1
|
Sekiguchi F, Koike N, Shimada Y, Sugimoto K, Masuda H, Nakamura T, Yamaguchi H, Tanabe G, Marumoto S, Kasanami Y, Tsubota M, Ohkubo T, Yoshida S, Kawabata A. A hydrolysate of poly-trans-[(2-carboxyethyl)germasesquioxane] (Ge-132) suppresses Ca v3.2-dependent pain by sequestering exogenous and endogenous sulfide. Redox Biol 2023; 59:102579. [PMID: 36563535 PMCID: PMC9800310 DOI: 10.1016/j.redox.2022.102579] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 12/05/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022] Open
Abstract
Poly-trans-[(2-carboxyethyl)germasesquioxane] (Ge-132), an organogermanium, is hydrolyzed to 3-(trihydroxygermyl)propanoic acid (THGP) in aqueous solutions, and reduces inflammation, pain and cancer, whereas the underlying mechanisms remain unknown. Sulfides including H2S, a gasotransmitter, generated from l-cysteine by some enzymes including cystathionine-γ-lyase (CSE), are pro-nociceptive, since they enhance Cav3.2 T-type Ca2+ channel activity expressed in the primary afferents, most probably by canceling the channel inhibition by Zn2+ linked via coordinate bonding to His191 of Cav3.2. Given that germanium is reactive to sulfur, we tested whether THGP would directly trap sulfide, and inhibit sulfide-induced enhancement of Cav3.2 activity and sulfide-dependent pain in mice. Using mass spectrometry and 1H NMR techniques, we demonstrated that THGP directly reacted with sulfides including Na2S and NaSH, and formed a sulfur-containing reaction product, which decreased in the presence of ZnCl2. In Cav3.2-transfected HEK293 cells, THGP inhibited the sulfide-induced enhancement of T-type Ca2+ channel-dependent membrane currents. In mice, THGP, administered systemically or locally, inhibited the mechanical allodynia caused by intraplantar Na2S. In the mice with cyclophosphamide-induced cystitis and cerulein-induced pancreatitis, which exhibited upregulation of CSE in the bladder and pancreas, respectively, systemic administration of THGP as well as a selective T-type Ca2+ channel inhibitor suppressed the cystitis-related and pancreatitis-related visceral pain. These data suggest that THGP traps sulfide and inhibits sulfide-induced enhancement of Cav3.2 activity, leading to suppression of Cav3.2-dependent pain caused by sulfide applied exogenously and generated endogenously.
Collapse
Affiliation(s)
- Fumiko Sekiguchi
- Laboratory of Pharmacology and Pathophysiology, Faculty of Pharmacy, Kindai University, Kowakae 3-4-1, Higashi-Osaka, 577-8502, Japan
| | - Nene Koike
- Laboratory of Pharmacology and Pathophysiology, Faculty of Pharmacy, Kindai University, Kowakae 3-4-1, Higashi-Osaka, 577-8502, Japan
| | - Yasuhiro Shimada
- Asai Germanium Research Institute Co., Ltd., Suzuranoka, Hakodate, Hokkaido, 042-0958, Japan
| | - Kaho Sugimoto
- Laboratory of Pharmacology and Pathophysiology, Faculty of Pharmacy, Kindai University, Kowakae 3-4-1, Higashi-Osaka, 577-8502, Japan
| | - Hiroshi Masuda
- Laboratory of Pharmacology and Pathophysiology, Faculty of Pharmacy, Kindai University, Kowakae 3-4-1, Higashi-Osaka, 577-8502, Japan
| | - Takashi Nakamura
- Asai Germanium Research Institute Co., Ltd., Suzuranoka, Hakodate, Hokkaido, 042-0958, Japan
| | - Hiroaki Yamaguchi
- Yamagata University Graduate School of Medicine, Iida-nishi 2-2-2, Yamagata, 990-9585, Japan; Department of Pharmacy, Yamagata University Hospital, Iida-nishi 2-2-2, Yamagata, 990-9585, Japan
| | - Genzoh Tanabe
- Laboratory of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Kindai University, 3-4-1 Kowakae, Higashi-Osaka, 577-8502, Japan
| | - Shinsuke Marumoto
- Joint Research Center, Kindai University, 3-4-1 Kowakae, Higashi-Osaka, 577-8502, Japan
| | - Yoshihito Kasanami
- Laboratory of Pharmacology and Pathophysiology, Faculty of Pharmacy, Kindai University, Kowakae 3-4-1, Higashi-Osaka, 577-8502, Japan
| | - Maho Tsubota
- Laboratory of Pharmacology and Pathophysiology, Faculty of Pharmacy, Kindai University, Kowakae 3-4-1, Higashi-Osaka, 577-8502, Japan
| | - Tsuyako Ohkubo
- Division of Basic Medical Sciences and Fundamental Nursing, Faculty of Nursing, Fukuoka Nursing College, Fukuoka, 814-0193, Japan
| | - Shigeru Yoshida
- Department of Life Science, Faculty of Science and Engineering, Kindai University, 3-4-1 Kowakae, Higashi-Osaka, 577-8502, Japan
| | - Atsufumi Kawabata
- Laboratory of Pharmacology and Pathophysiology, Faculty of Pharmacy, Kindai University, Kowakae 3-4-1, Higashi-Osaka, 577-8502, Japan.
| |
Collapse
|
2
|
Trypsin Depolarizes Pacemaker Potentials in Murine Small Intestinal Interstitial Cells of Cajal. APPLIED SCIENCES-BASEL 2022. [DOI: 10.3390/app12094755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Interstitial cells of Cajal (ICCs) generate pacemaker potentials in the gastrointestinal (GI) tract. In this study, the effects of trypsin on pacemaker potentials in murine small intestinal ICCs were examined. We used whole-cell patch-clamp analysis. The results of whole-cell patch-clamp analysis revealed that trypsin dose-dependently depolarized pacemaker potentials and decreased their amplitude. Treatments with the antagonists of neurokinin1 (NK1) and NK2 receptors (SR-140333 and SR-48968, respectively) slightly inhibited the trypsin-induced responses. However, treatment with the combination of SR-140333 and SR-48968 completely inhibited trypsin-induced responses. Trypsin slightly depolarized pacemaker potentials and increased their amplitude after the intracellular application of GDP-β-S. Additionally, incubation in external Ca2+-free solution inhibited trypsin-induced responses. In the presence of U-73122, staurosporine, Go6976, or xestospongin C, trypsin did not depolarize the pacemaker’s potentials. However, trypsin depolarized the pacemaker potentials in the presence of rottlerin. Finally, HC067047, a TRPV4 inhibitor, did not affect the trypsin-induced responses. These results suggest that trypsin depolarized pacemaker potentials through NK1 and NK2 receptors in the murine small intestinal ICCs, with this effect being dependent on the G protein, phospholipase C, protein kinase C, inositol triphosphate pathways, and extracellular Ca2+ but being independent of the TRPV4 pathway. Hence, trypsin-mediated GI motility regulation must be considered for prokinetic drug developments.
Collapse
|
3
|
Oliveira KA, Torquato RJS, Lustosa DCGG, Ribeiro T, Nascimento BWL, de Oliveira LCG, Juliano MA, Paschoalin T, Lemos VS, Araujo RN, Pereira MH, Tanaka AS. Proteolytic activity of Triatoma infestans saliva associated with PAR-2 activation and vasodilation. J Venom Anim Toxins Incl Trop Dis 2021; 27:e20200098. [PMID: 33747067 PMCID: PMC7939238 DOI: 10.1590/1678-9199-jvatitd-2020-0098] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Background Triatoma infestans (Hemiptera: Reduviidae) is a
hematophagous insect and the main vector of Trypanosoma
cruzi (Kinetoplastida: Trypanosomatidae). In the present study,
the authors investigated whether a serine protease activity from the saliva
of T. infestans has a role in vasomotor modulation, and in
the insect-blood feeding by cleaving and activating protease-activated
receptors (PARs). Methods T. infestans saliva was chromatographed as previously
reported for purification of triapsin, a serine protease. The cleavage
activity of triapsin on PAR peptides was investigated based on FRET
technology. Mass spectrometry was used to analyze the sites of PAR-2 peptide
cleaved by triapsin. NO measurements were performed using the DAN assay
(2,3-diaminonapthalene). The vasorelaxant activity of triapsin was measured
in vessels with or without functional endothelium pre-contracted with
phenylephrine (3 µM). Intravital microscopy was used to assess the effect of
triapsin on mouse skin microcirculation. Results Triapsin was able to induce hydrolysis of PAR peptides and showed a higher
preference for cleavage of the PAR-2 peptide. Analysis by mass spectrometry
confirmed a single cleavage site, which corresponds to the activation site
of the PAR-2 receptor. Triapsin induced dose-dependent NO release in
cultured human umbilical vein endothelial cells (HUVECs), reaching a maximum
effect at 17.58 nM. Triapsin purified by gel-filtration chromatography
(10-16 to 10-9 M) was applied cumulatively to
mouse mesenteric artery rings and showed a potent endothelium-dependent
vasodilator effect (EC30 = 10-12 M). Nitric oxide
seems to be partially responsible for this vasodilator effect because L-NAME
(L-NG-nitroarginine methyl ester 300 µM), a nitric oxide synthetase
inhibitor, did not abrogate the vasodilation activated by triapsin.
Anti-PAR-2 antibody completely inhibited vasodilation observed in the
presence of triapsin activity. Triapsin activity also induced an increase in
the mouse ear venular diameter. Conclusion Data from this study suggest a plausible association between triapsin
activity mediated PAR-2 activation and vasodilation caused by T.
infestans saliva.
Collapse
Affiliation(s)
- Karla A Oliveira
- Department of Biochemistry and Pharmacology, Federal University of Piauí, Teresina, PI, Brazil
| | - Ricardo J S Torquato
- Department of Biochemistry, Federal University of São Paulo (Unifesp), São Paulo, SP, Brazil
| | - Daniela C G Garcia Lustosa
- Department of Pharmacology, Institute of Biomedical Sciences, Federal University of Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Tales Ribeiro
- Department of Parasitology, Institute of Biomedical Sciences, Federal University of Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Bruno W L Nascimento
- Department of Parasitology, Institute of Biomedical Sciences, Federal University of Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Lilian C G de Oliveira
- Department of Biophysics, National Institute of Pharmacology and Molecular Biology (INFAR), Federal University of São Paulo (Unifesp), São Paulo, SP, Brazil
| | - Maria A Juliano
- Department of Biophysics, National Institute of Pharmacology and Molecular Biology (INFAR), Federal University of São Paulo (Unifesp), São Paulo, SP, Brazil
| | - Thaysa Paschoalin
- Department of Biophysics, National Institute of Pharmacology and Molecular Biology (INFAR), Federal University of São Paulo (Unifesp), São Paulo, SP, Brazil
| | - Virginia S Lemos
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, Federal University of Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Ricardo N Araujo
- Department of Parasitology, Institute of Biomedical Sciences, Federal University of Minas Gerais (UFMG), Belo Horizonte, MG, Brazil.,National Institute of Science and Technology in Molecular Entomology (INCT-EM), Rio de Janeiro, RJ, Brazil
| | - Marcos H Pereira
- Department of Parasitology, Institute of Biomedical Sciences, Federal University of Minas Gerais (UFMG), Belo Horizonte, MG, Brazil.,National Institute of Science and Technology in Molecular Entomology (INCT-EM), Rio de Janeiro, RJ, Brazil
| | - Aparecida S Tanaka
- Department of Biochemistry, Federal University of São Paulo (Unifesp), São Paulo, SP, Brazil.,National Institute of Science and Technology in Molecular Entomology (INCT-EM), Rio de Janeiro, RJ, Brazil
| |
Collapse
|
4
|
Hassler SN, Kume M, Mwirigi JM, Ahmad A, Shiers S, Wangzhou A, Ray PR, Belugin SN, Naik DK, Burton MD, Vagner J, Boitano S, Akopian AN, Dussor G, Price TJ. The cellular basis of protease-activated receptor 2-evoked mechanical and affective pain. JCI Insight 2020; 5:137393. [PMID: 32352932 PMCID: PMC7308051 DOI: 10.1172/jci.insight.137393] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 04/24/2020] [Indexed: 12/31/2022] Open
Abstract
Protease-activated receptor 2 (PAR2) has long been implicated in inflammatory and visceral pain, but the cellular basis of PAR2-evoked pain has not been delineated. Although PAR2-evoked pain has been attributed to sensory neuron expression, RNA-sequencing experiments show ambiguous F2rl1 mRNA detection. Moreover, many pharmacological tools for PAR2 are nonspecific, acting also on the Mas-related GPCR family (Mrg) that are highly enriched in sensory neurons. We sought to clarify the cellular basis of PAR2-evoked pain. We developed a PAR2-conditional knockout mouse and specifically deleted PAR2 in all sensory neurons using the PirtCre mouse line. Our behavioral findings show that PAR2 agonist-evoked mechanical hyperalgesia and facial grimacing, but not thermal hyperalgesia, are dependent on PAR2 expression in sensory neurons that project to the hind paw in male and female mice. F2rl1 mRNA is expressed in a discrete population (~4%) of mostly small-diameter sensory neurons that coexpress the Nppb and IL31ra genes. This cell population has been implicated in itch, but our work shows that PAR2 activation in these cells causes clear pain-related behaviors from the skin. Our findings show that a discrete population of DRG sensory neurons mediate PAR2-evoked pain.
Collapse
Affiliation(s)
- Shayne N. Hassler
- School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, University of Texas at Dallas, Dallas, Texas, USA
| | - Moeno Kume
- School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, University of Texas at Dallas, Dallas, Texas, USA
| | - Juliet M. Mwirigi
- School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, University of Texas at Dallas, Dallas, Texas, USA
| | - Ayesha Ahmad
- School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, University of Texas at Dallas, Dallas, Texas, USA
| | - Stephanie Shiers
- School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, University of Texas at Dallas, Dallas, Texas, USA
| | - Andi Wangzhou
- School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, University of Texas at Dallas, Dallas, Texas, USA
| | - Pradipta R. Ray
- School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, University of Texas at Dallas, Dallas, Texas, USA
| | - Serge N. Belugin
- Department of Endodontics, School of Dentistry, University of Texas Health San Antonio, San Antonio, Texas, USA
| | - Dhananjay K. Naik
- School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, University of Texas at Dallas, Dallas, Texas, USA
| | - Michael D. Burton
- School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, University of Texas at Dallas, Dallas, Texas, USA
| | | | - Scott Boitano
- BIO5 Research Institute and
- Department of Physiology, Asthma & Airway Disease Research Center, University of Arizona, Tucson, Arizona, USA
| | - Armen N. Akopian
- Department of Endodontics, School of Dentistry, University of Texas Health San Antonio, San Antonio, Texas, USA
| | - Gregory Dussor
- School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, University of Texas at Dallas, Dallas, Texas, USA
| | - Theodore J. Price
- School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, University of Texas at Dallas, Dallas, Texas, USA
| |
Collapse
|
5
|
Nitric Oxide Stimulates Acute Pancreatitis Pain via Activating the NF- κB Signaling Pathway and Inhibiting the Kappa Opioid Receptor. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:9230958. [PMID: 32454946 PMCID: PMC7231422 DOI: 10.1155/2020/9230958] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 03/03/2020] [Accepted: 03/11/2020] [Indexed: 12/03/2022]
Abstract
Pain is the most important clinical feature of acute pancreatitis (AP); however, its specific mechanism is currently unclear. In this study, we showed that AP caused an increase in nitric oxide (NO) secretion, activated the NF-κB pathway in the dorsal root ganglia (DRGs), and caused pain. We established an AP model in vivo and tested the expression of NO, the kappa opioid receptor (KOR), and pain factors. We showed that NO in AP was significantly elevated and increased the expression of pain factors. Next, by treating DRGs in vitro, it was found that NO activated the NF-κB pathway; conversely, NF-κB had no effect on NO. Moreover, inhibition of NF-κB promoted the KOR, whereas NF-κB did not change after KOR activation. Finally, behavioral experiments showed that a NO donor increased the pain behavior of mice, while a NO scavenger, NF-κB inhibitor, or KOR agonist attenuated the pain response in mice. These results suggest that iNOS/NO/NF-κB/KOR may be a key mechanism of pain in AP, providing a theoretical basis for the use of peripheral-restricted KOR agonists for pain treatment in AP.
Collapse
|
6
|
Saloman JL, Albers KM, Cruz-Monserrate Z, Davis BM, Edderkaoui M, Eibl G, Epouhe AY, Gedeon JY, Gorelick FS, Grippo PJ, Groblewski GE, Husain SZ, Lai KK, Pandol SJ, Uc A, Wen L, Whitcomb DC. Animal Models: Challenges and Opportunities to Determine Optimal Experimental Models of Pancreatitis and Pancreatic Cancer. Pancreas 2019; 48:759-779. [PMID: 31206467 PMCID: PMC6581211 DOI: 10.1097/mpa.0000000000001335] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
At the 2018 PancreasFest meeting, experts participating in basic research met to discuss the plethora of available animal models for studying exocrine pancreatic disease. In particular, the discussion focused on the challenges currently facing the field and potential solutions. That meeting culminated in this review, which describes the advantages and limitations of both common and infrequently used models of exocrine pancreatic disease, namely, pancreatitis and exocrine pancreatic cancer. The objective is to provide a comprehensive description of the available models but also to provide investigators with guidance in the application of these models to investigate both environmental and genetic contributions to exocrine pancreatic disease. The content covers both nongenic and genetically engineered models across multiple species (large and small). Recommendations for choosing the appropriate model as well as how to conduct and present results are provided.
Collapse
Affiliation(s)
- Jami L. Saloman
- Department of Neurobiology, Pittsburgh Center for Pain Research, University of Pittsburgh, Pittsburgh, PA
| | - Kathryn M. Albers
- Department of Neurobiology, Pittsburgh Center for Pain Research, University of Pittsburgh, Pittsburgh, PA
| | - Zobeida Cruz-Monserrate
- Division of Gastroenterology, Hepatology, and Nutrition; Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH
| | - Brian M. Davis
- Department of Neurobiology, Pittsburgh Center for Pain Research, University of Pittsburgh, Pittsburgh, PA
| | - Mouad Edderkaoui
- Basic and Translational Pancreas Research, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Guido Eibl
- Department of Surgery, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, CA
| | - Ariel Y. Epouhe
- Department of Neurobiology, Pittsburgh Center for Pain Research, University of Pittsburgh, Pittsburgh, PA
| | - Jeremy Y. Gedeon
- Department of Neurobiology, Pittsburgh Center for Pain Research, University of Pittsburgh, Pittsburgh, PA
| | - Fred S. Gorelick
- Department of Internal Medicine, Section of Digestive Diseases & Department of Cell Biology Yale University School of Medicine; Veterans Affairs Connecticut Healthcare, West Haven, CT
| | - Paul J. Grippo
- Department of Medicine, Division of Gastroenterology and Hepatology, UI Cancer Center, University of Illinois at Chicago, Chicago, IL
| | - Guy E. Groblewski
- Department of Nutritional Sciences, University of Wisconsin, Madison, WI
| | | | - Keane K.Y. Lai
- Department of Pathology (National Medical Center), Department of Molecular Medicine (Beckman Research Institute), and Comprehensive Cancer Center, City of Hope, Duarte, CA
| | - Stephen J. Pandol
- Department of Surgery, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, CA
| | - Aliye Uc
- Stead Family Department of Pediatrics, University of Iowa, Stead Family Children’s Hospital, Iowa City, IA
| | - Li Wen
- Department of Pediatrics, Stanford University, Palo Alto, CA
| | | |
Collapse
|
7
|
Macrophage-derived HMGB1 as a Pain Mediator in the Early Stage of Acute Pancreatitis in Mice: Targeting RAGE and CXCL12/CXCR4 Axis. J Neuroimmune Pharmacol 2017; 12:693-707. [DOI: 10.1007/s11481-017-9757-2] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Accepted: 07/20/2017] [Indexed: 12/11/2022]
|
8
|
Abreu FF, Souza ACA, Teixeira SA, Soares AG, Teixeira DF, Soares RC, Santana MT, Lauton Santos S, Costa SKP, Muscará MN, Camargo EA. Elucidating the role of oxidative stress in the therapeutic effect of rutin on experimental acute pancreatitis. Free Radic Res 2016; 50:1350-1360. [DOI: 10.1080/10715762.2016.1247494] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Affiliation(s)
| | | | - Simone Aparecida Teixeira
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Antônio Garcia Soares
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | | | | | | | - Sandra Lauton Santos
- Department of Physiology, Federal University of Sergipe, São Cristóvão, SE, Brazil
| | - Soraia Kátia Pereira Costa
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Marcelo Nicolas Muscará
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | | |
Collapse
|
9
|
Yau MK, Lim J, Liu L, Fairlie DP. Protease activated receptor 2 (PAR2) modulators: a patent review (2010-2015). Expert Opin Ther Pat 2016; 26:471-83. [PMID: 26936077 DOI: 10.1517/13543776.2016.1154540] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
INTRODUCTION Protease activated receptor 2 (PAR2) is a self-activated G protein-coupled receptor that has been implicated in several diseases, including inflammatory, gastrointestinal, respiratory, metabolic diseases, cancers and others, making it an important prospective drug target. No known endogenous ligands are available for PAR2, so having potent exogenous agonists and antagonists can be helpful for studying physiological functions of PAR2. AREAS COVERED This review covers agonist-, antagonist-, antibody- and pepducin-based modulators of PAR2 reported in patent applications between 2010-2015, along with their available structure-activity relationships, biological activities and potential uses for studying PAR2. EXPERT OPINION In the last six years, substantial efforts were made towards developing PAR2 modulators, but most lack potency or selectivity or have poor pharmacokinetic profiles. Many PAR2 modulators were assessed by measuring Gαq protein-mediated calcium release in cells. This may be insufficient to fully characterize ligand function, since different ligands signal through PAR2 via multiple signaling pathways. It may be feasible to develop biased ligands as drugs that can selectively modulate one or more specific signaling pathways linking PAR2 to a specific diseased state. Accordingly, potent, orally bioavailable, pathway- and receptor-selective PAR2 modulators may be an achievable goal to realizing effective drugs that can treat PAR2-mediated diseases.
Collapse
Affiliation(s)
- Mei-Kwan Yau
- a Division of Chemistry and Structural Biology, Institute for Molecular Bioscience , The University of Queensland , Brisbane , Australia
| | - Junxian Lim
- a Division of Chemistry and Structural Biology, Institute for Molecular Bioscience , The University of Queensland , Brisbane , Australia
| | - Ligong Liu
- a Division of Chemistry and Structural Biology, Institute for Molecular Bioscience , The University of Queensland , Brisbane , Australia
| | - David P Fairlie
- a Division of Chemistry and Structural Biology, Institute for Molecular Bioscience , The University of Queensland , Brisbane , Australia
| |
Collapse
|
10
|
Protease-activated receptor 2 activation is sufficient to induce the transition to a chronic pain state. Pain 2016; 156:859-867. [PMID: 25734998 DOI: 10.1097/j.pain.0000000000000125] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Protease-activated receptor type 2 (PAR2) is known to play an important role in inflammatory, visceral, and cancer-evoked pain based on studies using PAR2 knockout (PAR2(-/-)) mice. We have tested the hypothesis that specific activation of PAR2 is sufficient to induce a chronic pain state through extracellular signal-regulated kinase (ERK) signaling to protein synthesis machinery. We have further tested whether the maintenance of this chronic pain state involves a brain-derived neurotrophic factor (BDNF)/tropomyosin-related kinase B (trkB)/atypical protein kinase C (aPKC) signaling axis. We observed that intraplantar injection of the novel highly specific PAR2 agonist, 2-aminothiazol-4-yl-LIGRL-NH2 (2-at), evokes a long-lasting acute mechanical hypersensitivity (median effective dose ∼12 pmoles), facial grimacing, and causes robust hyperalgesic priming as revealed by a subsequent mechanical hypersensitivity and facial grimacing to prostaglandin E2 (PGE2) injection. The promechanical hypersensitivity effect of 2-at is completely absent in PAR2(-/-) mice as is hyperalgesic priming. Intraplantar injection of the upstream ERK inhibitor, U0126, and the eukaryotic initiation factor (eIF) 4F complex inhibitor, 4EGI-1, prevented the development of acute mechanical hypersensitivity and hyperalgesic priming after 2-at injection. Systemic injection of the trkB antagonist ANA-12 similarly inhibited PAR2-mediated mechanical hypersensitivity, grimacing, and hyperalgesic priming. Inhibition of aPKC (intrathecal delivery of ZIP) or trkB (systemic administration of ANA-12) after the resolution of 2-at-induced mechanical hypersensitivity reversed the maintenance of hyperalgesic priming. Hence, PAR2 activation is sufficient to induce neuronal plasticity leading to a chronic pain state, the maintenance of which is dependent on a BDNF/trkB/aPKC signaling axis.
Collapse
|
11
|
Anderson MA, Akshintala V, Albers KM, Amann ST, Belfer I, Brand R, Chari S, Cote G, Davis BM, Frulloni L, Gelrud A, Guda N, Humar A, Liddle RA, Slivka A, Gupta RS, Szigethy E, Talluri J, Wassef W, Wilcox CM, Windsor J, Yadav D, Whitcomb DC. Mechanism, assessment and management of pain in chronic pancreatitis: Recommendations of a multidisciplinary study group. Pancreatology 2016; 16:83-94. [PMID: 26620965 PMCID: PMC4761301 DOI: 10.1016/j.pan.2015.10.015] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Revised: 10/29/2015] [Accepted: 10/31/2015] [Indexed: 12/11/2022]
Abstract
DESCRIPTION Pain in patients with chronic pancreatitis (CP) remains the primary clinical complaint and source of poor quality of life. However, clear guidance on evaluation and treatment is lacking. METHODS Pancreatic Pain working groups reviewed information on pain mechanisms, clinical pain assessment and pain treatment in CP. Levels of evidence were assigned using the Oxford system, and consensus was based on GRADE. A consensus meeting was held during PancreasFest 2012 with substantial post-meeting discussion, debate, and manuscript refinement. RESULTS Twelve discussion questions and proposed guidance statements were presented. Conference participates concluded: Disease Mechanism: Pain etiology is multifactorial, but data are lacking to effectively link symptoms with pathologic feature and molecular subtypes. Assessment of Pain: Pain should be assessed at each clinical visit, but evidence to support an optimal approach to assessing pain character, frequency and severity is lacking. MANAGEMENT There was general agreement on the roles for endoscopic and surgical therapies, but less agreement on optimal patient selection for medical, psychological, endoscopic, surgical and other therapies. CONCLUSIONS Progress is occurring in pain biology and treatment options, but pain in patients with CP remains a major problem that is inadequately understood, measured and managed. The growing body of information needs to be translated into more effective clinical care.
Collapse
Affiliation(s)
| | | | - Kathryn M Albers
- Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA, USA
| | | | - Inna Belfer
- Department of Anesthesiology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Randall Brand
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Suresh Chari
- Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Greg Cote
- Department of Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Brian M Davis
- Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Luca Frulloni
- Department of Medicine, University of Verona, Verona, Italy
| | - Andres Gelrud
- Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Nalini Guda
- Department of Gastroenterology, Aurora St. Luke's Medical Center, Milwaukee, WI, USA
| | - Abhinav Humar
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | | | - Adam Slivka
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | | | - Eva Szigethy
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jyothsna Talluri
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Wahid Wassef
- Department of Medicine, University of Massachusetts, Worcester, MA, USA
| | - C Mel Wilcox
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - John Windsor
- Department of Surgery, University of Auckland, Auckland, New Zealand
| | - Dhiraj Yadav
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - David C Whitcomb
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA; Department of Cell Biology & Molecular Physiology, University of Pittsburgh, Pittsburgh, PA, USA; Department of Human Genetics, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
12
|
Atsawarungruangkit A, Pongprasobchai S. Current understanding of the neuropathophysiology of pain in chronic pancreatitis. World J Gastrointest Pathophysiol 2015; 6:193-202. [PMID: 26600977 PMCID: PMC4644883 DOI: 10.4291/wjgp.v6.i4.193] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Revised: 07/22/2015] [Accepted: 09/16/2015] [Indexed: 02/06/2023] Open
Abstract
Chronic pancreatitis (CP) is a chronic inflammatory disease of the pancreas. The main symptom of patients with CP is chronic and severe abdominal pain. However, the pathophysiology of pain in CP remains obscure. Traditionally, researchers believed that the pain was caused by anatomical changes in pancreatic structure. However, treatment outcomes based on such beliefs are considered unsatisfactory. The emerging explanations of pain in CP are trending toward neurobiological theories. This article aims to review current evidence regarding the neuropathophysiology of pain in CP and its potential implications for the development of new treatments for pain in CP.
Collapse
|
13
|
Abstract
The human exocrine pancreas consists of 2 main cell types: acinar and ductal cells. These exocrine cells interact closely to contribute to the secretion of pancreatic juice. The most important ion in terms of the pancreatic ductal secretion is HCO3. In fact, duct cells produce an alkaline fluid that may contain up to 140 mM NaHCO3, which is essential for normal digestion. This article provides an overview of the basics of pancreatic ductal physiology and pathophysiology. In the first part of the article, we discuss the ductal electrolyte and fluid transporters and their regulation. The central role of cystic fibrosis transmembrane conductance regulator (CFTR) is highlighted, which is much more than just a Cl channel. We also review the role of pancreatic ducts in severe debilitating diseases such as cystic fibrosis (caused by various genetic defects of cftr), pancreatitis, and diabetes mellitus. Stimulation of ductal secretion in cystic fibrosis and pancreatitis may have beneficial effects in their treatment.
Collapse
|
14
|
Abstract
PURPOSE OF REVIEW Pain is the most common symptom of chronic pancreatitis, with a profound socioeconomic impact. Historical management paradigms failed, as they did not adequately address the fundamental underlying mechanisms. The present article describes the neurobiology of pain and sensitization in this condition, in an effort to explain prior failings and provide future directions for managing pain in chronic pancreatitis. RECENT FINDINGS A number of recent advances have been made in understanding the neurobiology of pain for this condition. This has been coupled with clinical advances in assessing sensitization to pain in these patients, which has been shown to predict response to medical and surgical therapy. SUMMARY Pain in chronic pancreatitis is complex. Addressing the mechanical and morphological findings in chronic pancreatitis without addressing the underlying neurobiological mechanisms is destined to fail. New advances in our understanding of the neurobiology of pain in chronic pancreatitis helps to explain prior failings and provides future direction for managing pain in patients afflicted by this disease.
Collapse
|
15
|
Abstract
Proteinase-activated receptors (PARs) are a family of G protein-coupled receptor that are activated by extracellular cleavage of the receptor in the N-terminal domain. This slicing of the receptor exposes a tethered ligand which binds to a specific docking point on the receptor surface to initiate intracellular signalling. PARs are expressed by numerous tissues in the body, and they are involved in various physiological and pathological processes such as food digestion, tissue remodelling and blood coagulation. This chapter will summarise how serine proteinases activate PARs leading to the development of pain in several chronic pain conditions. The potential of PARs as a drug target for pain relief is also discussed.
Collapse
Affiliation(s)
- Jason J McDougall
- Departments of Pharmacology and Anaesthesia, Pain Management and Perioperative Medicine, Dalhousie University, 5850 College Street, Halifax, NS, Canada, B3H 4R2,
| | | |
Collapse
|
16
|
Veldhuis NA, Poole DP, Grace M, McIntyre P, Bunnett NW. The G Protein–Coupled Receptor–Transient Receptor Potential Channel Axis: Molecular Insights for Targeting Disorders of Sensation and Inflammation. Pharmacol Rev 2014; 67:36-73. [DOI: 10.1124/pr.114.009555] [Citation(s) in RCA: 114] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
|
17
|
Suen JY, Cotterell A, Lohman RJ, Lim J, Han A, Yau MK, Liu L, Cooper MA, Vesey DA, Fairlie DP. Pathway-selective antagonism of proteinase activated receptor 2. Br J Pharmacol 2014; 171:4112-24. [PMID: 24821440 DOI: 10.1111/bph.12757] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2014] [Revised: 04/04/2014] [Accepted: 04/30/2014] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND AND PURPOSE Proteinase activated receptor 2 (PAR2) is a GPCR associated with inflammation, metabolism and disease. Clues to understanding how to block PAR2 signalling associated with disease without inhibiting PAR2 activation in normal physiology could be provided by studies of biased signalling. EXPERIMENTAL APPROACH PAR2 ligand GB88 was profiled for PAR2 agonist and antagonist properties by several functional assays associated with intracellular G-protein-coupled signalling in vitro in three cell types and with PAR2-induced rat paw oedema in vivo. KEY RESULTS In HT29 cells, GB88 was a PAR2 antagonist in terms of Ca(2+) mobilization and PKC phosphorylation, but a PAR2 agonist in attenuating forskolin-induced cAMP accumulation, increasing ERK1/2 phosphorylation, RhoA activation, myosin phosphatase phosphorylation and actin filament rearrangement. In CHO-hPAR2 cells, GB88 inhibited Ca(2+) release, but activated G(i/o) and increased ERK1/2 phosphorylation. In human kidney tubule cells, GB88 inhibited cytokine secretion (IL6, IL8, GM-CSF, TNF-α) mediated by PAR2. A rat paw oedema induced by PAR2 agonists was also inhibited by orally administered GB88 and compared with effects of locally administered inhibitors of G-protein coupled pathways. CONCLUSIONS AND IMPLICATIONS GB88 is a biased antagonist of PAR2 that selectively inhibits PAR2/G(q/11)/Ca(2+)/PKC signalling, leading to anti-inflammatory activity in vivo, while being an agonist in activating three other PAR2-activated pathways (cAMP, ERK, Rho) in human cells. These findings highlight opportunities to design drugs to block specific PAR2-linked signalling pathways in disease, without blocking beneficial PAR2 signalling in normal physiology, and to dissect PAR2-associated mechanisms of disease in vivo.
Collapse
Affiliation(s)
- J Y Suen
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Qld, Australia
| | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Development and evaluation of small peptidomimetic ligands to protease-activated receptor-2 (PAR2) through the use of lipid tethering. PLoS One 2014; 9:e99140. [PMID: 24927179 PMCID: PMC4057235 DOI: 10.1371/journal.pone.0099140] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Accepted: 05/09/2014] [Indexed: 02/05/2023] Open
Abstract
Protease-activated receptor-2 (PAR2) is a G-Protein Coupled Receptor (GPCR) activated by proteolytic cleavage to expose an attached, tethered ligand (SLIGRL). We evaluated the ability for lipid-tethered-peptidomimetics to activate PAR2 with in vitro physiological and Ca2+ signaling assays to determine minimal components necessary for potent, specific and full PAR2 activation. A known PAR2 activating compound containing a hexadecyl (Hdc) lipid via three polyethylene glycol (PEG) linkers (2at-LIGRL-PEG3-Hdc) provided a potent agonist starting point (physiological EC50 = 1.4 nM; 95% CI: 1.2-2.3 nM). In a set of truncated analogs, 2at-LIGR-PEG3-Hdc retained potency (EC50 = 2.1 nM; 1.3-3.4 nM) with improved selectivity for PAR2 over Mas1 related G-protein coupled receptor type C11, a GPCR that can be activated by the PAR2 peptide agonist, SLIGRL-NH2. 2at-LIG-PEG3-Hdc was the smallest full PAR2 agonist, albeit with a reduced EC50 (46 nM; 20-100 nM). 2at-LI-PEG3-Hdc retained specific activity for PAR2 with reduced EC50 (310 nM; 260-360 nM) but displayed partial PAR2 activation in both physiological and Ca2+ signaling assays. Further truncation (2at-L-PEG3-Hdc and 2at-PEG3-Hdc) eliminated in vitro activity. When used in vivo, full and partial PAR2 in vitro agonists evoked mechanical hypersensitivity at a 15 pmole dose while 2at-L-PEG3-Hdc lacked efficacy. Minimum peptidomimetic PAR2 agonists were developed with known heterocycle substitutes for Ser1 (isoxazole or aminothiazoyl) and cyclohexylalanine (Cha) as a substitute for Leu2. Both heterocycle-tetrapeptide and heterocycle-dipeptides displayed PAR2 specificity, however, only the heterocycle-tetrapeptides displayed full PAR2 agonism. Using the lipid-tethered-peptidomimetic approach we have developed novel structure activity relationships for PAR2 that allows for selective probing of PAR2 function across a broad range of physiological systems.
Collapse
|
19
|
A systematic review of the evidence for central nervous system plasticity in animal models of inflammatory-mediated gastrointestinal pain. Inflamm Bowel Dis 2014; 20:176-95. [PMID: 24284415 DOI: 10.1097/01.mib.0000437499.52922.b1] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND Abdominal pain frequently accompanies inflammatory disorders of the gastrointestinal tract (GIT), and animal models of GIT inflammation have been developed to explore the role of the central nervous system (CNS) in this process. Here, we summarize the evidence from animal studies for CNS plasticity following GIT inflammation. METHODS A systematic review was conducted to identify studies that: (1) used inflammation of GIT organs, (2) assessed pain or visceral hypersensitivity, and (3) presented evidence of CNS involvement. Two hundred and eight articles were identified, and 79 were eligible for analysis. RESULTS Rats were most widely used (76%). Most studies used adult animals (42%) with a bias toward males (74%). Colitis was the most frequently used model (78%) and 2,4,6-trinitrobenzenesulfonic acid the preferred inflammatory agent (33%). Behavioral (58%), anatomical/molecular (44%), and physiological (24%) approaches were used alone or in combination to assess CNS involvement during or after GIT inflammation. Measurement times varied widely (<1 h-> 2 wk after inflammation). Blinded outcomes were used in 42% studies, randomization in 10%, and evidence of visceral inflammation in 54%. Only 3 studies fulfilled our criteria for high methodological quality, and no study reported sample size calculations. CONCLUSIONS The included studies provide strong evidence for CNS plasticity following GIT inflammation, specifically in the spinal cord dorsal horn. This evidence includes altered visceromotor responses and indices of referred pain, elevated neural activation and peptide content, and increased neuronal excitability. This evidence supports continued use of this approach for preclinical studies; however, there is substantial scope to improve study design.
Collapse
|
20
|
Yau MK, Liu L, Fairlie DP. Toward drugs for protease-activated receptor 2 (PAR2). J Med Chem 2013; 56:7477-97. [PMID: 23895492 DOI: 10.1021/jm400638v] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
PAR2 has a distinctive functional phenotype among an unusual group of GPCRs called protease activated receptors, which self-activate after cleavage of their N-termini by mainly serine proteases. PAR2 is the most highly expressed PAR on certain immune cells, and it is activated by multiple proteases (but not thrombin) in inflammation. PAR2 is expressed on many types of primary human cells and cancer cells. PAR2 knockout mice and PAR2 agonists and antagonists have implicated PAR2 as a promising target in inflammatory conditions; respiratory, gastrointestinal, metabolic, cardiovascular, and neurological dysfunction; and cancers. This article summarizes salient features of PAR2 structure, activation, and function; opportunities for disease intervention via PAR2; pharmacological properties of published or patented PAR2 modulators (small molecule agonists and antagonists, pepducins, antibodies); and some personal perspectives on limitations of assessing their properties and on promising new directions for PAR2 modulation.
Collapse
Affiliation(s)
- Mei-Kwan Yau
- Division of Chemistry and Structural Biology, Institute for Molecular Bioscience, The University of Queensland , Brisbane, Queensland 4072, Australia
| | | | | |
Collapse
|
21
|
Flynn AN, Hoffman J, Tillu DV, Sherwood CL, Zhang Z, Patek R, Asiedu MNK, Vagner J, Price TJ, Boitano S. Development of highly potent protease-activated receptor 2 agonists via synthetic lipid tethering. FASEB J 2013; 27:1498-510. [PMID: 23292071 DOI: 10.1096/fj.12-217323] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Protease-activated receptor-2 (PAR₂) is a G-protein coupled receptor (GPCR) associated with a variety of pathologies. However, the therapeutic potential of PAR₂ is limited by a lack of potent and specific ligands. Following proteolytic cleavage, PAR₂ is activated through a tethered ligand. Hence, we reasoned that lipidation of peptidomimetic ligands could promote membrane targeting and thus significantly improve potency and constructed a series of synthetic tethered ligands (STLs). STLs contained a peptidomimetic PAR₂ agonist (2-aminothiazol-4-yl-LIGRL-NH₂) bound to a palmitoyl group (Pam) via polyethylene glycol (PEG) linkers. In a high-throughput physiological assay, these STL agonists displayed EC₅₀ values as low as 1.47 nM, representing a ∼200 fold improvement over the untethered parent ligand. Similarly, these STL agonists were potent activators of signaling pathways associated with PAR₂: EC₅₀ for Ca(2+) response as low as 3.95 nM; EC₅₀ for MAPK response as low as 9.49 nM. Moreover, STLs demonstrated significant improvement in potency in vivo, evoking mechanical allodynia with an EC₅₀ of 14.4 pmol. STLs failed to elicit responses in PAR2(-/-) cells at agonist concentrations of >300-fold their EC₅₀ values. Our results demonstrate that the STL approach is a powerful tool for increasing ligand potency at PAR₂ and represent opportunities for drug development at other protease activated receptors and across GPCRs.
Collapse
Affiliation(s)
- Andrea N Flynn
- Department of Physiology, Arizona Health Sciences Center, Tucson, AZ 85724-5030, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Terada Y, Fujimura M, Nishimura S, Tsubota M, Sekiguchi F, Nishikawa H, Kawabata A. Contribution of TRPA1 as a Downstream Signal of Proteinase-Activated Receptor-2 to Pancreatic Pain. J Pharmacol Sci 2013; 123:284-7. [DOI: 10.1254/jphs.13128sc] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
|
23
|
Lee YT, Wei J, Chuang YC, Chang CY, Chen IC, Weng CF, Schmid-Schönbein GW. Successful treatment with continuous enteral protease inhibitor in a patient with severe septic shock. Transplant Proc 2012; 44:817-9. [PMID: 22483504 DOI: 10.1016/j.transproceed.2012.03.032] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
OBJECTIVE The mortality rate among patients with septic shock is high despite current therapy. We present a case of Fournier's gangrene and septic shock at 4 years post-heart transplantation that was reversed by "continuous enteral feeding" of the digestive enzyme inhibitor, gabexate mesilate. Recently, powerful pancreatic digestive proteases in the lumen of the intestine have been identified as initiators of the systemic inflammatory response. Intraluminal inhibitions of the proteases significantly attenuates intestinal damage, system inflammation, and multiorgan failure in experimental forms of shock but it has not been tested in man. METHODS AND RESULTS Gabexate mesilate, a synthetic digestive protease inhibitor, was continuously administered in two liters of crystalloid solution to a patient by enteral feeding during septic shock. The condition and markers for shock due to sepsis reversed in a few days. CONCLUSION This case suggested that "enteral" digestive protease inhibition may decrease and even reverse the sequelae of shock and sepsis.
Collapse
Affiliation(s)
- Y-T Lee
- Heart Center, Cheng Hsin General Hospital, Taipei, Taiwan
| | | | | | | | | | | | | |
Collapse
|
24
|
Camargol EA, Zanonil CI, Toyamal MH, Muscaral MN, Dochertyl RJ, Costal SK. Abdominal hyperalgesia in secretory phospholipase A2-induced rat pancreatitis: Distinct roles of NK1receptors. Eur J Pain 2012; 15:900-6. [DOI: 10.1016/j.ejpain.2011.04.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2010] [Revised: 02/19/2011] [Accepted: 04/05/2011] [Indexed: 11/28/2022]
|
25
|
Pallagi P, Venglovecz V, Rakonczay Z, Borka K, Korompay A, Ózsvári B, Judák L, Sahin-Tóth M, Geisz A, Schnúr A, Maléth J, Takács T, Gray MA, Argent BE, Mayerle J, Lerch MM, Wittmann T, Hegyi P. Trypsin reduces pancreatic ductal bicarbonate secretion by inhibiting CFTR Cl⁻ channels and luminal anion exchangers. Gastroenterology 2011; 141:2228-2239.e6. [PMID: 21893120 PMCID: PMC3273991 DOI: 10.1053/j.gastro.2011.08.039] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2010] [Revised: 07/14/2011] [Accepted: 08/05/2011] [Indexed: 12/16/2022]
Abstract
BACKGROUND & AIMS The effects of trypsin on pancreatic ductal epithelial cells (PDECs) vary among species and depend on the localization of proteinase-activated receptor 2 (PAR-2). We compared PAR-2 localization in human and guinea-pig PDECs, and used isolated guinea pig ducts to study the effects of trypsin and a PAR-2 agonist on bicarbonate secretion. METHODS PAR-2 localization was analyzed by immunohistochemistry in guinea pig and human pancreatic tissue samples (from 15 patients with chronic pancreatitis and 15 without pancreatic disease). Functionally, guinea pig PDECs were studied by microperfusion of isolated ducts, measurements of intracellular pH and intracellular Ca(2+) concentration, and patch clamp analysis. The effect of pH on trypsinogen autoactivation was assessed using recombinant human cationic trypsinogen. RESULTS PAR-2 localized to the apical membrane of human and guinea pig PDECs. Trypsin increased intracellular Ca(2+) concentration and intracellular pH and inhibited secretion of bicarbonate by the luminal anion exchanger and the cystic fibrosis transmembrane conductance regulator (CFTR) Cl(-) channel. Autoactivation of human cationic trypsinogen accelerated when the pH was reduced from 8.5 to 6.0. PAR-2 expression was strongly down-regulated, at transcriptional and protein levels, in the ducts of patients with chronic pancreatitis, consistent with increased activity of intraductal trypsin. Importantly, in PAR-2 knockout mice, the effects of trypsin were markedly reduced. CONCLUSIONS Trypsin reduces pancreatic ductal bicarbonate secretion via PAR-2-dependent inhibition of the apical anion exchanger and the CFTR Cl(-) channel. This could contribute to the development of chronic pancreatitis by decreasing luminal pH and promoting premature activation of trypsinogen in the pancreatic ducts.
Collapse
Affiliation(s)
- Petra Pallagi
- First Dept. of Medicine, University of Szeged, Szeged, Hungary
| | | | | | - Katalin Borka
- 2nd Dept. of Pathology, Semmelweis University, Budapest, Hungary
| | - Anna Korompay
- 2nd Dept. of Pathology, Semmelweis University, Budapest, Hungary
| | - Béla Ózsvári
- First Dept. of Medicine, University of Szeged, Szeged, Hungary
| | - Linda Judák
- First Dept. of Medicine, University of Szeged, Szeged, Hungary
| | - Miklós Sahin-Tóth
- Dept. of Molecular and Cell Biology, Henry M. Goldman School of Dental Medicine, Boston University, Boston, MA, USA
| | - Andrea Geisz
- First Dept. of Medicine, University of Szeged, Szeged, Hungary,Dept. of Molecular and Cell Biology, Henry M. Goldman School of Dental Medicine, Boston University, Boston, MA, USA
| | - Andrea Schnúr
- First Dept. of Medicine, University of Szeged, Szeged, Hungary,Dept. of Molecular and Cell Biology, Henry M. Goldman School of Dental Medicine, Boston University, Boston, MA, USA
| | - József Maléth
- First Dept. of Medicine, University of Szeged, Szeged, Hungary
| | - Tamás Takács
- First Dept. of Medicine, University of Szeged, Szeged, Hungary
| | - Mike A. Gray
- Institute for Cell & Molecular Biosciences, Newcastle University, Newcastle upon Tyne, UK
| | - Barry E. Argent
- Institute for Cell & Molecular Biosciences, Newcastle University, Newcastle upon Tyne, UK
| | - Julia Mayerle
- Department of Medicine A, Greifswald University Hospital, Greifswald, Germany
| | - Markus M. Lerch
- Department of Medicine A, Greifswald University Hospital, Greifswald, Germany
| | - Tibor Wittmann
- First Dept. of Medicine, University of Szeged, Szeged, Hungary
| | - Péter Hegyi
- First Dept. of Medicine, University of Szeged, Szeged, Hungary
| |
Collapse
|
26
|
Ceppa EP, Lyo V, Grady EF, Knecht W, Grahn S, Peterson A, Bunnett NW, Kirkwood KS, Cattaruzza F. Serine proteases mediate inflammatory pain in acute pancreatitis. Am J Physiol Gastrointest Liver Physiol 2011; 300:G1033-42. [PMID: 21436316 PMCID: PMC3774216 DOI: 10.1152/ajpgi.00305.2010] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Acute pancreatitis is a life-threatening inflammatory disease characterized by abdominal pain of unknown etiology. Trypsin, a key mediator of pancreatitis, causes inflammation and pain by activating protease-activated receptor 2 (PAR(2)), but the isoforms of trypsin that cause pancreatitis and pancreatic pain are unknown. We hypothesized that human trypsin IV and rat P23, which activate PAR(2) and are resistant to pancreatic trypsin inhibitors, contribute to pancreatic inflammation and pain. Injections of a subinflammatory dose of exogenous trypsin increased c-Fos immunoreactivity, indicative of spinal nociceptive activation, but did not cause inflammation, as assessed by measuring serum amylase and myeloperoxidase activity and by histology. The same dose of trypsin IV and P23 increased some inflammatory end points and caused a more robust effect on nociception, which was blocked by melagatran, a trypsin inhibitor that also inhibits polypeptide-resistant trypsin isoforms. To determine the contribution of endogenous activation of trypsin and its minor isoforms, recombinant enterokinase (ENK), which activates trypsins in the duodenum, was administered into the pancreas. Intraductal ENK caused nociception and inflammation that were diminished by polypeptide inhibitors, including soybean trypsin inhibitor and a specific trypsin inhibitor (type I-P), and by melagatran. Finally, the secretagogue cerulein induced pancreatic nociceptive activation and nocifensive behavior that were reversed by melagatran. Thus trypsin and its minor isoforms mediate pancreatic pain and inflammation. In particular, the inhibitor-resistant isoforms trypsin IV and P23 may be important in mediating prolonged pancreatic inflammatory pain in pancreatitis. Our results suggest that inhibitors of these isoforms could be novel therapies for pancreatitis pain.
Collapse
Affiliation(s)
- Eugene P. Ceppa
- 1Department of Surgery, Duke University Medical Center, Durham, North Carolina;
| | | | | | - Wolfgang Knecht
- 4Molecular Pharmacology and Lead Generation, AstraZeneca Research and Development, Mölndal, Sweden
| | | | - Anders Peterson
- 4Molecular Pharmacology and Lead Generation, AstraZeneca Research and Development, Mölndal, Sweden
| | - Nigel W. Bunnett
- Departments of 2Surgery and ,3Physiology, University of California, San Francisco, San Francisco, California; and
| | | | | |
Collapse
|
27
|
Proteinase-activated receptor 2 mediates thermal hyperalgesia and is upregulated in a rat model of chronic pancreatitis. Pancreas 2011; 40:300-7. [PMID: 21311307 DOI: 10.1097/mpa.0b013e318201cbc1] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
OBJECTIVES The mechanism of pain in chronic pancreatitis (CP) has yet to be explored. Proteinase-activated receptor 2 (PAR2) plays a pronociceptive role in visceral pain. The study aimed to assess the expression of PAR2 in dorsal root ganglia (DRGs) and validate its role of thermal hyperalgesia in CP. METHODS Chronic pancreatitis model was induced by trinitrobenzene sulfonic acid infusion into rat pancreatic ducts. Abdominal hyperalgesia was measured by thermal withdrawal latencies. The expression of PAR2 and transient receptor potential vanilloid 1 (TRPV1) were analyzed by immunofluorescence and Western blot. The messenger RNA encoding PAR2 was quantitated by real-time polymerase chain reaction. The effects of short-term and long-term ulinastatin treatment on abdominal thermal hyperalgesia of rats with CP were measured. RESULTS Rats with CP showed a decreased thermal withdrawal latency. Proteinase-activated receptor 2 and TRPV1 were significantly upregulated in DRGs. The increased PAR2 protein expression was tightly correlated with thermal withdrawal latencies and TRPV1 expression. Short-term ulinastatin treatment inhibited the development of thermal hyperalgesia of rats with CP in a dose-dependent manner. CONCLUSIONS The thermal hyperalgesia in CP is associated with an up-regulation of the PAR2 in DRGs. Proteinase-activated receptor 2 was involved in the pain generation in rats with CP.
Collapse
|
28
|
Takemura Y, Furuta S, Hirayama S, Miyashita K, Imai S, Narita M, Kuzumaki N, Tsukiyama Y, Yamazaki M, Suzuki T, Narita M. Upregulation of bradykinin receptors is implicated in the pain associated with caerulein-induced acute pancreatitis. Synapse 2010; 65:608-16. [DOI: 10.1002/syn.20880] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2010] [Accepted: 09/23/2010] [Indexed: 01/01/2023]
|
29
|
Nishimura S, Ishikura H, Matsunami M, Shinozaki Y, Sekiguchi F, Naruse M, Kitamura T, Akashi R, Matsumura K, Kawabata A. The proteinase/proteinase-activated receptor-2/transient receptor potential vanilloid-1 cascade impacts pancreatic pain in mice. Life Sci 2010; 87:643-50. [PMID: 20932849 DOI: 10.1016/j.lfs.2010.09.030] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2010] [Revised: 07/29/2010] [Accepted: 09/28/2010] [Indexed: 01/12/2023]
Abstract
AIMS Proteinase-activated receptor-2 (PAR2) and transient receptor potential vanilloid-1 (TRPV1) are co-localized in the primary afferents, and the trans-activation of TRPV1 by PAR2 activation is involved in processing of somatic pain. Given evidence for contribution of PAR2 to pancreatic pain, the present study aimed at clarifying the involvement of TRPV1 in processing of pancreatic pain by the proteinase/PAR2 pathway in mice. MAIN METHODS Acute pancreatitis was created by repeated administration of cerulein in conscious mice, and the referred allodynia/hyperalgesia was assessed using von Frey filaments. Injection of PAR2 agonists into the pancreatic duct was achieved in anesthetized mice, and expression of Fos in the spinal cord was determined by immunohistochemistry. KEY FINDINGS The established referred allodynia/hyperalgesia following cerulein treatment was abolished by post-treatment with nafamostat mesilate, a proteinase inhibitor, and with capsazepine, a TRPV1 antagonist, in mice. Injection of trypsin, an endogenous PAR2 agonist, or SLIGRL-NH(2), a PAR2-activating peptide, into the pancreatic duct caused expression of Fos protein in the spinal superficial layers at T8-T10 levels in the mice. The spinal Fos expression caused by trypsin and by SLIGRL-NH(2) was partially blocked by capsazepine, the former effect abolished by nafamostat mesilate. SIGNIFICANCE Our data thus suggest that the proteinase/PAR2/TRPV1 cascade might impact pancreatic pain, in addition to somatic pain, and play a role in the maintenance of pancreatitis-related pain in mice.
Collapse
Affiliation(s)
- Sachiyo Nishimura
- Division of Pharmacology and Pathophysiology, Kinki University School of Pharmacy, Higashi-Osaka, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Barry GD, Suen JY, Le GT, Cotterell A, Reid RC, Fairlie DP. Novel Agonists and Antagonists for Human Protease Activated Receptor 2. J Med Chem 2010; 53:7428-40. [DOI: 10.1021/jm100984y] [Citation(s) in RCA: 85] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Grant D. Barry
- Division of Chemistry and Structural Biology, Institute for Molecular Bioscience, The University of Queensland, Brisbane Qld 4072, Australia
| | - Jacky Y. Suen
- Division of Chemistry and Structural Biology, Institute for Molecular Bioscience, The University of Queensland, Brisbane Qld 4072, Australia
| | - Giang T. Le
- Division of Chemistry and Structural Biology, Institute for Molecular Bioscience, The University of Queensland, Brisbane Qld 4072, Australia
| | - Adam Cotterell
- Division of Chemistry and Structural Biology, Institute for Molecular Bioscience, The University of Queensland, Brisbane Qld 4072, Australia
| | - Robert C. Reid
- Division of Chemistry and Structural Biology, Institute for Molecular Bioscience, The University of Queensland, Brisbane Qld 4072, Australia
| | - David P. Fairlie
- Division of Chemistry and Structural Biology, Institute for Molecular Bioscience, The University of Queensland, Brisbane Qld 4072, Australia
| |
Collapse
|
31
|
Fukushima O, Nishimura S, Matsunami M, Aoki Y, Nishikawa H, Ishikura H, Kawabata A. Phosphorylation of ERK in the spinal dorsal horn following pancreatic pronociceptive stimuli with proteinase-activated receptor-2 agonists and hydrogen sulfide in rats: Evidence for involvement of distinct mechanisms. J Neurosci Res 2010; 88:3198-205. [DOI: 10.1002/jnr.22480] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
32
|
Tanaka Y, Sekiguchi F, Hong H, Kawabata A. PAR2 triggers IL-8 release via MEK/ERK and PI3-kinase/Akt pathways in GI epithelial cells. Biochem Biophys Res Commun 2008; 377:622-626. [PMID: 18854173 DOI: 10.1016/j.bbrc.2008.10.018] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2008] [Accepted: 10/07/2008] [Indexed: 12/18/2022]
Abstract
Proteinase-activated receptor-2 (PAR2) plays pro-inflammatory roles in many organs including the gastrointestinal (GI) tract. To clarify the downstream pro-inflammatory signaling of PAR2 in the GI tract, we examined interleukin-8 (IL-8) release and the underlying cellular signaling following PAR2 stimulation in human colorectal cancer-derived HCT-15 cells and human gastric adenocarcinoma-derived MKN-45 cells. A PAR2-activating peptide, but not a PAR2-inactive scrambled peptide or a PAR1- activating peptide, caused IL-8 release in these GI epithelial cells. The PAR2-triggered IL-8 release was suppressed by inhibitors of MEK (U0126) or PI3-kinase (LY294002), and PAR2 stimulation indeed activated the downstream kinases, ERK and Akt. U0126 blocked the phosphorylation of ERK, but not Akt, and LY294002 blocked the phosphorylation of Akt, but not ERK. Together, PAR2 triggers IL-8 release via two independent signaling pathways, MEK/ERK and PI3-kinase/Akt, suggesting a role of PAR2 as a pro-inflammatory receptor in the GI tract.
Collapse
Affiliation(s)
- Yusuke Tanaka
- Division of Pharmacology and Pathophysiology, Kinki University School of Pharmacy, 3-4-1 Kowakae, Higashi-Osaka 577-8502, Japan
| | - Fumiko Sekiguchi
- Division of Pharmacology and Pathophysiology, Kinki University School of Pharmacy, 3-4-1 Kowakae, Higashi-Osaka 577-8502, Japan
| | - Hao Hong
- Division of Pharmacology and Pathophysiology, Kinki University School of Pharmacy, 3-4-1 Kowakae, Higashi-Osaka 577-8502, Japan
| | - Atsufumi Kawabata
- Division of Pharmacology and Pathophysiology, Kinki University School of Pharmacy, 3-4-1 Kowakae, Higashi-Osaka 577-8502, Japan.
| |
Collapse
|
33
|
Gardell LR, Ma JN, Seitzberg JG, Knapp AE, Schiffer HH, Tabatabaei A, Davis CN, Owens M, Clemons B, Wong KK, Lund B, Nash NR, Gao Y, Lameh J, Schmelzer K, Olsson R, Burstein ES. Identification and characterization of novel small-molecule protease-activated receptor 2 agonists. J Pharmacol Exp Ther 2008; 327:799-808. [PMID: 18768780 DOI: 10.1124/jpet.108.142570] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
We report the first small-molecule protease-activated receptor (PAR) 2 agonists, AC-55541 [N-[[1-(3-bromo-phenyl)-eth-(E)-ylidene-hydrazinocarbonyl]-(4-oxo-3,4-dihydro-phthalazin-1-yl)-methyl]-benzamide] and AC-264613 [2-oxo-4-phenylpyrrolidine-3-carboxylic acid [1-(3-bromo-phenyl)-(E/Z)-ethylidene]-hydrazide], each representing a distinct chemical series. AC-55541 and AC-264613 each activated PAR2 signaling in cellular proliferation assays, phosphatidylinositol hydrolysis assays, and Ca(2+) mobilization assays, with potencies ranging from 200 to 1000 nM for AC-55541 and 30 to 100 nM for AC-264613. In comparison, the PAR2-activating peptide 2-furoyl-LIGRLO-NH(2) had similar potency, whereas SLIGRL-NH(2) was 30 to 300 times less potent. Neither AC-55541 nor AC-264613 had activity at any of the other PAR receptor subtypes, nor did they have any significant affinity for over 30 other molecular targets involved in nociception. Visualization of EYFP-tagged PAR2 receptors showed that each compound stimulated internalization of PAR2 receptors. AC-55541 and AC-264613 were well absorbed when administered intraperitoneally to rats, each reaching micromolar peak plasma concentrations. AC-55541 and AC-264613 were each stable to metabolism by liver microsomes and maintained sustained exposure in rats, with elimination half-lives of 6.1 and 2.5 h, respectively. Intrapaw administration of AC-55541 or AC-264613 elicited robust and persistent thermal hyperalgesia and edema. Coadministration of either a tachykinin 1 (neurokinin 1) receptor antagonist or a transient receptor potential vanilloid (TRPV) 1 antagonist completely blocked these effects. Systemic administration of either AC-55541 or AC-264613 produced a similar degree of hyperalgesia as was observed when the compounds were administered locally. These compounds represent novel small-molecule PAR2 agonists that will be useful in probing the physiological functions of PAR2 receptors.
Collapse
|
34
|
Guerios SD, Wang ZY, Boldon K, Bushman W, Bjorling DE. Blockade of NGF and trk receptors inhibits increased peripheral mechanical sensitivity accompanying cystitis in rats. Am J Physiol Regul Integr Comp Physiol 2008; 295:R111-22. [PMID: 18448607 DOI: 10.1152/ajpregu.00728.2007] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Visceral inflammation, including that arising from bladder inflammation, reduces the threshold to sensation of innocuous or noxious stimuli applied to peripheral structures (referred hyperalgesia). Cystitis may induce transient or persistent plastic changes mediated by neurotrophins, particularly nerve growth factor (NGF), which contribute to increased nociceptive input. In this study, acute or subacute cystitis was induced in female rats by one or three (at 72-h intervals) 400-microl intravesical instillations of 1 mM acrolein. Sensitivity of the hindpaws to mechanical and thermal stimuli was determined before and 4, 24, 48, 72, and 96 h after treatment. Other groups of rats were treated with intravesical or intrathecal k252a [a nonspecific antagonist of tyrosine kinase (trk) receptors, including trkA, the high-affinity receptor for NGF] before the first or third acrolein instillation. Some rats were intraperitoneally injected with specific NGF-neutralizing antiserum or normal serum before acrolein instillation. Acute and subacute cystitis induced mechanical, but not thermal, referred hyperalgesia that was attenuated by intravesical pretreatment with k252a. Systemic treatment with NGF-neutralizing antiserum before instillation of acrolein suppressed subsequent mechanical referred hyperalgesia. Expression of NGF was increased within the bladder by acute or subacute cystitis and in L6/S1 dorsal root ganglia by subacute cystitis. These results suggest that the bladder-derived NGF acting via trk receptors at least partially mediates peripheral sensitization to mechanical stimuli associated with acute and subacute acrolein-induced cystitis.
Collapse
Affiliation(s)
- Simone D Guerios
- Department of Surgical Sciences, School of Veterinary Medicine, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin, USA
| | | | | | | | | |
Collapse
|
35
|
Yang H, McNearney TA, Chu R, Lu Y, Ren Y, Yeomans DC, Wilson SP, Westlund KN. Enkephalin-encoding herpes simplex virus-1 decreases inflammation and hotplate sensitivity in a chronic pancreatitis model. Mol Pain 2008; 4:8. [PMID: 18307791 PMCID: PMC2292157 DOI: 10.1186/1744-8069-4-8] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2008] [Accepted: 02/28/2008] [Indexed: 12/15/2022] Open
Abstract
Background A chronic pancreatitis model was developed in young male Lewis rats fed a high-fat and alcohol liquid diet beginning at three weeks. The model was used to assess time course and efficacy of a replication defective herpes simplex virus type 1 vector construct delivering human cDNA encoding preproenkephalin (HSV-ENK). Results Most surprising was the relative lack of inflammation and tissue disruption after HSV-ENK treatment compared to the histopathology consistent with pancreatitis (inflammatory cell infiltration, edema, acinar cell hypertrophy, fibrosis) present as a result of the high-fat and alcohol diet in controls. The HSV-ENK vector delivered to the pancreatic surface at week 3 reversed pancreatitis-associated hotplate hypersensitive responses for 4–6 weeks, while control virus encoding β-galactosidase cDNA (HSV-β-gal) had no effect. Increased Fos expression seen bilaterally in pain processing regions in control animals with pancreatitis was absent in HSV-ENK-treated animals. Increased met-enkephalin staining was evident in pancreas and lower thoracic spinal cord laminae I–II in the HSV-ENK-treated rats. Conclusion Thus, clear evidence is provided that site specific HSV-mediated transgene delivery of human cDNA encoding preproenkephalin ameliorates pancreatic inflammation and significantly reduces hypersensitive hotplate responses for an extended time consistent with HSV mediated overexpression, without tolerance or evidence of other opiate related side effects.
Collapse
Affiliation(s)
- Hong Yang
- Dept of Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, TX, USA.
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Kawabata A, Matsunami M, Sekiguchi F. Gastrointestinal roles for proteinase-activated receptors in health and disease. Br J Pharmacol 2007; 153 Suppl 1:S230-40. [PMID: 17994114 DOI: 10.1038/sj.bjp.0707491] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
It has been almost a decade since the molecular cloning of all four members of the proteinase-activated receptor (PAR) family was completed. This unique family of G protein-coupled receptors (GPCRs) mediates specific cellular actions of various endogenous proteinases including thrombin, trypsin, tryptase, etc. and also certain exogenous enzymes. Increasing evidence has been clarifying the emerging roles played by PARs in health and disease. PARs, particularly PAR1 and PAR2, are distributed throughout the gastrointestinal (GI) tract, modulating various GI functions. One of the most important GI functions of PARs is regulation of exocrine secretion in the salivary glands, pancreas and GI mucosal epithelium. PARs also modulate motility of GI smooth muscle, involving multiple mechanisms. PAR2 appears to play dual roles in pancreatitis and related pain, being pro-inflammatory/pro-nociceptive and anti-inflammatory/anti-nociceptive. Similarly, dual roles for PAR1 and PAR2 have been demonstrated in mucosal inflammation/damage throughout the GI tract. There is also fundamental and clinical evidence for involvement of PAR2 in colonic pain. PARs are thus considered key molecules in regulation of GI functions and targets for development of drugs for treatment of various GI diseases.
Collapse
Affiliation(s)
- A Kawabata
- Division of Pharmacology and Pathophysiology, Kinki University School of Pharmacy, Higashi-Osaka, Japan.
| | | | | |
Collapse
|
37
|
Barry GD, Suen JY, Low HB, Pfeiffer B, Flanagan B, Halili M, Le GT, Fairlie DP. A refined agonist pharmacophore for protease activated receptor 2. Bioorg Med Chem Lett 2007; 17:5552-7. [PMID: 17765542 DOI: 10.1016/j.bmcl.2007.08.026] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2007] [Revised: 08/10/2007] [Accepted: 08/13/2007] [Indexed: 11/20/2022]
Abstract
Protease activated receptor 2 (PAR(2)) is a G protein-coupled receptor implicated in inflammation and cancer. Only a few peptide agonists are known with greater potency than the native agonist SLIGRL-NH(2). Here we report 52 peptide agonists of PAR(2), 26 with activity at sub-micromolar concentrations, and one being iodinated for radioligand experiments. Potency was highest when the N- or C-termini of SLIGRL-NH(2) were modified, pointing to a new ligand pharmacophore model that may aid development of drug-like PAR(2) modulators.
Collapse
Affiliation(s)
- Grant D Barry
- Centre for Drug Design and Development, Institute for Molecular Bioscience, University of Queensland, Brisbane, Qld. 4072, Australia
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Fregni F, Pascual-Leone A, Freedman SD. Pain in chronic pancreatitis: a salutogenic mechanism or a maladaptive brain response? Pancreatology 2007; 7:411-22. [PMID: 17898531 PMCID: PMC2826873 DOI: 10.1159/000108958] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Pain in chronic pancreatitis is frequently refractory to medical and even surgical treatment. This refractoriness leads us to believe that a pancreas-independent, brain-mediated mechanism must be responsible. If so, several scenarios are worth considering. First, chronic pain could be the consequence of undesirable neuroplastic changes, by which pathology becomes established and causes disability. Alternatively, pain may be linked to the salutogenic (from salutogenesis, the Latin word for health and well-being) central nervous system response (we defined 'salutogenic response' as the specific modulation of the immune system induced by brain activity changes) to promote healing of the injured viscera. If so, chronic pain could index the ongoing nervous system attempt to promote healing. In this review, we discuss (1) the mechanisms of pain in chronic pancreatitis; (2) potential brain-related salutogenic mechanisms, and (3) the potential relationship of these two factors to the disease status. Furthermore, we consider these aspects in light of a new approach to treat visceral pain: transcranial magnetic stimulation, a noninvasive method of brain stimulation.
Collapse
Affiliation(s)
- Felipe Fregni
- Departments of Medicine, Harvard Medical School, Boston, Mass., USA
- Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Mass., USA
| | - Alvaro Pascual-Leone
- Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Mass., USA
| | - Steven D. Freedman
- Departments of Medicine, Harvard Medical School, Boston, Mass., USA
- *Steven Freedman, MD, PhD, Department of Medicine, 330 Brookline Ave – FN 204, Boston, MA 02215 (USA), Tel. +1 617 667 2581, Fax +1 617 667 0536, E-Mail
| |
Collapse
|
39
|
Ishikura H, Nishimura S, Matsunami M, Tsujiuchi T, Ishiki T, Sekiguchi F, Naruse M, Nakatani T, Kamanaka Y, Kawabata A. The proteinase inhibitor camostat mesilate suppresses pancreatic pain in rodents. Life Sci 2007; 80:1999-2004. [PMID: 17433371 DOI: 10.1016/j.lfs.2007.02.044] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2006] [Revised: 02/20/2007] [Accepted: 02/28/2007] [Indexed: 02/01/2023]
Abstract
Camostat mesilate, an orally available proteinase inhibitor, is clinically used for treatment of pancreatitis. Given recent evidence that pancreatic proteinases including trypsin and/or proteinase-activated receptor-2 (PAR2) might be involved in pancreatic pain, we examined if camostat mesilate could suppress spinal Fos expression, a marker for neuronal activation, following specific application of trypsin to the pancreas, and pancreatitis-related referred allodynia. Trypsin, administered into the pancreatic duct, caused delayed expression of Fos proteins in the superficial layer of the bilateral T8 and T9 spinal dorsal horns in rats. The trypsin-induced spinal Fos expression was completely abolished by oral pre-administration of camostat mesilate at 300 mg/kg. After hourly repeated (6 times in total) administration of caerulein, mice showed typical symptoms of pancreatitis, accompanied by mechanical allodynia in the upper abdomen (i.e., referred hyperalgesia/allodynia), as assessed by use of von Frey filaments. Camostat mesilate at 100-300 mg/kg, given orally twice before the 1st and 4th doses of caerulein, abolished the pancreatitis-related abdominal allodynia, while it partially prevented the inflammatory signs. The same doses of camostat mesilate, when administered once after the final dose of caerulein, also revealed significant anti-allodynic effect. These data suggest that camostat mesilate prevents and/or depresses pancreatitis-induced pain and/or referred hyperalgesia/allodynia, in which proteinases including trypsin would play a critical role.
Collapse
Affiliation(s)
- Hiroyasu Ishikura
- Division of Endocrinology, Clinical Research Institute for Endocrine and Metabolic Diseases, National Hospital Organization, Kyoto Medical Center, 1-1 Fukakusa, Fushimiku 612-8555, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Michalski CW, Laukert T, Sauliunaite D, Pacher P, Bergmann F, Agarwal N, Su Y, Giese T, Giese NA, Bátkai S, Friess H, Kuner R. Cannabinoids ameliorate pain and reduce disease pathology in cerulein-induced acute pancreatitis. Gastroenterology 2007; 132:1968-78. [PMID: 17484889 PMCID: PMC2268094 DOI: 10.1053/j.gastro.2007.02.035] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2006] [Accepted: 01/24/2007] [Indexed: 12/11/2022]
Abstract
BACKGROUND & AIMS The functional involvement of the endocannabinoid system in modulation of pancreatic inflammation, such as acute pancreatitis, has not been studied to date. Moreover, the therapeutic potential of cannabinoids in pancreatitis has not been addressed. METHODS We quantified endocannabinoid levels and expression of cannabinoid receptors 1 and 2 (CB1 and CB2) in pancreas biopsies from patients and mice with acute pancreatitis. Functional studies were performed in mice using pharmacological interventions. Histological examination, serological, and molecular analyses (lipase, myeloperoxidase, cytokines, and chemokines) were performed to assess disease pathology and inflammation. Pain resulting from pancreatitis was studied as abdominal hypersensitivity to punctate von Frey stimuli. Behavioral analyses in the open-field, light-dark, and catalepsy tests were performed to judge cannabinoid-induced central side effects. RESULTS Patients with acute pancreatitis showed an up-regulation of cannabinoid receptors and elevated levels of endocannabinoids in the pancreas. HU210, a synthetic agonist at CB1 and CB2, abolished abdominal pain associated with pancreatitis and also reduced inflammation and decreased tissue pathology in mice without producing central, adverse effects. Antagonists at CB1- and CB2-receptors were effective in reversing HU210-induced antinociception, whereas a combination of CB1- and CB2-antagonists was required to block the anti-inflammatory effects of HU210 in pancreatitis. CONCLUSIONS In humans, acute pancreatitis is associated with up-regulation of ligands as well as receptors of the endocannabinoid system in the pancreas. Furthermore, our results suggest a therapeutic potential for cannabinoids in abolishing pain associated with acute pancreatitis and in partially reducing inflammation and disease pathology in the absence of adverse side effects.
Collapse
MESH Headings
- Acute Disease
- Animals
- Biopsy
- Cannabinoid Receptor Modulators/physiology
- Cannabinoids/adverse effects
- Cannabinoids/therapeutic use
- Ceruletide
- Dronabinol/adverse effects
- Dronabinol/analogs & derivatives
- Dronabinol/pharmacology
- Female
- Gene Expression Regulation
- Humans
- Male
- Mice
- Mice, Inbred C57BL
- Neuroprotective Agents/adverse effects
- Neuroprotective Agents/pharmacology
- Pain/drug therapy
- Pain/etiology
- Pancreas/metabolism
- Pancreas/pathology
- Pancreatitis/chemically induced
- Pancreatitis/complications
- Pancreatitis/drug therapy
- Pancreatitis/pathology
- Receptor, Cannabinoid, CB1/agonists
- Receptor, Cannabinoid, CB1/genetics
- Receptor, Cannabinoid, CB1/metabolism
- Receptor, Cannabinoid, CB2/agonists
- Receptor, Cannabinoid, CB2/genetics
- Receptor, Cannabinoid, CB2/metabolism
Collapse
|
41
|
Liu J, Li F. New developments in the relationship between protease activated recerptor-2 and alimentary system diseases. Shijie Huaren Xiaohua Zazhi 2007; 15:986-990. [DOI: 10.11569/wcjd.v15.i9.986] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Protease-activated receptor-2 (PAR-2) is a G-protein-coupled receptor, and its special molecule structure and activation way are similar to other protease-activated receptors. PAR-2 is widely distributed in alimentary system and series of effects are produced when it is activated by certain proteases. For example, PAR-2 exerts gastric mucosal cytoprotective effect, influences the secretion of digestive glands and movement of gastrointestinal tract, participates in the development of pancreatitis, and associates closely with malignant tumors in alimentary system. More and more researches are now focusing on the relationship between PAR-2 and alimentary system diseases.
Collapse
|
42
|
Kawabata A. [Proteinase-activated receptors and gastrointestinal functions]. Nihon Yakurigaku Zasshi 2006; 128:82-7. [PMID: 16943642 DOI: 10.1254/fpj.128.82] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
43
|
Lechin F, van der Dijs B. Central nervous system circuitry involved in the hyperinsulinism syndrome. Neuroendocrinology 2006; 84:222-34. [PMID: 17167239 DOI: 10.1159/000098005] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2006] [Accepted: 10/30/2006] [Indexed: 12/29/2022]
Abstract
Raised plasma levels of insulin, glucose and glucagon are found in patients affected by 'hyperinsulinism'. Obesity, hypertension, mammary plus ovary cysts and rheumatic symptoms are frequently observed in these patients. Sleep disorders and depression are also present in most subjects affected by this polysymptomatic disorder. The simultaneous increases of glucose, insulin and glucagon plasma levels seen in these patients indicate that the normal crosstalk between A cells, B cells and D cells is disrupted. With respect to this, it is well known that glucose excites B cells (which secrete insulin) and inhibits A cells (which secrete glucagon), which in turn excites D cells (which secrete somatostatin). Gastrointestinal hormones (incretins) modulate this crosstalk both directly and indirectly throughout pancreatic and hepatobiliary mechanisms. The above factors depend on autonomic nervous system mediation. For instance, acetylcholine released from parasympathetic nerves excites both B and A cells. Noradrenaline released from sympathetic nerves and adrenaline secreted from the adrenal glands inhibit B cells and excite A cells, which are crowded with beta(2)- and alpha(2)-receptors, respectively. Noradrenaline released from sympathetic nerves also excites A cells by acting at alpha(1)-receptors located at this level. According to this, the excessive release of noradrenaline from these nerves should provoke an enhancement of glucagon secretion which will result in overexcitation of insulin secretion from B cells. That is the disorder seen in the so-called 'hyperinsulinism', in which raised plasma levels of glucose, insulin and glucagon coexist. Taking into account that neural sympathetic activity is positively correlated to the A5 noradrenergic nucleus and median raphe serotonergic neurons, and negatively correlated to the A6 noradrenergic, the dorsal raphe serotonergic and the C1 adrenergic neurons, we postulate that this unbalanced central nervous system circuitry is responsible for the hyperinsulinism syndrome.
Collapse
Affiliation(s)
- Fuad Lechin
- Department of Physiological Sciences, Sections of Neurochemistry, Neurophysiology, Neuroimmunology and Neuropharmacology, Instituto de Medicina Experimental, Universidad Central de Venezuela, Caracas, Venezuela.
| | | |
Collapse
|