation. DESIGN Colonic biopsies, gut fluid, serum and stool were collected from healthy adults during endoscopy. Der p1 was measured by ELISA. Effect of HDM was assessed on gut permeability, tight-junction and mucin expression, and cytokine production, in presence or absence of cysteine protease inhibitors or serine protease inhibitors. In vivo effect of HDM was examined in mice given oral HDM or protease-neutralised HDM. Role of HDM in low-grade inflammation was studied in patients with IBS. RESULTS HDM Der p1 was detected in the human gut. In colonic biopsies from healthy patients, HDM increased epithelial permeability (p<0.001), reduced expression of tight-junction proteins and mucus barrier. These effects were associated with increased tumour necrosis factor (TNF)-α and interleukin (IL)-10 production and were abolished by cysteine-protease inhibitor (p<0.01). HDM effects did not require Th2 immunity. Results were confirmed in vivo in mice. In patients with IBS, HDM further deteriorated gut barrier function, induced TNF-α but failed to induce IL-10 secretion (p<0.001). CONCLUSIONS HDM, a ubiquitous environmental factor, is present in the human gut where it directly affects gut function through its proteolytic activity. HDM may be an important trigger of gut dysfunction and warrants further investigation.
Collapse
Affiliation(s)
- Meri K Tulic
- Université de Nice Sophia-Antipolis, EA 6302 Immune Tolerance (TIM), Nice, France The International Inflammation 'in-FLAME' Network, Worldwide Universities Network
| | - Mylene Vivinus-Nébot
- Université de Nice Sophia-Antipolis, EA 6302 Immune Tolerance (TIM), Nice, France Department of Immunology, Hôpital Archet 1, CHU de Nice, Université de Nice Sophia-Antipolis, Nice, France
| | - Akila Rekima
- Université de Nice Sophia-Antipolis, EA 6302 Immune Tolerance (TIM), Nice, France
| | - Samara Rabelo Medeiros
- Université de Nice Sophia-Antipolis, EA 6302 Immune Tolerance (TIM), Nice, France Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Chrystelle Bonnart
- INSERM U1043, Centre de Physiopathologie de Toulouse Purpan (CPTP), Toulouse, France CNRS, U5282, Toulouse, France Université de Toulouse, Site Paul Sabatier (UPS), Toulouse, France
| | - Haining Shi
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital, Charlestown, Massachusetts, USA
| | - Allan Walker
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital, Charlestown, Massachusetts, USA
| | - Raffaella Dainese
- Université de Nice Sophia-Antipolis, EA 6302 Immune Tolerance (TIM), Nice, France Department of Gastroenterology and Nutrition, Hôpital Archet 2, CHU de Nice, Université de Nice Sophia-Antipolis, Nice, France
| | - Julien Boyer
- Université de Nice Sophia-Antipolis, EA 6302 Immune Tolerance (TIM), Nice, France Department of Gastroenterology and Nutrition, Hôpital Archet 2, CHU de Nice, Université de Nice Sophia-Antipolis, Nice, France
| | - Nathalie Vergnolle
- INSERM U1043, Centre de Physiopathologie de Toulouse Purpan (CPTP), Toulouse, France CNRS, U5282, Toulouse, France Université de Toulouse, Site Paul Sabatier (UPS), Toulouse, France
| | - Thierry Piche
- Université de Nice Sophia-Antipolis, EA 6302 Immune Tolerance (TIM), Nice, France Department of Gastroenterology and Nutrition, Hôpital Archet 2, CHU de Nice, Université de Nice Sophia-Antipolis, Nice, France
| | - Valérie Verhasselt
- Université de Nice Sophia-Antipolis, EA 6302 Immune Tolerance (TIM), Nice, France The International Inflammation 'in-FLAME' Network, Worldwide Universities Network
| |
Collapse
|
41
|
Moloney RD, Johnson AC, O'Mahony SM, Dinan TG, Greenwood‐Van Meerveld B, Cryan JF. Stress and the Microbiota-Gut-Brain Axis in Visceral Pain: Relevance to Irritable Bowel Syndrome. CNS Neurosci Ther 2016; 22:102-17. [PMID: 26662472 PMCID: PMC6492884 DOI: 10.1111/cns.12490] [Citation(s) in RCA: 236] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Revised: 11/05/2015] [Accepted: 11/05/2015] [Indexed: 02/06/2023] Open
Abstract
Visceral pain is a global term used to describe pain originating from the internal organs of the body, which affects a significant proportion of the population and is a common feature of functional gastrointestinal disorders (FGIDs) such as irritable bowel syndrome (IBS). While IBS is multifactorial, with no single etiology to completely explain the disorder, many patients also experience comorbid behavioral disorders, such as anxiety or depression; thus, IBS is described as a disorder of the gut-brain axis. Stress is implicated in the development and exacerbation of visceral pain disorders. Chronic stress can modify central pain circuitry, as well as change motility and permeability throughout the gastrointestinal (GI) tract. More recently, the role of the gut microbiota in the bidirectional communication along the gut-brain axis, and subsequent changes in behavior, has emerged. Thus, stress and the gut microbiota can interact through complementary or opposing factors to influence visceral nociceptive behaviors. This review will highlight the evidence by which stress and the gut microbiota interact in the regulation of visceral nociception. We will focus on the influence of stress on the microbiota and the mechanisms by which microbiota can affect the stress response and behavioral outcomes with an emphasis on visceral pain.
Collapse
Affiliation(s)
- Rachel D. Moloney
- Laboratory of NeurogastroenterologyAPC Microbiome InstituteUniversity College CorkCorkIreland
- Present address:
Oklahoma Center for NeuroscienceUniversity of Oklahoma Health Science CenterOklahoma CityOKUSA
| | - Anthony C. Johnson
- Oklahoma Center for NeuroscienceUniversity of Oklahoma Health Science CenterOklahoma CityOKUSA
| | - Siobhain M. O'Mahony
- Laboratory of NeurogastroenterologyAPC Microbiome InstituteUniversity College CorkCorkIreland
- Department of Anatomy and NeuroscienceUniversity College CorkCorkIreland
| | - Timothy G. Dinan
- Laboratory of NeurogastroenterologyAPC Microbiome InstituteUniversity College CorkCorkIreland
- Department of Psychiatry and Neurobehavioural ScienceUniversity College CorkCorkIreland
| | - Beverley Greenwood‐Van Meerveld
- Oklahoma Center for NeuroscienceUniversity of Oklahoma Health Science CenterOklahoma CityOKUSA
- V.A. Medical CenterOklahoma CityOKUSA
| | - John F. Cryan
- Laboratory of NeurogastroenterologyAPC Microbiome InstituteUniversity College CorkCorkIreland
- Department of Anatomy and NeuroscienceUniversity College CorkCorkIreland
| |
Collapse
|
42
|
Jin M, Yang HW, Tao AL, Wei JF. Evolution of the protease-activated receptor family in vertebrates. Int J Mol Med 2016; 37:593-602. [PMID: 26820116 PMCID: PMC4771116 DOI: 10.3892/ijmm.2016.2464] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Accepted: 01/13/2016] [Indexed: 01/18/2023] Open
Abstract
Belonging to the G protein-coupled receptor (GPcr) family, the protease-activated receptors (Pars) consist of 4 members, PAR1-4. PARs mediate the activation of cells via thrombin, serine and other proteases. Such protease-triggered signaling events are thought to be critical for hemostasis, thrombosis and other normal pathological processes. In the present study, we examined the evolution of PARs by analyzing phylogenetic trees, chromosome location, selective pressure and functional divergence based on the 169 functional gene alignment sequences from 57 vertebrate gene sequences. We found that the 4 Pars originated from 4 invertebrate ancestors by phylogenetic trees analysis. The selective pressure results revealed that only PAR1 appeared by positive selection during its evolution, while the other PAR members did not. In addition, we noticed that although these PARs evolved separately, the results of functional divergence indicated that their evolutional rates were similar and their functions did not significantly diverge. The findings of our study provide valuable insight into the evolutionary history of the vertebrate PAR family.
Collapse
Affiliation(s)
- Min Jin
- Guangdong Provincial Key Laboratory of Allergy and Clinical Immunology, The State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510260, P.R. China
| | - Hai-Wei Yang
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Ai-Lin Tao
- Guangdong Provincial Key Laboratory of Allergy and Clinical Immunology, The State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510260, P.R. China
| | - Ji-Fu Wei
- Guangdong Provincial Key Laboratory of Allergy and Clinical Immunology, The State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510260, P.R. China
| |
Collapse
|
43
|
Abreu IS, Euzebio Alves VT, Benedete APS, Bueno da Silva HA, França BN, Saraiva L, Lima LA, Carvalho MH, Holzhausen M. Gingival crevicular fluid levels of protease-activated receptors type 1 and type 2 in diabetic patients with periodontitis. J Periodontal Res 2015; 51:577-85. [PMID: 26564991 DOI: 10.1111/jre.12336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/25/2015] [Indexed: 11/28/2022]
Abstract
BACKGROUND AND OBJECTIVE Protease activated receptor type 1 (PAR1 ) seems to play a role in periodontal repair, while PAR2 is associated with periodontal inflammation. As diabetes is a known risk factor for periodontal disease, the aim of this study was to evaluate the influence of type 2 diabetes on PAR1 and PAR2 mRNA expression in the gingival crevicular fluid of patients with chronic periodontitis before and after non-surgical periodontal treatment. MATERIAL AND METHODS Gingival crevicular fluid samples and clinical parameters consisting of measuring probing depth, clinical attachment level, bleeding on probing and plaque index were collected from systemically healthy patients and patients with type 2 diabetes and chronic periodontitis, at baseline and after non-surgical periodontal therapy. PAR1 and PAR2 , as well as the presence of the proteases RgpB gingipain and neutrophil proteinase-3 were assessed by quantitative polymerase chain reaction in the gingival crevicular fluid. RESULTS The periodontal clinical parameters significantly improved after periodontal therapy (p < 0.01). Diabetes led to increased expression of PAR1 in gingival crevicular fluid, and in the presence of chronic periodontitis, it significantly decreased the expression of PAR1 and PAR2 (p < 0.05). Moreover, non-surgical periodontal treatment in diabetics resulted in increased expression of PAR1 and PAR2 (p < 0.05), and decreased expression of RgpB gingipain and proteinase-3 (p < 0.05). CONCLUSION The present data demonstrated that diabetes was associated with an altered expression of PAR1 and PAR2 in the gingival crevicular fluid cells of subjects with chronic periodontitis. Future studies are necessary to elucidate the effects of PAR1 upregulation in periodontally healthy sites and PAR2 downregulation in chronic periodontitis sites on the increased susceptibility and severity of periodontitis in diabetes.
Collapse
Affiliation(s)
- I S Abreu
- Division of Periodontics, Department of Stomatology, School of Dentistry, University of São Paulo, São Paulo, SP, Brazil
| | - V T Euzebio Alves
- Division of Periodontics, Department of Stomatology, School of Dentistry, University of São Paulo, São Paulo, SP, Brazil
| | - A P S Benedete
- Division of Periodontics, Department of Stomatology, School of Dentistry, University of São Paulo, São Paulo, SP, Brazil
| | - H A Bueno da Silva
- Division of Periodontics, Department of Stomatology, School of Dentistry, University of São Paulo, São Paulo, SP, Brazil
| | - B N França
- Division of Periodontics, Department of Stomatology, School of Dentistry, University of São Paulo, São Paulo, SP, Brazil
| | - L Saraiva
- Division of Periodontics, Department of Stomatology, School of Dentistry, University of São Paulo, São Paulo, SP, Brazil
| | - L A Lima
- Division of Periodontics, Department of Stomatology, School of Dentistry, University of São Paulo, São Paulo, SP, Brazil
| | - M H Carvalho
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - M Holzhausen
- Division of Periodontics, Department of Stomatology, School of Dentistry, University of São Paulo, São Paulo, SP, Brazil
| |
Collapse
|
44
|
Patricio ES, Costa R, Figueiredo CP, Gers-Barlag K, Bicca MA, Manjavachi MN, Segat GC, Gentry C, Luiz AP, Fernandes ES, Cunha TM, Bevan S, Calixto JB. Mechanisms Underlying the Scratching Behavior Induced by the Activation of Proteinase-Activated Receptor-4 in Mice. J Invest Dermatol 2015; 135:2484-2491. [DOI: 10.1038/jid.2015.183] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Revised: 04/20/2015] [Accepted: 04/27/2015] [Indexed: 02/08/2023]
|
45
|
Boitano S, Hoffman J, Flynn AN, Asiedu MN, Tillu DV, Zhang Z, Sherwood CL, Rivas CM, DeFea KA, Vagner J, Price TJ. The novel PAR2 ligand C391 blocks multiple PAR2 signalling pathways in vitro and in vivo. Br J Pharmacol 2015; 172:4535-4545. [PMID: 26140338 DOI: 10.1111/bph.13238] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Revised: 06/18/2015] [Accepted: 06/28/2015] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND AND PURPOSE Proteinase-activated receptor-2 (PAR2) is a GPCR linked to diverse pathologies, including acute and chronic pain. PAR2 is one of the four PARs that are activated by proteolytic cleavage of the extracellular amino terminus, resulting in an exposed, tethered peptide agonist. Several peptide and peptidomimetic agonists, with high potency and efficacy, have been developed to probe the functions of PAR2, in vitro and in vivo. However, few similarly potent and effective antagonists have been described. EXPERIMENTAL APPROACH We modified the peptidomimetic PAR2 agonist, 2-furoyl-LIGRLO-NH2 , to create a novel PAR2 peptidomimetic ligand, C391. C391 was evaluated for PAR2 agonist/antagonist activity to PAR2 across Gq signalling pathways using the naturally expressing PAR2 cell line 16HBE14o-. For antagonist studies, a highly potent and specific peptidomimetic agonist (2-aminothiazo-4-yl-LIGRL-NH2 ) and proteinase agonist (trypsin) were used to activate PAR2. C391 was also evaluated in vivo for reduction of thermal hyperalgesia, mediated by mast cell degranulation, in mice. KEY RESULTS C391 is a potent and specific peptidomimetic antagonist, blocking multiple signalling pathways (Gq -dependent Ca2+ , MAPK) induced following peptidomimetic or proteinase activation of human PAR2. In a PAR2-dependent behavioural assay in mice, C391 dose-dependently (75 μg maximum effect) blocked the thermal hyperalgesia, mediated by mast cell degranulation. CONCLUSIONS AND IMPLICATIONS C391 is the first low MW antagonist to block both PAR2 Ca2+ and MAPK signalling pathways activated by peptidomimetics and/or proteinase activation. C391 represents a new molecular structure for PAR2 antagonism and can serve as a basis for further development for this important therapeutic target.
Collapse
Affiliation(s)
- Scott Boitano
- Arizona Respiratory Center and Department of Physiology, University of Arizona, Tucson, AZ, USA.,The BIO5 Collaborative Research Institute, University of Arizona, Tucson, AZ, USA
| | - Justin Hoffman
- Arizona Respiratory Center and Department of Physiology, University of Arizona, Tucson, AZ, USA.,The BIO5 Collaborative Research Institute, University of Arizona, Tucson, AZ, USA
| | - Andrea N Flynn
- Arizona Respiratory Center and Department of Physiology, University of Arizona, Tucson, AZ, USA.,The BIO5 Collaborative Research Institute, University of Arizona, Tucson, AZ, USA
| | - Marina N Asiedu
- Department of Pharmacology, University of Arizona, Tucson, AZ, USA
| | - Dipti V Tillu
- Department of Pharmacology, University of Arizona, Tucson, AZ, USA
| | - Zhenyu Zhang
- The BIO5 Collaborative Research Institute, University of Arizona, Tucson, AZ, USA
| | - Cara L Sherwood
- Arizona Respiratory Center and Department of Physiology, University of Arizona, Tucson, AZ, USA.,The BIO5 Collaborative Research Institute, University of Arizona, Tucson, AZ, USA
| | - Candy M Rivas
- Arizona Respiratory Center and Department of Physiology, University of Arizona, Tucson, AZ, USA.,The BIO5 Collaborative Research Institute, University of Arizona, Tucson, AZ, USA.,Graduate Interdisciplinary Program in Physiological Sciences, University of Arizona, Tucson, AZ, USA
| | - Kathryn A DeFea
- Biomedical Sciences Division, University of California, Riverside, CA, USA
| | - Josef Vagner
- The BIO5 Collaborative Research Institute, University of Arizona, Tucson, AZ, USA
| | - Theodore J Price
- The BIO5 Collaborative Research Institute, University of Arizona, Tucson, AZ, USA.,Department of Pharmacology, University of Arizona, Tucson, AZ, USA.,School of Behavioral and Brain Sciences, University of Texas at Dallas, Dallas, TX, USA
| |
Collapse
|
46
|
Hurley MJ, Durrenberger PF, Gentleman SM, Walls AF, Dexter DT. Altered Expression of Brain Proteinase-Activated Receptor-2, Trypsin-2 and Serpin Proteinase Inhibitors in Parkinson's Disease. J Mol Neurosci 2015; 57:48-62. [PMID: 25982926 DOI: 10.1007/s12031-015-0576-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Accepted: 05/05/2015] [Indexed: 10/23/2022]
Abstract
Neuroinflammation is thought to contribute to cell death in neurodegenerative disorders, but the factors involved in the inflammatory process are not completely understood. Proteinase-activated receptor-2 (PAR2) expression in brain is increased in Alzheimer's disease and multiple sclerosis, but the status of PAR2 in Parkinson's disease is unknown. This study examined expression of PAR2 and endogenous proteinase activators (trypsin-2, mast cell tryptase) and proteinase inhibitors (serpin-A5, serpin-A13) in areas vulnerable and resistant to neurodegeneration in Parkinson's disease at different Braak α-synuclein stages of the disease in post-mortem brain. In normal aged brain, expression of PAR-2, trypsin-2, and serpin-A5 and serpin-A13 was found in neurons and microglia, and alterations in the amount of immunoreactivity for these proteins were found in some brain regions. Namely, there was a decrease in neurons positive for serpin-A5 in the dorsal motor nucleus, and serpin-A13 expression was reduced in the locus coeruleus and primary motor cortex, while expression of PAR2, trypsin-2 and both serpins was reduced in neurons within the substantia nigra. There was an increased number of microglia that expressed serpin-A5 in the dorsal motor nucleus of vagus and elevated numbers of microglia that expressed serpin-A13 in the substantia nigra of late Parkinson's disease cases. The number of microglia that expressed trypsin-2 increased in primary motor cortex of incidental Lewy body disease cases. Analysis of Parkinson's disease cases alone indicated that serpin-A5 and serpin-A13, and trypsin-2 expression in midbrain and cerebral cortex was different in cases with a high incidence of L-DOPA-induced dyskinesia and psychosis compared to those with low levels of these treatment-induced side effects. This study showed that there was altered expression in brain of PAR2 and some proteins that can control its function in Parkinson's disease. Given the role of PAR2 in neuroinflammation, drugs that mitigate these changes may be neuroprotective when administered to patients with Parkinson's disease.
Collapse
Affiliation(s)
- Michael J Hurley
- Centre for Neuroinflammation and Neurodegeneration, Division of Brain Sciences, Department of Medicine, Imperial College London, Hammersmith Campus, Du Cane Road, London, W12 0NN, UK,
| | | | | | | | | |
Collapse
|
47
|
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
|
48
|
Charest-Morin X, Raghavan A, Charles ML, Kolodka T, Bouthillier J, Jean M, Robbins MS, Marceau F. Pharmacological effects of recombinant human tissue kallikrein on bradykinin B2 receptors. Pharmacol Res Perspect 2015; 3:e00119. [PMID: 26038695 PMCID: PMC4448978 DOI: 10.1002/prp2.119] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Revised: 09/23/2014] [Accepted: 09/26/2014] [Indexed: 12/13/2022] Open
Abstract
Tissue kallikrein (KLK-1), a serine protease, initiates the release of bradykinin (BK)-related peptides from low-molecular weight kininogen. KLK-1 and the BK B2 receptor (B2R) mediate beneficial effects on the progression of type 2 diabetes and renal disease, but the precise role of KLK-1 independent of its kinin-forming activity remains unclear. We used DM199, a recombinant form of human KLK-1, along with the isolated human umbilical vein, a robust bioassay of the B2R, to address the previous claims that KLK-1 directly binds to and activates the human B2R, with possible receptor cleavage. DM199 (1–10 nmol/L) contracted the isolated vein via the B2R, but in a tachyphylactic, kinin-dependent manner, without desensitization of the tissue to exogenously added BK. In binding experiments with recombinant N-terminally tagged myc-B2Rs expressed in HEK 293a cells, DM199 displaced [3H]BK binding from the rabbit myc-B2R, but not from the human or rat myc-B2Rs. No evidence of myc-B2R degradation by immunoblot analysis was apparent following treatment of these 3 myc-B2R constructs with DM199 (30 min, ≤10 nmol/L). In HEK 293 cells stably expressing rabbit B2R-GFP, DM199 (11–108 pmol/L) elicited signaling-dependent endocytosis and reexpression, while a higher concentration (1.1 nmol/L) induced a partially irreversible endocytosis of the construct (microscopy), paralleled by the appearance of free GFP in cells (immunoblotting, indicative of incomplete receptor down-regulation). The pharmacology of DM199 at relevant concentrations (<10 nmol/L) is essentially based on the activity of locally generated kinins. Binding to and mild down-regulation of the B2R is possibly a species-dependent idiosyncratic response to DM199.
Collapse
Affiliation(s)
- Xavier Charest-Morin
- Centre de recherche en rhumatologie et immunologie, CHU de Québec Québec City, Québec, Canada, G1V 4G2
| | - Arvind Raghavan
- DiaMedica Inc. One Carlson Parkway, Suite 124, Minneapolis, Minnesota, 55447
| | - Matthew L Charles
- DiaMedica Inc. One Carlson Parkway, Suite 124, Minneapolis, Minnesota, 55447
| | - Tadeusz Kolodka
- DiaMedica Inc. One Carlson Parkway, Suite 124, Minneapolis, Minnesota, 55447
| | - Johanne Bouthillier
- Centre de recherche en rhumatologie et immunologie, CHU de Québec Québec City, Québec, Canada, G1V 4G2
| | - Mélissa Jean
- Centre de recherche en rhumatologie et immunologie, CHU de Québec Québec City, Québec, Canada, G1V 4G2
| | - Mark S Robbins
- DiaMedica Inc. One Carlson Parkway, Suite 124, Minneapolis, Minnesota, 55447
| | - François Marceau
- Centre de recherche en rhumatologie et immunologie, CHU de Québec Québec City, Québec, Canada, G1V 4G2
| |
Collapse
|
49
|
An Introduction to Pain Pathways and Pain “Targets”. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2015; 131:1-30. [DOI: 10.1016/bs.pmbts.2015.01.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
50
|
Hollenberg MD. Proteinases, their receptors and inflammatory signalling: the Oxford South Parks Road connection. Br J Pharmacol 2014; 172:3196-211. [PMID: 25521749 DOI: 10.1111/bph.13041] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Revised: 11/18/2014] [Accepted: 12/07/2014] [Indexed: 01/22/2023] Open
Abstract
In keeping with the aim of the Paton Memorial Lecture to 'facilitate the historical study of pharmacology', this overview, which is my distinct honour to write, represents a 'Janus-like' personal perspective looking both backwards and forwards at the birth and growth of 'receptor molecular pharmacology' with special relevance to inflammatory diseases. The overview begins in the Oxford Department of Pharmacology in the mid-1960s and then goes on to provide a current perspective of signalling by proteinases. Looking backwards, the synopsis describes the fruitful Oxford Pharmacology Department infrastructure that Bill Paton generated in keeping with the blueprint begun by his predecessor, J H Burn. Looking forwards, the overview illustrates the legacy of that environment in generating some of the first receptor ligand-binding data and providing the inspiration and vision for those like me who were training in the department at the same time. With apologies, I mention only in passing a number of individuals who benefitted from the 'South Parks Road connection' using myself as one of the 'outcome study' examples. It is also by looking forward that I can meet the complementary aim of summarizing the lecture presented at a 'BPS 2014 Focused Meeting on Cell Signalling' to provide an overview of the role of proteinases and their signalling mechanisms in the setting of inflammation.
Collapse
Affiliation(s)
- M D Hollenberg
- Inflammation Research Network-Snyder Institute for Chronic Disease, Department of Physiology and Pharmacology and Department of Medicine, University of Calgary Cumming School of Medicine, Calgary, AB, Canada
| |
Collapse
|