1
|
Liu W, Zhang W, Wang C, Song J, Li K, Zhang X, Wu X, Guo H. TRPV4 antagonist suppresses retinal ganglion cell apoptosis by regulating the activation of CaMKII and TNF-α expression in a chronic ocular hypertension rat model. Int Immunopharmacol 2024; 130:111811. [PMID: 38457929 DOI: 10.1016/j.intimp.2024.111811] [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: 01/13/2024] [Revised: 02/24/2024] [Accepted: 03/04/2024] [Indexed: 03/10/2024]
Abstract
Glaucoma is characterized by a progressive loss of retinal ganglion cells (RGCs), leading to irreversible visual function impairment. Sustained increase in intraocular pressure represents a major risk factor for glaucoma, yet the underlying mechanisms of RGC apoptosis induced by intraocular pressure remains unclear. This study aims to investigate the role of TRPV4 in RGC apoptosis in a rat model of chronic ocular hypertension (COH) and the underlying molecular mechanism. In the COH rat models, we evaluated the visual function, retinal pathological changes and RGC apoptosis. TRPV4 expression and downstream signaling molecules were also detected. We found that RGC density decreased and RGC apoptosis was induced in COH eyes compared with control eyes. TRPV4 expression increased significantly in response to elevated IOP. TRPV4 inhibition by the TRPV4 antagonist HC-067047 (HC-067) suppressed RGC apoptosis and protected visual function. HC-067 treatment upregulated the phosphorylation of CaMKII in both control and COH eyes. Finally, HC-067 treatment suppressed the production of TNF-α induced by ocular hypertension. The TRPV4 antagonist HC-067 might suppress RGC apoptosis by regulating the activation of CaMKII and inhibiting the production of TNF-α in the COH model. This indicated that TRPV4 antagonists may be a potential and novel therapeutic strategy for glaucoma.
Collapse
Affiliation(s)
- Wenhui Liu
- Department of Ophthalmology, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
| | - Wenzhe Zhang
- Department of Ophthalmology, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
| | - Chen Wang
- Department of Ophthalmology, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
| | - Jiarun Song
- Department of Ophthalmology, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
| | - Kaiyue Li
- Department of Nuclear Medicine, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
| | - Xia Zhang
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| | - Xinyi Wu
- Department of Ophthalmology, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China.
| | - Hui Guo
- Department of Ophthalmology, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China.
| |
Collapse
|
2
|
Jesus RLC, Araujo FA, Alves QL, Dourado KC, Silva DF. Targeting temperature-sensitive transient receptor potential channels in hypertension: far beyond the perception of hot and cold. J Hypertens 2023; 41:1351-1370. [PMID: 37334542 DOI: 10.1097/hjh.0000000000003487] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Transient receptor potential (TRP) channels are nonselective cation channels and participate in various physiological roles. Thus, changes in TRP channel function or expression have been linked to several disorders. Among the many TRP channel subtypes, the TRP ankyrin type 1 (TRPA1), TRP melastatin type 8 (TRPM8), and TRP vanilloid type 1 (TRPV1) channels are temperature-sensitive and recognized as thermo-TRPs, which are expressed in the primary afferent nerve. Thermal stimuli are converted into neuronal activity. Several studies have described the expression of TRPA1, TRPM8, and TRPV1 in the cardiovascular system, where these channels can modulate physiological and pathological conditions, including hypertension. This review provides a complete understanding of the functional role of the opposing thermo-receptors TRPA1/TRPM8/TRPV1 in hypertension and a more comprehensive appreciation of TRPA1/TRPM8/TRPV1-dependent mechanisms involved in hypertension. These channels varied activation and inactivation have revealed a signaling pathway that may lead to innovative future treatment options for hypertension and correlated vascular diseases.
Collapse
Affiliation(s)
- Rafael Leonne C Jesus
- Laboratory of Cardiovascular Physiology and Pharmacology, Federal University of Bahia, Salvador
| | - Fênix A Araujo
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation - FIOCRUZ, Bahia, Brazil
| | - Quiara L Alves
- Laboratory of Cardiovascular Physiology and Pharmacology, Federal University of Bahia, Salvador
| | - Keina C Dourado
- Laboratory of Cardiovascular Physiology and Pharmacology, Federal University of Bahia, Salvador
| | - Darizy F Silva
- Laboratory of Cardiovascular Physiology and Pharmacology, Federal University of Bahia, Salvador
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation - FIOCRUZ, Bahia, Brazil
| |
Collapse
|
3
|
Tang L, Gao J, Cao X, Chen L, Wang H, Ding H. TRPV1 mediates itch-associated scratching and skin barrier dysfunction in DNFB-induced atopic dermatitis mice. Exp Dermatol 2021; 31:398-405. [PMID: 34608683 DOI: 10.1111/exd.14464] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 08/05/2021] [Accepted: 09/30/2021] [Indexed: 11/29/2022]
Abstract
In chronic pruritic diseases such as atopic dermatitis (AD), pruritus and skin lesions are exacerbated by scratching in clinical and experimental settings. TRPV1 is known to mediate itch and neurogenic inflammation, but the role of TRPV1 in itch-associated scratching in AD is poorly understood. In this study, we examined the efficacy of cutting off nails and TRPV1 antagonist, ruthenium red (RR) in a murine model of AD induced by DNFB and further investigated the underlying mechanism. Nail clipping or RR could markedly ameliorate the general AD-like symptoms as manifested by the reduced clinical severity of dermatitis, IgE and Th2-related cytokine levels, and mast cell degranulation. Moreover, scratching behaviour, the levels of pruritogenic mediators, including HIS, TSLP, IL-31 and SP, and skin pH and TEWL were all significantly decreased in nail clipping or RR-treated mice, suggesting a reduction in itch-associated scratching and skin barrier defects. Immunofluorescence staining and Western blot results revealed that antipruritic effect of nail clipping or RR in AD may be explained, at least in part, by the suppression of TRPV1 activation. In summary, these data show that TRPV1 mediates itch-associated scratching and subsequent skin barrier defects, suggesting its potential as a therapeutic target in AD.
Collapse
Affiliation(s)
- Liu Tang
- Department of Pharmacy, Renmin Hospital of Wuhan University, Wuhan, China.,School of Pharmaceutical Sciences, Wuhan University, Wuhan, China
| | - Jiefang Gao
- School of Pharmaceutical Sciences, Wuhan University, Wuhan, China
| | - Xiaoqin Cao
- College of Medicine, Jianghan University, Wuhan, China
| | - Lu Chen
- School of Pharmaceutical Sciences, Wuhan University, Wuhan, China
| | - Huiling Wang
- School of Pharmaceutical Sciences, Wuhan University, Wuhan, China
| | - Hong Ding
- School of Pharmaceutical Sciences, Wuhan University, Wuhan, China
| |
Collapse
|
4
|
Wu AJ, Tong BCK, Huang AS, Li M, Cheung KH. Mitochondrial Calcium Signaling as a Therapeutic Target for Alzheimer's Disease. Curr Alzheimer Res 2021; 17:329-343. [PMID: 31820698 DOI: 10.2174/1567205016666191210091302] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Revised: 10/17/2019] [Accepted: 12/09/2019] [Indexed: 11/22/2022]
Abstract
Mitochondria absorb calcium (Ca2+) at the expense of the electrochemical gradient generated during respiration. The influx of Ca2+ into the mitochondrial matrix helps maintain metabolic function and results in increased cytosolic Ca2+ during intracellular Ca2+ signaling. Mitochondrial Ca2+ homeostasis is tightly regulated by proteins located in the inner and outer mitochondrial membranes and by the cross-talk with endoplasmic reticulum Ca2+ signals. Increasing evidence indicates that mitochondrial Ca2+ overload is a pathological phenotype associated with Alzheimer's Disease (AD). As intracellular Ca2+ dysregulation can be observed before the appearance of typical pathological hallmarks of AD, it is believed that mitochondrial Ca2+ overload may also play an important role in AD etiology. The high mitochondrial Ca2+ uptake can easily compromise neuronal functions and exacerbate AD progression by impairing mitochondrial respiration, increasing reactive oxygen species formation and inducing apoptosis. Additionally, mitochondrial Ca2+ overload can damage mitochondrial recycling via mitophagy. This review will discuss the molecular players involved in mitochondrial Ca2+ dysregulation and the pharmacotherapies that target this dysregulation. As most of the current AD therapeutics are based on amyloidopathy, tauopathy, and the cholinergic hypothesis, they achieve only symptomatic relief. Thus, determining how to reestablish mitochondrial Ca2+ homeostasis may aid in the development of novel AD therapeutic interventions.
Collapse
Affiliation(s)
- Aston J Wu
- School of Chinese Medicine, Hong Kong Baptist University, 7 Baptist University Road, Kowloon Tong, Kowloon, Hong Kong, China.,Mr. and Mrs. Ko Chi Ming Centre for Parkinson's Disease Research, Hong Kong Baptist University, 7 Baptist University Road, Kowloon Tong, Kowloon, Hong Kong, China
| | - Benjamin C-K Tong
- School of Chinese Medicine, Hong Kong Baptist University, 7 Baptist University Road, Kowloon Tong, Kowloon, Hong Kong, China.,Mr. and Mrs. Ko Chi Ming Centre for Parkinson's Disease Research, Hong Kong Baptist University, 7 Baptist University Road, Kowloon Tong, Kowloon, Hong Kong, China
| | - Alexis S Huang
- School of Chinese Medicine, Hong Kong Baptist University, 7 Baptist University Road, Kowloon Tong, Kowloon, Hong Kong, China.,Mr. and Mrs. Ko Chi Ming Centre for Parkinson's Disease Research, Hong Kong Baptist University, 7 Baptist University Road, Kowloon Tong, Kowloon, Hong Kong, China
| | - Min Li
- School of Chinese Medicine, Hong Kong Baptist University, 7 Baptist University Road, Kowloon Tong, Kowloon, Hong Kong, China.,Mr. and Mrs. Ko Chi Ming Centre for Parkinson's Disease Research, Hong Kong Baptist University, 7 Baptist University Road, Kowloon Tong, Kowloon, Hong Kong, China
| | - King-Ho Cheung
- School of Chinese Medicine, Hong Kong Baptist University, 7 Baptist University Road, Kowloon Tong, Kowloon, Hong Kong, China.,Mr. and Mrs. Ko Chi Ming Centre for Parkinson's Disease Research, Hong Kong Baptist University, 7 Baptist University Road, Kowloon Tong, Kowloon, Hong Kong, China
| |
Collapse
|
5
|
Guarino BD, Paruchuri S, Thodeti CK. The role of TRPV4 channels in ocular function and pathologies. Exp Eye Res 2020; 201:108257. [PMID: 32979394 DOI: 10.1016/j.exer.2020.108257] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 09/17/2020] [Accepted: 09/18/2020] [Indexed: 12/15/2022]
Abstract
Transient potential receptor vanilloid 4 (TRPV4) is an ion channel responsible for sensing osmotic and mechanical signals, which in turn regulates calcium signaling across cell membranes. TRPV4 is widely expressed throughout the body, and plays an important role in normal physiological function, as well as different pathologies, however, its role in the eye is not well known. In the eye, TRPV4 is expressed in various tissues, such as the retina, corneal epithelium, ciliary body, and the lens. In this review, we provide an overview on TRPV4 structure, activation, mutations, and summarize the current knowledge of TRPV4 function and signaling mechanisms in various locations throughout the eye, as well as its role in ocular diseases, such as glaucoma and diabetic retinopathy. Based on the available data, we highlight the therapeutic potential of TRPV4 as well as the shortcomings of current research. Finally, we provide future perspectives on the implications of targeting TRPV4 to treat various ocular pathologies.
Collapse
Affiliation(s)
- Brianna D Guarino
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH, 44272, USA
| | | | - Charles K Thodeti
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH, 44272, USA.
| |
Collapse
|
6
|
Yang W, Wang Y, Guo J, He L, Zhou Y, Zheng H, Liu Z, Zhu P, Zhang XC. Cryo-electron microscopy structure of CLHM1 ion channel from Caenorhabditis elegans. Protein Sci 2020; 29:1803-1815. [PMID: 32557855 PMCID: PMC7380676 DOI: 10.1002/pro.3904] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 06/15/2020] [Accepted: 06/16/2020] [Indexed: 02/02/2023]
Abstract
Calcium homeostasis modulators (CALHMs/CLHMs) comprise a family of pore-forming protein complexes assembling into voltage-gated, Ca2+ -sensitive, nonselective channels. These complexes contain an ion-conduction pore sufficiently wide to permit the passing of ATP molecules serving as neurotransmitters. While their function and structure information is accumulating, the precise mechanisms of these channel complexes remain to be full understood. Here, we present the structure of the Caenorhabditis elegans CLHM1 channel in its open state solved through single-particle cryo-electron microscopy at 3.7-Å resolution. The transmembrane region of the channel structure of the dominant class shows an assembly of 10-fold rotational symmetry in one layer, and its cytoplasmic region is involved in additional twofold symmetrical packing in a tail-to-tail manner. Furthermore, we identified a series of amino acid residues critical for the regulation of CeCLHM1 channel using functional assays, electrophysiological analyses as well as structural-based analysis. Our structure and function analyses provide new insights into the mechanisms of CALHM channels.
Collapse
Affiliation(s)
- Weixin Yang
- National Laboratory of Biomacromolecules, CAS Center for Excellence in BiomacromoleculesInstitute of Biophysics, Chinese Academy of SciencesBeijingChina
- College of Life SciencesUniversity of Chinese Academy of SciencesBeijingChina
| | - Youwang Wang
- National Laboratory of Biomacromolecules, CAS Center for Excellence in BiomacromoleculesInstitute of Biophysics, Chinese Academy of SciencesBeijingChina
- College of Life SciencesUniversity of Chinese Academy of SciencesBeijingChina
| | - Jianli Guo
- National Laboratory of Biomacromolecules, CAS Center for Excellence in BiomacromoleculesInstitute of Biophysics, Chinese Academy of SciencesBeijingChina
| | - Lingli He
- National Laboratory of Biomacromolecules, CAS Center for Excellence in BiomacromoleculesInstitute of Biophysics, Chinese Academy of SciencesBeijingChina
| | - Ye Zhou
- National Laboratory of Biomacromolecules, CAS Center for Excellence in BiomacromoleculesInstitute of Biophysics, Chinese Academy of SciencesBeijingChina
| | - Hui Zheng
- National Laboratory of Biomacromolecules, CAS Center for Excellence in BiomacromoleculesInstitute of Biophysics, Chinese Academy of SciencesBeijingChina
| | - Zhenfeng Liu
- National Laboratory of Biomacromolecules, CAS Center for Excellence in BiomacromoleculesInstitute of Biophysics, Chinese Academy of SciencesBeijingChina
- College of Life SciencesUniversity of Chinese Academy of SciencesBeijingChina
| | - Ping Zhu
- National Laboratory of Biomacromolecules, CAS Center for Excellence in BiomacromoleculesInstitute of Biophysics, Chinese Academy of SciencesBeijingChina
- College of Life SciencesUniversity of Chinese Academy of SciencesBeijingChina
| | - Xuejun C. Zhang
- National Laboratory of Biomacromolecules, CAS Center for Excellence in BiomacromoleculesInstitute of Biophysics, Chinese Academy of SciencesBeijingChina
- College of Life SciencesUniversity of Chinese Academy of SciencesBeijingChina
| |
Collapse
|
7
|
Entin-Meer M, Keren G. Potential roles in cardiac physiology and pathology of the cation channel TRPV2 expressed in cardiac cells and cardiac macrophages: a mini-review. Am J Physiol Heart Circ Physiol 2019; 318:H181-H188. [PMID: 31809212 DOI: 10.1152/ajpheart.00491.2019] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
TRPV2 is a well-conserved channel protein expressed in almost all tissues. Cardiomyocyte TRPV2 is expressed in the intercalated disks of the cardiac sarcomeres, where it is involved in maintaining the proper mechanoelectric coupling and structure. It is also abundantly expressed in the intracellular pools, mainly the endoplasmic reticulum. Under pathological conditions, TRPV2 is translocated to the sarcolemma, where it mediates an abnormal [Ca]2+ entry that may contribute to disease progression. In addition, an intracellularly diffused TRPV2 expression is present in resident cardiac macrophages. Upon infection or inflammation, TRPV2 is engaged in early phagosomes and is, therefore, potentially involved in protecting the cardiac tissue. Following acute myocardial infarction, a profound elevated expression of TRPV2 is observed on the cell membrane of the peri-infarct macrophages. The macrophage TRPV2 may harbor a detrimental effect in cardiac recovery by increasing unfavorable migration and phagocytosis processes in the injured heart. Most reports suggest that while cardiac TRPV2 activation may be beneficial under specific physiological conditions, both cardiac- and macrophage-related TRPV2 blocking can significantly ameliorate disease progression in various pathological states. To verify this possibility, the time frame of TRPV2 overexpression and its mediated signaling need to be fully characterized in both cardiomyocyte and cardiac macrophage populations.
Collapse
Affiliation(s)
- Michal Entin-Meer
- Cardiovascular Research Laboratory, Tel Aviv Sourasky Medical Center, affiliated with the Sackler School of Medicine, Tel-Aviv, Israel
| | - Gad Keren
- Cardiovascular Research Laboratory, Tel Aviv Sourasky Medical Center, affiliated with the Sackler School of Medicine, Tel-Aviv, Israel
| |
Collapse
|
8
|
Reyes-Pardo H, Sánchez-Herrera DP. Mechanosensitive ion channel inhibitors promote the stiffening of the plasma membrane of mouse sensory neurons. SOFT MATTER 2019; 15:8320-8328. [PMID: 31565715 DOI: 10.1039/c9sm01230c] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
The mechanosensitivity of cells depends on the lipid-protein interactions of the plasma membrane. Affectations in the lipid region of the plasma membrane affect the transduction of mechanical forces, and any molecule that modifies the biophysical integrity of the lipid bilayer can alter the mechanical activity of the proteins inside the membrane. To understand whether inhibitors of mechanically activated ion channels affect the mechanical properties of the plasma membrane, we evaluated the rigidity of the membrane of sensory neurons of the DRG of mice using a variant of the scanning ion conductance microscopy method, which allows us to calculate the Young's modulus of individual cells before and after the perfusion of different doses of Gd3+, ruthenium red and GsMTx-4. Our results suggest that these molecules compromise the membrane by increasing the Young's modulus value, which indicates that the membrane becomes more rigid; these compounds act through different mechanisms and by a non-specific manner, each one shows a certain preference for specific cell subpopulations, depending on their cell size and their reactivity to isolectin B4. Our results support the idea that the biophysical properties that result from the interactions that arise in the membranes are part of the mechanotransduction process.
Collapse
Affiliation(s)
- Humberto Reyes-Pardo
- Unidad Monterrey, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Apodaca, Nuevo León, Mexico.
| | | |
Collapse
|
9
|
Ottolini M, Hong K, Sonkusare SK. Calcium signals that determine vascular resistance. WILEY INTERDISCIPLINARY REVIEWS. SYSTEMS BIOLOGY AND MEDICINE 2019; 11:e1448. [PMID: 30884210 PMCID: PMC6688910 DOI: 10.1002/wsbm.1448] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2018] [Revised: 02/07/2019] [Accepted: 02/14/2019] [Indexed: 12/19/2022]
Abstract
Small arteries in the body control vascular resistance, and therefore, blood pressure and blood flow. Endothelial and smooth muscle cells in the arterial walls respond to various stimuli by altering the vascular resistance on a moment to moment basis. Smooth muscle cells can directly influence arterial diameter by contracting or relaxing, whereas endothelial cells that line the inner walls of the arteries modulate the contractile state of surrounding smooth muscle cells. Cytosolic calcium is a key driver of endothelial and smooth muscle cell functions. Cytosolic calcium can be increased either by calcium release from intracellular stores through IP3 or ryanodine receptors, or the influx of extracellular calcium through ion channels at the cell membrane. Depending on the cell type, spatial localization, source of a calcium signal, and the calcium-sensitive target activated, a particular calcium signal can dilate or constrict the arteries. Calcium signals in the vasculature can be classified into several types based on their source, kinetics, and spatial and temporal properties. The calcium signaling mechanisms in smooth muscle and endothelial cells have been extensively studied in the native or freshly isolated cells, therefore, this review is limited to the discussions of studies in native or freshly isolated cells. This article is categorized under: Biological Mechanisms > Cell Signaling Laboratory Methods and Technologies > Imaging Models of Systems Properties and Processes > Mechanistic Models.
Collapse
Affiliation(s)
- Matteo Ottolini
- Robert M. Berne Cardiovascular Research Center, University of Virginia-School of Medicine, Charlottesville, VA, 22908, USA
- Department of Pharmacology, University of Virginia-School of Medicine, Charlottesville, VA, 22908, USA
| | - Kwangseok Hong
- Robert M. Berne Cardiovascular Research Center, University of Virginia-School of Medicine, Charlottesville, VA, 22908, USA
- Department of Physical Education, Chung-Ang University, Seoul, 06974, South Korea
| | - Swapnil K. Sonkusare
- Robert M. Berne Cardiovascular Research Center, University of Virginia-School of Medicine, Charlottesville, VA, 22908, USA
- Department of Pharmacology, University of Virginia-School of Medicine, Charlottesville, VA, 22908, USA
- Department of Molecular Physiology and Biological Physics, University of Virginia-School of Medicine, Charlottesville, VA, 22908, USA
| |
Collapse
|
10
|
Byrsonima intermedia A. Juss partitions promote gastroprotection against peptic ulcers and improve healing through antioxidant and anti-inflammatory activities. Biomed Pharmacother 2019; 111:1112-1123. [DOI: 10.1016/j.biopha.2018.12.132] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 12/21/2018] [Accepted: 12/30/2018] [Indexed: 12/16/2022] Open
|
11
|
Nakamura M, Yamanaka H, Oguro A, Imaoka S. Bisphenol A induces Nrf2-dependent drug-metabolizing enzymes through nitrosylation of Keap1. Drug Metab Pharmacokinet 2018; 33:194-202. [DOI: 10.1016/j.dmpk.2018.04.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 03/01/2018] [Accepted: 04/16/2018] [Indexed: 11/27/2022]
|
12
|
De Vriese K, Costa A, Beeckman T, Vanneste S. Pharmacological Strategies for Manipulating Plant Ca 2+ Signalling. Int J Mol Sci 2018; 19:E1506. [PMID: 29783646 PMCID: PMC5983822 DOI: 10.3390/ijms19051506] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 05/10/2018] [Accepted: 05/12/2018] [Indexed: 11/20/2022] Open
Abstract
Calcium is one of the most pleiotropic second messengers in all living organisms. However, signalling specificity is encoded via spatio-temporally regulated signatures that act with surgical precision to elicit highly specific cellular responses. How this is brought about remains a big challenge in the plant field, in part due to a lack of specific tools to manipulate/interrogate the plant Ca2+ toolkit. In many cases, researchers resort to tools that were optimized in animal cells. However, the obviously large evolutionary distance between plants and animals implies that there is a good chance observed effects may not be specific to the intended plant target. Here, we provide an overview of pharmacological strategies that are commonly used to activate or inhibit plant Ca2+ signalling. We focus on highlighting modes of action where possible, and warn for potential pitfalls. Together, this review aims at guiding plant researchers through the Ca2+ pharmacology swamp.
Collapse
Affiliation(s)
- Kjell De Vriese
- Department of Plant Biotechnology and Bioinformatics, Ghent University, Technologiepark 927, 9052 Ghent, Belgium.
- VIB Center for Plant Systems Biology, VIB, Technologiepark 927, 9052 Ghent, Belgium.
| | - Alex Costa
- Department of Biosciences, University of Milan, 20133 Milan, Italy.
- Instititute of Biophysics, Consiglio Nazionale delle Ricerche, 20133 Milan, Italy.
| | - Tom Beeckman
- Department of Plant Biotechnology and Bioinformatics, Ghent University, Technologiepark 927, 9052 Ghent, Belgium.
- VIB Center for Plant Systems Biology, VIB, Technologiepark 927, 9052 Ghent, Belgium.
| | - Steffen Vanneste
- Department of Plant Biotechnology and Bioinformatics, Ghent University, Technologiepark 927, 9052 Ghent, Belgium.
- VIB Center for Plant Systems Biology, VIB, Technologiepark 927, 9052 Ghent, Belgium.
- Lab of Plant Growth Analysis, Ghent University Global Campus, Songdomunhwa-Ro, 119, Yeonsu-gu, Incheon 21985, Korea.
| |
Collapse
|
13
|
Highly sensitive avoidance plays a key role in sensory adaptation to deep-sea hydrothermal vent environments. PLoS One 2018; 13:e0189902. [PMID: 29298328 PMCID: PMC5752015 DOI: 10.1371/journal.pone.0189902] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Accepted: 12/04/2017] [Indexed: 01/07/2023] Open
Abstract
The environments around deep-sea hydrothermal vents are very harsh conditions for organisms due to the possibility of exposure to highly toxic compounds and extremely hot venting there. Despite such extreme environments, some indigenous species have thrived there. Alvinellid worms (Annelida) are among the organisms best adapted to high-temperature and oxidatively stressful venting regions. Although intensive studies of the adaptation of these worms to the environments of hydrothermal vents have been made, little is known about the worms' sensory adaptation to the severe chemical conditions there. To examine the sensitivity of the vent-endemic worm Paralvinella hessleri to low pH and oxidative stress, we determined the concentration of acetic acid and hydrogen peroxide that induced avoidance behavior of this worm, and compared these concentrations to those obtained for related species inhabiting intertidal zones, Thelepus sp. The concentrations of the chemicals that induced avoidance behavior of P. hessleri were 10-100 times lower than those for Thelepus sp. To identify the receptors for these chemicals, chemical avoidance tests were performed with the addition of ruthenium red, a blocker of transient receptor potential (TRP) channels. This treatment suppressed the chemical avoidance behavior of P. hessleri, which suggests that TRP channels are involved in the chemical avoidance behavior of this species. Our results revealed for the first time hypersensitive detection systems for acid and for oxidative stress in the vent-endemic worm P. hessleri, possibly mediated by TRP channels, suggesting that such sensory systems may have facilitated the adaptation of this organism to harsh vent environments.
Collapse
|
14
|
Mishra J, Jhun BS, Hurst S, O-Uchi J, Csordás G, Sheu SS. The Mitochondrial Ca 2+ Uniporter: Structure, Function, and Pharmacology. Handb Exp Pharmacol 2017; 240:129-156. [PMID: 28194521 PMCID: PMC5554456 DOI: 10.1007/164_2017_1] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
Mitochondrial Ca2+ uptake is crucial for an array of cellular functions while an imbalance can elicit cell death. In this chapter, we briefly reviewed the various modes of mitochondrial Ca2+ uptake and our current understanding of mitochondrial Ca2+ homeostasis in regards to cell physiology and pathophysiology. Further, this chapter focuses on the molecular identities, intracellular regulators as well as the pharmacology of mitochondrial Ca2+ uniporter complex.
Collapse
Affiliation(s)
- Jyotsna Mishra
- Center for Translational Medicine, Department of Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, 1020 Locust Street, Suite 543D, Philadelphia, PA, 19107, USA
| | - Bong Sook Jhun
- Cardiovascular Research Center, Department of Medicine, Rhode Island Hospital, Warren Alpert Medical School of Brown University, Providence, RI, 02903, USA
| | - Stephen Hurst
- Center for Translational Medicine, Department of Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, 1020 Locust Street, Suite 543D, Philadelphia, PA, 19107, USA
| | - Jin O-Uchi
- Cardiovascular Research Center, Department of Medicine, Rhode Island Hospital, Warren Alpert Medical School of Brown University, Providence, RI, 02903, USA.
| | - György Csordás
- MitoCare Center for Mitochondrial Imaging Research and Diagnostics, Department of Pathology, Anatomy and Cell Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, 19107, USA.
| | - Shey-Shing Sheu
- Center for Translational Medicine, Department of Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, 1020 Locust Street, Suite 543D, Philadelphia, PA, 19107, USA.
| |
Collapse
|
15
|
Yang BH, Piao ZG, Kim YB, Lee CH, Lee JK, Park K, Kim JS, Oh SB. Activation of Vanilloid Receptor 1 (VR1) by Eugenol. J Dent Res 2016; 82:781-5. [PMID: 14514756 DOI: 10.1177/154405910308201004] [Citation(s) in RCA: 129] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
The structural similarity of eugenol with capsaicin suggests that these two agents may share molecular mechanisms to produce their effects. We investigated the effects of eugenol in comparison with those of capsaicin using whole-cell patch clamp and Fura-2-based calcium-imaging techniques in a heterologous expression system and with sensory neurons. In vanilloid receptor 1 (VR1)-expressing human embryonic kidney (HEK) 293 cells and trigeminal ganglion (TG) neurons, eugenol activated inward currents, whereas capsazepine, a competitive VR antagonist, and ruthenium red (RR), a functional VR antagonist, completely blocked eugenol-induced inward currents. Moreover, eugenol caused elevation of [Ca2+]i, and this was completely abolished by both capsazepine and ruthenium red in VR1-expressing HEK 293 cells and TG neurons. Our results provide strong evidence that eugenol produces its effects, at least in part, via VR1 expressed by the sensory nerve endings in the teeth.
Collapse
Affiliation(s)
- B H Yang
- Department of Physiology, College of Dentistry and Dental Research Institute, Seoul National University, 28-2 Yeongeon-Dong ChongNo-Ku, Seoul, Korea 110-749
| | | | | | | | | | | | | | | |
Collapse
|
16
|
Silva JC, Almeida JR, Quintans JS, Gopalsamy RG, Shanmugam S, Serafini MR, Oliveira MR, Silva BA, Martins AO, Castro FF, Menezes IR, Coutinho HD, Oliveira RC, Thangaraj P, Araújo AA, Quintans-Júnior LJ. Enhancement of orofacial antinociceptive effect of carvacrol, a monoterpene present in oregano and thyme oils, by β-cyclodextrin inclusion complex in mice. Biomed Pharmacother 2016; 84:454-461. [DOI: 10.1016/j.biopha.2016.09.065] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Revised: 09/16/2016] [Accepted: 09/17/2016] [Indexed: 12/21/2022] Open
|
17
|
Mickle AD, Shepherd AJ, Mohapatra DP. Nociceptive TRP Channels: Sensory Detectors and Transducers in Multiple Pain Pathologies. Pharmaceuticals (Basel) 2016; 9:ph9040072. [PMID: 27854251 PMCID: PMC5198047 DOI: 10.3390/ph9040072] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Revised: 11/07/2016] [Accepted: 11/09/2016] [Indexed: 02/07/2023] Open
Abstract
Specialized receptors belonging to the transient receptor potential (TRP) family of ligand-gated ion channels constitute the critical detectors and transducers of pain-causing stimuli. Nociceptive TRP channels are predominantly expressed by distinct subsets of sensory neurons of the peripheral nervous system. Several of these TRP channels are also expressed in neurons of the central nervous system, and in non-neuronal cells that communicate with sensory nerves. Nociceptive TRPs are activated by specific physico-chemical stimuli to provide the excitatory trigger in neurons. In addition, decades of research has identified a large number of immune and neuromodulators as mediators of nociceptive TRP channel activation during injury, inflammatory and other pathological conditions. These findings have led to aggressive targeting of TRP channels for the development of new-generation analgesics. This review summarizes the complex activation and/or modulation of nociceptive TRP channels under pathophysiological conditions, and how these changes underlie acute and chronic pain conditions. Furthermore, development of small-molecule antagonists for several TRP channels as analgesics, and the positive and negative outcomes of these drugs in clinical trials are discussed. Understanding the diverse functional and modulatory properties of nociceptive TRP channels is critical to function-based drug targeting for the development of evidence-based and efficacious new generation analgesics.
Collapse
Affiliation(s)
- Aaron D Mickle
- Department of Anesthesiology, Washington University School of Medicine, 660 S. Euclid Avenue, St. Louis, MO 63110, USA.
- Washington University Pain Center, Department of Anesthesiology, Washington University School of Medicine, 660 S. Euclid Avenue, St. Louis, MO 63110, USA.
| | - Andrew J Shepherd
- Department of Anesthesiology, Washington University School of Medicine, 660 S. Euclid Avenue, St. Louis, MO 63110, USA.
- Washington University Pain Center, Department of Anesthesiology, Washington University School of Medicine, 660 S. Euclid Avenue, St. Louis, MO 63110, USA.
| | - Durga P Mohapatra
- Department of Anesthesiology, Washington University School of Medicine, 660 S. Euclid Avenue, St. Louis, MO 63110, USA.
- Washington University Pain Center, Department of Anesthesiology, Washington University School of Medicine, 660 S. Euclid Avenue, St. Louis, MO 63110, USA.
- Center for Investigation of Membrane Excitability Diseases, Washington University School of Medicine, 660 S. Euclid Avenue, St. Louis, MO 63110, USA.
- Siteman Cancer Center, Washington University School of Medicine, 660 S. Euclid Avenue, St. Louis, MO 63110, USA.
| |
Collapse
|
18
|
Tsitoura P, Koussis K, Iatrou K. Inhibition of Anopheles gambiae odorant receptor function by mosquito repellents. J Biol Chem 2015; 290:7961-72. [PMID: 25657000 PMCID: PMC4367294 DOI: 10.1074/jbc.m114.632299] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Revised: 01/28/2015] [Indexed: 11/06/2022] Open
Abstract
The identification of molecular targets of insect repellents has been a challenging task, with their effects on odorant receptors (ORs) remaining a debatable issue. Here, we describe a study on the effects of selected mosquito repellents, including the widely used repellent N,N-diethyl-meta-toluamide (DEET), on the function of specific ORs of the African malaria vector Anopheles gambiae. This study, which has been based on quantitative measurements of a Ca(2+)-activated photoprotein biosensor of recombinant OR function in an insect cell-based expression platform and a sequential compound addition protocol, revealed that heteromeric OR (ORx/Orco) function was susceptible to strong inhibition by all tested mosquito repellents except DEET. Moreover, our results demonstrated that the observed inhibition was due to efficient blocking of Orco (olfactory receptor coreceptor) function. This mechanism of repellent action, which is reported for the first time, is distinct from the mode of action of other characterized insect repellents including DEET.
Collapse
Affiliation(s)
- Panagiota Tsitoura
- From the Insect Molecular Genetics and Biotechnology Group, Institute of Biosciences and Applications, National Centre for Scientific Research "Demokritos", 15310 Athens and
| | - Konstantinos Koussis
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, 70013 Heraklion, Crete, Greece
| | - Kostas Iatrou
- From the Insect Molecular Genetics and Biotechnology Group, Institute of Biosciences and Applications, National Centre for Scientific Research "Demokritos", 15310 Athens and
| |
Collapse
|
19
|
Min JW, Liu WH, He XH, Peng BW. Different types of toxins targeting TRPV1 in pain. Toxicon 2013; 71:66-75. [PMID: 23732125 DOI: 10.1016/j.toxicon.2013.05.016] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2012] [Revised: 05/14/2013] [Accepted: 05/22/2013] [Indexed: 02/06/2023]
Abstract
The transient receptor potential vanilloid 1(TRPV1) channels are members of the transient receptor potential (TRP) superfamily. Members of this family are expressed in primary sensory neurons and are best known for their role in nociception and sensory transmission. Multiple painful stimuli can activate these channels. In this review, we discussed the mechanisms of different types of venoms that target TRPV1, such as scorpion venom, botulinum neurotoxin, spider toxin, ciguatera fish poisoning (CFP) and neurotoxic shellfish poisoning (NSP). Some of these toxins activate TRPV1; however, some do not. Regardless of TRPV1 inhibition or activation, they occur through different pathways. For example, BoNT/A decreases TRPV1 expression levels by blocking TRPV1 trafficking to the plasma membrane, although the exact mechanism is still under debate. Vanillotoxins from tarantula (Psalmopoeus cambridgei) are proposed to activate TRPV1 via interaction with a region of TRPV1 that is homologous to voltage-dependent ion channels. Here, we offer a description of the present state of knowledge for this complex subject.
Collapse
Affiliation(s)
- Jia-Wei Min
- Department of Physiology, School of Basic Medical Sciences, Wuhan University, 185 Donghu Road, Wuhan, Hubei 430071, PR China
| | | | | | | |
Collapse
|
20
|
Jin H, Sun P, Takatori S, Koyama T, Zamami Y, Tangsucharit P, Kitamura Y, Kawasaki H. Involvement of perivascular nerves and transient receptor potential vanilloid 1 (TRPV1) in vascular responses to histamine in rat mesenteric resistance arteries. Eur J Pharmacol 2012; 680:73-80. [DOI: 10.1016/j.ejphar.2012.01.018] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2011] [Revised: 01/16/2012] [Accepted: 01/23/2012] [Indexed: 10/14/2022]
|
21
|
Borbíró I, Lisztes E, Tóth BI, Czifra G, Oláh A, Szöllősi AG, Szentandrássy N, Nánási PP, Péter Z, Paus R, Kovács L, Bíró T. Activation of Transient Receptor Potential Vanilloid-3 Inhibits Human Hair Growth. J Invest Dermatol 2011; 131:1605-14. [DOI: 10.1038/jid.2011.122] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
|
22
|
Tokita Y, Yamamoto M, Satoh K, Nishiyama M, Iizuka S, Imamura S, Kase Y. Possible involvement of the transient receptor potential vanilloid type 1 channel in postoperative adhesive obstruction and its prevention by a kampo (traditional Japanese) medicine, daikenchuto. J Pharmacol Sci 2010; 115:75-83. [PMID: 21186335 DOI: 10.1254/jphs.10226fp] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
This study focused on the localization of transient receptor potential vanilloid type 1 (TRPV1) in the intestines in postoperative adhesion model rats and investigated the underlying mechanism for the anti-adhesion action of daikenchuto (DKT), especially in relation to TRPV1. Postoperative intestinal adhesion was induced by sprinkling talc in the small intestine. The expression of TRPV1 mRNA was examined by in situ hybridization and real-time RT-PCR. The effects of DKT and its major ingredient, hydroxy sanshool, with or without ruthenium red, a TRP-channel antagonist, on talc-induced intestinal adhesions were evaluated. The level of TRPV1 mRNA was higher in the adhesion regions of talc-treated rats than in normal small intestine of sham-operated rats. Localization of TRPV1 mRNA expression was identified in the submucosal plexus of both sham-operated and talc-treated rats; and in talc-treated rats, it was observed also in the myenteric plexus and regions of adhesion. Capsaicin, DKT, and hydroxy sanshool significantly prevented formation of intestinal adhesions. The effects of DKT and hydroxy sanshool were abrogated by subcutaneous injection of ruthenium red. These results suggest that pharmacological modulation of TRPV1 might be a possible therapeutic option in postoperative intestinal adhesion, which might be relevant to the prevention of postoperative adhesive obstruction by DKT.
Collapse
|
23
|
St. Pierre M, Reeh PW, Zimmermann K. Differential effects of TRPV channel block on polymodal activation of rat cutaneous nociceptors in vitro. Exp Brain Res 2009; 196:31-44. [DOI: 10.1007/s00221-009-1808-3] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2009] [Accepted: 04/07/2009] [Indexed: 02/02/2023]
|
24
|
Brown RC, Wu L, Hicks K, O'neil RG. Regulation of blood-brain barrier permeability by transient receptor potential type C and type v calcium-permeable channels. Microcirculation 2008; 15:359-71. [PMID: 18464164 DOI: 10.1080/10739680701762656] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
OBJECTIVE To identify plasma membrane ion channels mediating calcium influx at the blood-brain barrier in response to disrupting stimuli. METHODS We examined the expression and function of candidate transient receptor potential channels using reverse transcriptase polymerase chain recation, Fura-2 calcium imaging, and permeability assays. RESULTS Immortalized mouse brain microvessel endothelial cells expressed multiple transient receptor potential isoforms: transient receptor potential C1, C2, C4, and C7, M2, M3, M4, and M7, and V2 and V4. Similar profiles were observed in freshly isolated cerebral microvessels and primary cultured rat brain endothelial cells. Thrombin-stimulated calcium influx in brain endothelial cells was blocked by transient receptor potential C inhibitors. Transient receptor potential V activating stimuli also increased intracellular calcium. This increase was inhibited by a transient receptor potential V blocker or by removal of extracellular calcium. Barrier integrity was compromised by thrombin, hypo-osmolar stress, and PMA treatment. The increase in barrier permeability induced by transient receptor potential V activators was blocked by transient receptor potential V inhibition, while thrombin effects were inhibited by transient receptor potential C inhibitors. CONCLUSIONS These results demonstrate that transient receptor potential C and transient receptor potential V channels mediate calcium influx at the blood-brain barrier, and as a consequence, may modulate barrier integrity.
Collapse
Affiliation(s)
- Rachel C Brown
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | | | | | | |
Collapse
|
25
|
Andrè E, Campi B, Materazzi S, Trevisani M, Amadesi S, Massi D, Creminon C, Vaksman N, Nassini R, Civelli M, Baraldi PG, Poole DP, Bunnett NW, Geppetti P, Patacchini R. Cigarette smoke-induced neurogenic inflammation is mediated by alpha,beta-unsaturated aldehydes and the TRPA1 receptor in rodents. J Clin Invest 2008; 118:2574-82. [PMID: 18568077 PMCID: PMC2430498 DOI: 10.1172/jci34886] [Citation(s) in RCA: 199] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2007] [Accepted: 04/23/2008] [Indexed: 01/21/2023] Open
Abstract
Cigarette smoke (CS) inhalation causes an early inflammatory response in rodent airways by stimulating capsaicin-sensitive sensory neurons that express transient receptor potential cation channel, subfamily V, member 1 (TRPV1) through an unknown mechanism that does not involve TRPV1. We hypothesized that 2 alpha,beta-unsaturated aldehydes present in CS, crotonaldehyde and acrolein, induce neurogenic inflammation by stimulating TRPA1, an excitatory ion channel coexpressed with TRPV1 on capsaicin-sensitive nociceptors. We found that CS aqueous extract (CSE), crotonaldehyde, and acrolein mobilized Ca2+ in cultured guinea pig jugular ganglia neurons and promoted contraction of isolated guinea pig bronchi. These responses were abolished by a TRPA1-selective antagonist and by the aldehyde scavenger glutathione but not by the TRPV1 antagonist capsazepine or by ROS scavengers. Treatment with CSE or aldehydes increased Ca2+ influx in TRPA1-transfected cells, but not in control HEK293 cells, and promoted neuropeptide release from isolated guinea pig airway tissue. Furthermore, the effect of CSE and aldehydes on Ca2+ influx in dorsal root ganglion neurons was abolished in TRPA1-deficient mice. These data identify alpha,beta-unsaturated aldehydes as the main causative agents in CS that via TRPA1 stimulation mediate airway neurogenic inflammation and suggest a role for TRPA1 in the pathogenesis of CS-induced diseases.
Collapse
Affiliation(s)
- Eunice Andrè
- Center of Excellence for the Study of Inflammation, University of Ferrara, Ferrara, Italy.
Clinical Pharmacology Unit, Department of Critical Care Medicine and Surgery, University of Florence, Florence, Italy.
Departments of Physiology and Surgery, UCSF, San Francisco, California, USA.
Department of Human Pathology and Oncology, University of Florence, Florence, Italy.
CEA, Institut de Biologie et Technologies de Saclay (iBiTec-S), Service de pharmacologie et d'immuno analyse (SPI), Gif sur Yvette, France.
Pharmacology Department, Chiesi Pharmaceuticals, Parma, Italy.
Department of Pharmaceutical Chemistry, University of Ferrara, Ferrara, Italy
| | - Barbara Campi
- Center of Excellence for the Study of Inflammation, University of Ferrara, Ferrara, Italy.
Clinical Pharmacology Unit, Department of Critical Care Medicine and Surgery, University of Florence, Florence, Italy.
Departments of Physiology and Surgery, UCSF, San Francisco, California, USA.
Department of Human Pathology and Oncology, University of Florence, Florence, Italy.
CEA, Institut de Biologie et Technologies de Saclay (iBiTec-S), Service de pharmacologie et d'immuno analyse (SPI), Gif sur Yvette, France.
Pharmacology Department, Chiesi Pharmaceuticals, Parma, Italy.
Department of Pharmaceutical Chemistry, University of Ferrara, Ferrara, Italy
| | - Serena Materazzi
- Center of Excellence for the Study of Inflammation, University of Ferrara, Ferrara, Italy.
Clinical Pharmacology Unit, Department of Critical Care Medicine and Surgery, University of Florence, Florence, Italy.
Departments of Physiology and Surgery, UCSF, San Francisco, California, USA.
Department of Human Pathology and Oncology, University of Florence, Florence, Italy.
CEA, Institut de Biologie et Technologies de Saclay (iBiTec-S), Service de pharmacologie et d'immuno analyse (SPI), Gif sur Yvette, France.
Pharmacology Department, Chiesi Pharmaceuticals, Parma, Italy.
Department of Pharmaceutical Chemistry, University of Ferrara, Ferrara, Italy
| | - Marcello Trevisani
- Center of Excellence for the Study of Inflammation, University of Ferrara, Ferrara, Italy.
Clinical Pharmacology Unit, Department of Critical Care Medicine and Surgery, University of Florence, Florence, Italy.
Departments of Physiology and Surgery, UCSF, San Francisco, California, USA.
Department of Human Pathology and Oncology, University of Florence, Florence, Italy.
CEA, Institut de Biologie et Technologies de Saclay (iBiTec-S), Service de pharmacologie et d'immuno analyse (SPI), Gif sur Yvette, France.
Pharmacology Department, Chiesi Pharmaceuticals, Parma, Italy.
Department of Pharmaceutical Chemistry, University of Ferrara, Ferrara, Italy
| | - Silvia Amadesi
- Center of Excellence for the Study of Inflammation, University of Ferrara, Ferrara, Italy.
Clinical Pharmacology Unit, Department of Critical Care Medicine and Surgery, University of Florence, Florence, Italy.
Departments of Physiology and Surgery, UCSF, San Francisco, California, USA.
Department of Human Pathology and Oncology, University of Florence, Florence, Italy.
CEA, Institut de Biologie et Technologies de Saclay (iBiTec-S), Service de pharmacologie et d'immuno analyse (SPI), Gif sur Yvette, France.
Pharmacology Department, Chiesi Pharmaceuticals, Parma, Italy.
Department of Pharmaceutical Chemistry, University of Ferrara, Ferrara, Italy
| | - Daniela Massi
- Center of Excellence for the Study of Inflammation, University of Ferrara, Ferrara, Italy.
Clinical Pharmacology Unit, Department of Critical Care Medicine and Surgery, University of Florence, Florence, Italy.
Departments of Physiology and Surgery, UCSF, San Francisco, California, USA.
Department of Human Pathology and Oncology, University of Florence, Florence, Italy.
CEA, Institut de Biologie et Technologies de Saclay (iBiTec-S), Service de pharmacologie et d'immuno analyse (SPI), Gif sur Yvette, France.
Pharmacology Department, Chiesi Pharmaceuticals, Parma, Italy.
Department of Pharmaceutical Chemistry, University of Ferrara, Ferrara, Italy
| | - Christophe Creminon
- Center of Excellence for the Study of Inflammation, University of Ferrara, Ferrara, Italy.
Clinical Pharmacology Unit, Department of Critical Care Medicine and Surgery, University of Florence, Florence, Italy.
Departments of Physiology and Surgery, UCSF, San Francisco, California, USA.
Department of Human Pathology and Oncology, University of Florence, Florence, Italy.
CEA, Institut de Biologie et Technologies de Saclay (iBiTec-S), Service de pharmacologie et d'immuno analyse (SPI), Gif sur Yvette, France.
Pharmacology Department, Chiesi Pharmaceuticals, Parma, Italy.
Department of Pharmaceutical Chemistry, University of Ferrara, Ferrara, Italy
| | - Natalya Vaksman
- Center of Excellence for the Study of Inflammation, University of Ferrara, Ferrara, Italy.
Clinical Pharmacology Unit, Department of Critical Care Medicine and Surgery, University of Florence, Florence, Italy.
Departments of Physiology and Surgery, UCSF, San Francisco, California, USA.
Department of Human Pathology and Oncology, University of Florence, Florence, Italy.
CEA, Institut de Biologie et Technologies de Saclay (iBiTec-S), Service de pharmacologie et d'immuno analyse (SPI), Gif sur Yvette, France.
Pharmacology Department, Chiesi Pharmaceuticals, Parma, Italy.
Department of Pharmaceutical Chemistry, University of Ferrara, Ferrara, Italy
| | - Romina Nassini
- Center of Excellence for the Study of Inflammation, University of Ferrara, Ferrara, Italy.
Clinical Pharmacology Unit, Department of Critical Care Medicine and Surgery, University of Florence, Florence, Italy.
Departments of Physiology and Surgery, UCSF, San Francisco, California, USA.
Department of Human Pathology and Oncology, University of Florence, Florence, Italy.
CEA, Institut de Biologie et Technologies de Saclay (iBiTec-S), Service de pharmacologie et d'immuno analyse (SPI), Gif sur Yvette, France.
Pharmacology Department, Chiesi Pharmaceuticals, Parma, Italy.
Department of Pharmaceutical Chemistry, University of Ferrara, Ferrara, Italy
| | - Maurizio Civelli
- Center of Excellence for the Study of Inflammation, University of Ferrara, Ferrara, Italy.
Clinical Pharmacology Unit, Department of Critical Care Medicine and Surgery, University of Florence, Florence, Italy.
Departments of Physiology and Surgery, UCSF, San Francisco, California, USA.
Department of Human Pathology and Oncology, University of Florence, Florence, Italy.
CEA, Institut de Biologie et Technologies de Saclay (iBiTec-S), Service de pharmacologie et d'immuno analyse (SPI), Gif sur Yvette, France.
Pharmacology Department, Chiesi Pharmaceuticals, Parma, Italy.
Department of Pharmaceutical Chemistry, University of Ferrara, Ferrara, Italy
| | - Pier Giovanni Baraldi
- Center of Excellence for the Study of Inflammation, University of Ferrara, Ferrara, Italy.
Clinical Pharmacology Unit, Department of Critical Care Medicine and Surgery, University of Florence, Florence, Italy.
Departments of Physiology and Surgery, UCSF, San Francisco, California, USA.
Department of Human Pathology and Oncology, University of Florence, Florence, Italy.
CEA, Institut de Biologie et Technologies de Saclay (iBiTec-S), Service de pharmacologie et d'immuno analyse (SPI), Gif sur Yvette, France.
Pharmacology Department, Chiesi Pharmaceuticals, Parma, Italy.
Department of Pharmaceutical Chemistry, University of Ferrara, Ferrara, Italy
| | - Daniel P. Poole
- Center of Excellence for the Study of Inflammation, University of Ferrara, Ferrara, Italy.
Clinical Pharmacology Unit, Department of Critical Care Medicine and Surgery, University of Florence, Florence, Italy.
Departments of Physiology and Surgery, UCSF, San Francisco, California, USA.
Department of Human Pathology and Oncology, University of Florence, Florence, Italy.
CEA, Institut de Biologie et Technologies de Saclay (iBiTec-S), Service de pharmacologie et d'immuno analyse (SPI), Gif sur Yvette, France.
Pharmacology Department, Chiesi Pharmaceuticals, Parma, Italy.
Department of Pharmaceutical Chemistry, University of Ferrara, Ferrara, Italy
| | - Nigel W. Bunnett
- Center of Excellence for the Study of Inflammation, University of Ferrara, Ferrara, Italy.
Clinical Pharmacology Unit, Department of Critical Care Medicine and Surgery, University of Florence, Florence, Italy.
Departments of Physiology and Surgery, UCSF, San Francisco, California, USA.
Department of Human Pathology and Oncology, University of Florence, Florence, Italy.
CEA, Institut de Biologie et Technologies de Saclay (iBiTec-S), Service de pharmacologie et d'immuno analyse (SPI), Gif sur Yvette, France.
Pharmacology Department, Chiesi Pharmaceuticals, Parma, Italy.
Department of Pharmaceutical Chemistry, University of Ferrara, Ferrara, Italy
| | - Pierangelo Geppetti
- Center of Excellence for the Study of Inflammation, University of Ferrara, Ferrara, Italy.
Clinical Pharmacology Unit, Department of Critical Care Medicine and Surgery, University of Florence, Florence, Italy.
Departments of Physiology and Surgery, UCSF, San Francisco, California, USA.
Department of Human Pathology and Oncology, University of Florence, Florence, Italy.
CEA, Institut de Biologie et Technologies de Saclay (iBiTec-S), Service de pharmacologie et d'immuno analyse (SPI), Gif sur Yvette, France.
Pharmacology Department, Chiesi Pharmaceuticals, Parma, Italy.
Department of Pharmaceutical Chemistry, University of Ferrara, Ferrara, Italy
| | - Riccardo Patacchini
- Center of Excellence for the Study of Inflammation, University of Ferrara, Ferrara, Italy.
Clinical Pharmacology Unit, Department of Critical Care Medicine and Surgery, University of Florence, Florence, Italy.
Departments of Physiology and Surgery, UCSF, San Francisco, California, USA.
Department of Human Pathology and Oncology, University of Florence, Florence, Italy.
CEA, Institut de Biologie et Technologies de Saclay (iBiTec-S), Service de pharmacologie et d'immuno analyse (SPI), Gif sur Yvette, France.
Pharmacology Department, Chiesi Pharmaceuticals, Parma, Italy.
Department of Pharmaceutical Chemistry, University of Ferrara, Ferrara, Italy
| |
Collapse
|
26
|
Matsuda H, Ochi M, Nagatomo A, Yoshikawa M. Effects of allyl isothiocyanate from horseradish on several experimental gastric lesions in rats. Eur J Pharmacol 2007; 561:172-81. [PMID: 17346695 DOI: 10.1016/j.ejphar.2006.12.040] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2006] [Revised: 12/05/2006] [Accepted: 12/18/2006] [Indexed: 11/24/2022]
Abstract
Allyl isothiocyanate is well known to be a principal pungent constituent of horseradish and an agonist for transient receptor potential (TRP) A1. Ally isothiocyanate markedly inhibited the formation of gastric lesions induced by ethanol (1.5 ml/rat, p.o.), 0.6 M HCl (1.5 ml/rat, p.o.), 1% ammonia (1.5 ml/rat, p.o.), and aspirin (150 mg/kg, p.o.) (ED(50)=1.6, 2.2, 1.7, ca. 6.5 mg/kg, p.o.). It also significantly inhibited the formation of gastric lesions induced by indomethacin (20 mg/kg, p.o.), though the inhibition was ca. 60% at a high dose (40 mg/kg, p.o.). Furthermore, several synthetic isothiocyanate compounds also significantly inhibited ethanol and indomethacin-induced gastric lesions. Whereas, TRPV1 agonists, capsaicin and piperine, inhibited gastric lesions induced by ethanol, 1% ammonia, and aspirin, but had less of an effect on 0.6 M HCl-induced gastric lesions. With regard to mode of action, the protective effects of ally isothiocyanate on ethanol-induced gastric lesions were attenuated by pretreatment with indomethacin, but not with N(G)-nitro-L-arginine methyl ester hydrochloride (L-NAME), or ruthenium red. Pretreatment with indomethacin reduced the protective effects of piperine, and L-NAME reduced the effects of capsaicin and omeprazole. Furthermore, ruthenium red reduced the effects of capsaicin, piperine, and omeprazole. These findings suggest that endogenous prostaglandins play an important role in the protective effect of allyl isothiocyanate in ethanol-induced gastric lesions different from capsaicin, piperine, and omeprazole.
Collapse
Affiliation(s)
- Hisashi Matsuda
- Department of Pharmacognosy, Kyoto Pharmaceutical University, Misasagi, Yamashina-ku, Kyoto 607-8412, Japan
| | | | | | | |
Collapse
|
27
|
De Petrocellis L, Starowicz K, Moriello AS, Vivese M, Orlando P, Di Marzo V. Regulation of transient receptor potential channels of melastatin type 8 (TRPM8): effect of cAMP, cannabinoid CB(1) receptors and endovanilloids. Exp Cell Res 2007; 313:1911-20. [PMID: 17428469 DOI: 10.1016/j.yexcr.2007.01.008] [Citation(s) in RCA: 111] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2006] [Revised: 01/09/2007] [Accepted: 01/10/2007] [Indexed: 11/17/2022]
Abstract
The transient receptor potential channel of melastatin type 8 (TRPM8), which is gated by low (<25 degrees C) temperature and chemical compounds, is regulated by protein kinase C-mediated phosphorylation in a way opposite to that observed with the transient receptor potential channel of vanilloid type 1 (TRPV1), i.e. by being desensitized and not sensitized. As TRPV1 is sensitized also by protein kinase A (PKA)-mediated phosphorylation, we investigated the effect of two activators of the PKA pathway, 8-Br-cAMP and forskolin, on the activity of menthol and icilin at TRPM8 in HEK-293 cells stably overexpressing the channel (TRPM8-HEK-293 cells). We also studied the effect on TRPM8 of: (1) a series of compounds previously shown to activate or antagonize TRPV1, and (2) co-stimulation of transiently co-expressed cannabinoid CB(1) receptors. Both 8-Br-cAMP (100 microM) and forskolin (10 microM) right-shifted the dose-response curves for the TRPM8-mediated effect of icilin and menthol on intracellular Ca(2+). The inhibitory effects of 8-Br-cAMP and forskolin were attenuated by the selective PKA inhibitor Rp-cAMP-S. Stimulation of human CB(1) receptors transiently co-expressed in TRPM8-HEK-293 cells also inhibited TRPM8 response to icilin. Finally, some TRPV1 agonists and antagonists, but not iodinated antagonists, antagonized icilin- and much less so menthol-, induced TRPM8 activation. Importantly, the endovanilloids/endocannabinoids, anandamide and NADA, also antagonized TRPM8 at submicromolar concentrations. Although these findings need to be confirmed by experiments directly measuring TRPM8 activity in natively TRPM8-expressing cells, they support the notion that the same regulatory events have opposing actions on TRPM8 and TRPV1 receptors and identify anandamide and NADA as the first potential endogenous functional antagonists of TRPM8 channels.
Collapse
Affiliation(s)
- Luciano De Petrocellis
- Endocannabinoid Research Group, Institute of Cybernetics, National Research Council, Via Campi Flegrei 34, Comprensorio Olivetti, 80078 Pozzuoli, Naples, Italy
| | | | | | | | | | | |
Collapse
|
28
|
Tugnoli V, Capone JG, Eleopra R, Quatrale R, Sensi M, Gastaldo E, Tola MR, Geppetti P. Botulinum toxin type A reduces capsaicin-evoked pain and neurogenic vasodilatation in human skin. Pain 2006; 130:76-83. [PMID: 17194546 DOI: 10.1016/j.pain.2006.10.030] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2006] [Revised: 10/19/2006] [Accepted: 10/30/2006] [Indexed: 10/23/2022]
Abstract
The effect of Botulinum Toxin type A (BoNT/A) on pain and neurogenic vasodilatation induced by application to the human skin of thermal stimuli and capsaicin was evaluated in a double blind study. A capsaicin cream (0.5 ml of a 0.075%) was applied to the skin of both forearms of eighteen subjects randomly pretreated with either BoNT/A (Botox) or 0.9% saline (NS). Capsaicin was applied to a skin area either inside (protocol A) or adjacent to the BoNT/A treated area (protocol B). Pre-treatment with BoNT/A did not affect thermal-specific and thermal-pain thresholds (by quantitative sensory testing). However, capsaicin-induced pain sensation (by a visual analogue scale), flare area (by acetate sheet) and changes in cutaneous blood flow (CBF, by laser Doppler flowmetry) were reduced when capsaicin was administered inside (protocol A) the BoNT/A treated area. In Protocol B, capsaicin-induced pain was unchanged, and capsaicin-induced flare/increase in CBF were reduced only in the area treated with BoNT/A, but not in the BoNT/A untreated area. Results indicate that (i) BoNT/A reduces capsaicin-induced pain and neurogenic vasodilatation without affecting the transmission of thermal and thermal-pain modalities; (ii) reduction in capsaicin-induced pain occurs only if capsaicin is administered into the BoNT/A pretreated area; (iii) reduction in neurogenic vasodilatation by BoNT/A does not contribute to its analgesic action. BoNT/A could be tested for the treatment of conditions characterised by neurogenic inflammation and inflammatory pain.
Collapse
Affiliation(s)
- Valeria Tugnoli
- Department of Clinical Neuroscience, S.Anna University Hospital of Ferrara, Ferrara, Italy.
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Hajnóczky G, Csordás G, Das S, Garcia-Perez C, Saotome M, Sinha Roy S, Yi M. Mitochondrial calcium signalling and cell death: approaches for assessing the role of mitochondrial Ca2+ uptake in apoptosis. Cell Calcium 2006; 40:553-60. [PMID: 17074387 PMCID: PMC2692319 DOI: 10.1016/j.ceca.2006.08.016] [Citation(s) in RCA: 460] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2006] [Accepted: 08/23/2006] [Indexed: 12/30/2022]
Abstract
Local Ca(2+) transfer between adjoining domains of the sarcoendoplasmic reticulum (ER/SR) and mitochondria allows ER/SR Ca(2+) release to activate mitochondrial Ca(2+) uptake and to evoke a matrix [Ca(2+)] ([Ca(2+)](m)) rise. [Ca(2+)](m) exerts control on several steps of energy metabolism to synchronize ATP generation with cell function. However, calcium signal propagation to the mitochondria may also ignite a cell death program through opening of the permeability transition pore (PTP). This occurs when the Ca(2+) release from the ER/SR is enhanced or is coincident with sensitization of the PTP. Recent studies have shown that several pro-apoptotic factors, including members of the Bcl-2 family proteins and reactive oxygen species (ROS) regulate the Ca(2+) sensitivity of both the Ca(2+) release channels in the ER and the PTP in the mitochondria. To test the relevance of the mitochondrial Ca(2+) accumulation in various apoptotic paradigms, methods are available for buffering of [Ca(2+)], for dissipation of the driving force of the mitochondrial Ca(2+) uptake and for inhibition of the mitochondrial Ca(2+) transport mechanisms. However, in intact cells, the efficacy and the specificity of these approaches have to be established. Here we discuss mechanisms that recruit the mitochondrial calcium signal to a pro-apoptotic cascade and the approaches available for assessment of the relevance of the mitochondrial Ca(2+) handling in apoptosis. We also present a systematic evaluation of the effect of ruthenium red and Ru360, two inhibitors of mitochondrial Ca(2+) uptake on cytosolic [Ca(2+)] and [Ca(2+)](m) in intact cultured cells.
Collapse
Affiliation(s)
- György Hajnóczky
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA.
| | | | | | | | | | | | | |
Collapse
|
30
|
Abstract
In recent years, infection of the stomach with the organism Helicobacter Pylori has been found to be the main cause of gastric ulcers, one of the common ailments afflicting humans. Excessive acid secretion in the stomach, reduction in gastric mucosal blood flow, constant intake of non-steroid anti-inflammatory drugs (NSAIDS), ethanol, smoking, stress etc. are also considered responsible for ulcer formation. The prevalent notion among sections of population in this country and perhaps in others is that "red pepper" popularly known as "Chilli," a common spice consumed in excessive amounts leads to "gastric ulcers" in view of its irritant and likely acid secreting nature. Persons with ulcers are advised either to limit or avoid its use. However, investigations carried out in recent years have revealed that chilli or its active principle "capsaicin" is not the cause for ulcer formation but a "benefactor." Capsaicin does not stimulate but inhibits acid secretion, stimulates alkali, mucus secretions and particularly gastric mucosal blood flow which help in prevention and healing of ulcers. Capsaicin acts by stimulating afferent neurons in the stomach and signals for protection against injury causing agents. Epidemiologic surveys in Singapore have shown that gastric ulcers are three times more common in the "Chinese" than among Malaysians and Indians who are in the habit of consuming more chillis. Ulcers are common among people who are in the habit of taking NSAIDS and are infected with the organism "Helicobacter Pylori," responsible for excessive acid secretion and erosion of the mucosal layer. Eradication of the bacteria by antibiotic treatment and avoiding the NSAIDS eliminates ulcers and restores normal acid secretion.
Collapse
|
31
|
Trevisani M, Patacchini R, Nicoletti P, Gatti R, Gazzieri D, Lissi N, Zagli G, Creminon C, Geppetti P, Harrison S. Hydrogen sulfide causes vanilloid receptor 1-mediated neurogenic inflammation in the airways. Br J Pharmacol 2005; 145:1123-31. [PMID: 15937520 PMCID: PMC1576227 DOI: 10.1038/sj.bjp.0706277] [Citation(s) in RCA: 118] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Hydrogen sulfide (H(2)S) is described as a mediator of diverse biological effects, and is known to produce irritation and injury in the lung following inhalation. Recently, H(2)S has been found to cause contraction in the rat urinary bladder via a neurogenic mechanism. Here, we studied whether sodium hydrogen sulfide (NaHS), used as donor of H(2)S, produces responses mediated by sensory nerve activation in the guinea-pig airways. NaHS evoked an increase in neuropeptide release in the airways that was significantly attenuated by capsaicin desensitization and by the transient receptor potential vanilloid 1 (TRPV1) antagonist capsazepine. In addition, NaHS caused an atropine-resistant contraction of isolated airways, which was completely prevented by capsaicin desensitization. Furthermore, NaHS-induced contraction was reduced by TRPV1 antagonism (ruthenium red, capsazepine and SB366791), and was abolished by pretreatment with the combination of tachykinin NK(1) (SR140333) and NK(2) (SR48968) receptor antagonists. In anesthetized guinea-pigs, intratracheal instillation of NaHS increased the total lung resistance and airway plasma protein extravasation. These two effects were reduced by TRPV1 antagonism (capsazepine) and tachykinin receptors (SR140333 and SR48968) blockade. Our results provide the first pharmacological evidence that H(2)S provokes tachykinin-mediated neurogenic inflammatory responses in guinea-pig airways, and that this effect is mediated by stimulation of TRPV1 receptors on sensory nerves endings. This novel mechanism may contribute to the irritative action of H(2)S in the respiratory system.
Collapse
Affiliation(s)
- Marcello Trevisani
- Center of Excellence for the Study of Inflammation, University of Ferrara, Ferrara, Italy
| | | | - Paola Nicoletti
- Department of Critical Care Medicine & Surgery, University of Florence, Florence, Italy
| | - Raffaele Gatti
- Center of Excellence for the Study of Inflammation, University of Ferrara, Ferrara, Italy
| | - David Gazzieri
- Center of Excellence for the Study of Inflammation, University of Ferrara, Ferrara, Italy
| | - Nicola Lissi
- Center of Excellence for the Study of Inflammation, University of Ferrara, Ferrara, Italy
| | - Giovanni Zagli
- Department of Critical Care Medicine & Surgery, University of Florence, Florence, Italy
| | - Christophe Creminon
- CEA, Service de Pharmacologie et d'Immunologie, DRM, CEA-Saclay, Gif sur Yvette, France
| | - Pierangelo Geppetti
- Center of Excellence for the Study of Inflammation, University of Ferrara, Ferrara, Italy
- Department of Critical Care Medicine & Surgery, University of Florence, Florence, Italy
| | - Selena Harrison
- Department of Critical Care Medicine & Surgery, University of Florence, Florence, Italy
- Author for correspondence:
| |
Collapse
|
32
|
Kwolek G, Zakrzeska A, Schlicker E, Göthert M, Godlewski G, Malinowska B. Central and peripheral components of the pressor effect of anandamide in urethane-anaesthetized rats. Br J Pharmacol 2005; 145:567-75. [PMID: 15834445 PMCID: PMC1576172 DOI: 10.1038/sj.bjp.0706195] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
1. We wanted to search for the mechanism(s) responsible for the brief pressor response induced by anandamide in urethane-anaesthetized rats. 2. The anandamide-induced pressor effect was not modified by the antagonists of cannabinoid CB(1) and vanilloid TRPV(1) receptors, SR 141716A (3 micromol kg(-1)) and capsazepine (1 micromol kg(-1)), respectively, by bilateral vagotomy and by pithing. Replacement of urethane by pentobarbitone virtually abolished the pressor effect of anandamide, both in pithed and vagotomized and in 'intact' rats (i.e. not treated in this manner). 3. The pressor effect of anandamide was reduced by the nonselective TRPV family inhibitor ruthenium red (3 micromol kg(-1)) and by the blocker of L-type calcium channels nifedipine (1 micromol kg(-1)), both in pithed urethane-anaesthetized rats and in 'intact' urethane-anaesthetized rats. The nonselective beta-adrenoceptor antagonist propranolol (0.1 or 0.3 micromol kg(-1)) and the nonselective NMDA receptor antagonist MK-801 (1 micromol kg(-1)) diminished the anandamide-induced vasopressor response in 'intact' but not in pithed rats. The inhibitory effect of propranolol in 'intact' rats was mimicked by the beta(2)-adrenoceptor antagonist ICI 118551 (1 micromol kg(-1)), but not by the beta(1)-adrenoceptor antagonist CGP 20712 (1 micromol kg(-1)). 4. The present study revealed that two mechanisms may be responsible for the anandamide-induced pressor response in urethane-anaesthetized rats. The first involves the central nervous system (probably the medulla oblongata) and is sensitive to propranolol and MK-801. The second, which is located peripherally (most probably in blood vessels), is sensitive to nifedipine, ruthenium red and pentobarbitone and, hence, probably represents a Ca(2+)-dependent mode of action.
Collapse
Affiliation(s)
- Grzegorz Kwolek
- Zakład Fizjologii Doświadczalnej, Akademia Medyczna w Białymstoku, ul. Mickiewicza 2A, 15-089 Białystok 8, Poland
| | - Agnieszka Zakrzeska
- Zakład Fizjologii Doświadczalnej, Akademia Medyczna w Białymstoku, ul. Mickiewicza 2A, 15-089 Białystok 8, Poland
| | - Eberhard Schlicker
- Institut für Pharmakologie und Toxikologie, Universität Bonn, Reuterstrasse 2b, 53113 Bonn, Germany
| | - Manfred Göthert
- Institut für Pharmakologie und Toxikologie, Universität Bonn, Reuterstrasse 2b, 53113 Bonn, Germany
| | - Grzegorz Godlewski
- Zakład Fizjologii Doświadczalnej, Akademia Medyczna w Białymstoku, ul. Mickiewicza 2A, 15-089 Białystok 8, Poland
| | - Barbara Malinowska
- Zakład Fizjologii Doświadczalnej, Akademia Medyczna w Białymstoku, ul. Mickiewicza 2A, 15-089 Białystok 8, Poland
- Author for correspondence:
| |
Collapse
|
33
|
Jakab B, Helyes Z, Varga A, Bölcskei K, Szabó A, Sándor K, Elekes K, Börzsei R, Keszthelyi D, Pintér E, Petho G, Németh J, Szolcsányi J. Pharmacological characterization of the TRPV1 receptor antagonist JYL1421 (SC0030) in vitro and in vivo in the rat. Eur J Pharmacol 2005; 517:35-44. [PMID: 15978575 DOI: 10.1016/j.ejphar.2005.05.002] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2005] [Revised: 04/28/2005] [Accepted: 05/05/2005] [Indexed: 11/20/2022]
Abstract
The TRPV1 capsaicin receptor is an integrator molecule on primary afferent neurones participating in inflammatory and nociceptive processes. The present paper characterizes the effects of JYL1421 (SC0030), a TRPV1 receptor antagonist, on capsaicin-evoked responses both in vitro and in vivo in the rat. JYL1421 concentration-dependently (0.1-2 microM) inhibited capsaicin-evoked substance P, calcitonin gene-related peptide and somatostatin release from isolated tracheae, while only 2 microM resulted in a significant inhibition of electrically induced neuropeptide release. Capsazepine (0.1-2 microM), as a reference compound, similarly diminished both capsaicin-evoked and electrically evoked peptide release. JYL1421 concentration-dependently decreased capsaicin-induced Ca(2+) accumulation in cultured trigeminal ganglion cells, while capsazepine was much less effective. In vivo 2 mg/kg i.p. JYL1421, but not capsazepine, inhibited capsaicin-induced hypothermia, eye wiping movements and reflex hypotension (a component of the pulmonary chemoreflex or Bezold-Jarisch reflex). Based on these data JYL1421 is a more selective and in most models also a more potent TRPV1 receptor antagonist than capsazepine, therefore it may promote the assessment of the (patho)physiological roles of the TRPV1 receptor.
Collapse
Affiliation(s)
- Balázs Jakab
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Pécs, H-7643, Pécs, Szigeti u. 12, Hungary
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Poblete IM, Orliac ML, Briones R, Adler-Graschinsky E, Huidobro-Toro JP. Anandamide elicits an acute release of nitric oxide through endothelial TRPV1 receptor activation in the rat arterial mesenteric bed. J Physiol 2005; 568:539-51. [PMID: 16081483 PMCID: PMC1474725 DOI: 10.1113/jphysiol.2005.094292] [Citation(s) in RCA: 90] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
In the isolated rat mesenteric bed, the 1 min perfusion with 100 nm anandamide, a concentration that did not evoke vasorelaxation, elicited an acute release of 165.1 +/- 9.2 pmol nitric oxide (NO) that was paralleled by a 2-fold increase in cGMP tissue levels. The rise in NO released was mimicked by either (R)-(+)-methanandamide or the vanilloid receptor agonists resiniferatoxin and (E)-capsaicin but not by its inactive cis-isomer (Z)-capsaicin. The NO release elicited by either anandamide or capsaicin was reduced by the TRPV1 receptor antagonists 5'-iodoresiniferatoxin, SB 366791 and capsazepine as well as by the cannabinoid CB(1) receptor antagonists SR 141716A or AM251. The outflow of NO elicited by anandamide and capsaicin was also reduced by endothelium removal or NO synthase inhibition, suggesting the specific participation of endothelial TRPV1 receptors, rather than the novel endothelial TRPV4 receptors. Consistently, RT-PCR showed the expression of the mRNA coding for the rat TRPV1 receptor in the endothelial cell layer, in addition to its expression in sensory nerves. The participation of sensory nerves on the release of NO was precluded on the basis that neonatal denervation of the myenteric plexus sensory nerves did not modify the pattern of NO release induced by anandamide and capsaicin. We propose that low concentrations of anandamide, devoid of vasorelaxing effects, elicit an acute release of NO mediated predominantly by the activation of endothelial TRPV1 receptors whose physiological significance remains elusive.
Collapse
Affiliation(s)
- Inés M Poblete
- Centro de Regulación Celular y Patología JV Luco, Instituto MIFAB, Departmento de Fisiología, Facultad de Ciencias Biológicas, P. Universidad Católica de Chile, Santiago
| | | | | | | | | |
Collapse
|
35
|
Laporte R, Hui A, Laher I. Pharmacological modulation of sarcoplasmic reticulum function in smooth muscle. Pharmacol Rev 2005; 56:439-513. [PMID: 15602008 DOI: 10.1124/pr.56.4.1] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The sarco/endoplasmic reticulum (SR/ER) is the primary storage and release site of intracellular calcium (Ca2+) in many excitable cells. The SR is a tubular network, which in smooth muscle (SM) cells distributes close to cellular periphery (superficial SR) and in deeper aspects of the cell (deep SR). Recent attention has focused on the regulation of cell function by the superficial SR, which can act as a buffer and also as a regulator of membrane channels and transporters. Ca2+ is released from the SR via two types of ionic channels [ryanodine- and inositol 1,4,5-trisphosphate-gated], whereas accumulation from thecytoplasm occurs exclusively by an energy-dependent sarco-endoplasmic reticulum Ca2+-ATPase pump (SERCA). Within the SR, Ca2+ is bound to various storage proteins. Emerging evidence also suggests that the perinuclear portion of the SR may play an important role in nuclear transcription. In this review, we detail the pharmacology of agents that alter the functions of Ca2+ release channels and of SERCA. We describe their use and selectivity and indicate the concentrations used in investigating various SM preparations. Important aspects of cell regulation and excitation-contractile activity coupling in SM have been uncovered through the use of such activators and inhibitors of processes that determine SR function. Likewise, they were instrumental in the recent finding of an interaction of the SR with other cellular organelles such as mitochondria. Thus, an appreciation of the pharmacology and selectivity of agents that interfere with SR function in SM has greatly assisted in unveiling the multifaceted nature of the SR.
Collapse
Affiliation(s)
- Régent Laporte
- Ferring Research Institute, Inc., Ferring Pharmaceuticals, San Diego, California, USA
| | | | | |
Collapse
|
36
|
Patacchini R, Santicioli P, Giuliani S, Maggi CA. Pharmacological investigation of hydrogen sulfide (H2S) contractile activity in rat detrusor muscle. Eur J Pharmacol 2005; 509:171-7. [PMID: 15733553 DOI: 10.1016/j.ejphar.2005.01.005] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2004] [Revised: 01/05/2005] [Accepted: 01/07/2005] [Indexed: 11/29/2022]
Abstract
We have investigated the mechanism through which hydrogen sulfide (H2S) stimulates capsaicin-sensitive primary afferent neurons in the rat isolated urinary bladder. Sodium hydrogen sulfide (NaHS), a donor of H2S, produced concentration-dependent contractile responses (pEC50=3.5+/-0.1) that were unaffected by the transient receptor potential vanilloid receptor 1 (TRPV1) antagonist capsazepine (30 microM) and SB 366791 (10 microM) and by the N-type Ca2+ channel blocker omega-conotoxin GVIA (omega-CTX; 100 nM). In contrast, the unselective transient receptor potential (TRP) cation channels blocker ruthenium red (30 microM) almost abolished NaHS-induced contractions. Ruthenium red (30 microM) greatly reduced capsaicin-induced contractions, whereas it did not attenuate the contractile response to neurokinin A. The putative TRPV1 receptor antagonist iodo-resiniferatoxin, from 100 nM upward, produced agonist responses per se, and could not be tested against NaHS. We conclude that H2S either acts at TRPV1 receptorial sites unblocked by capsazepine or SB 366791, or stimulates a still unidentified transient receptor potential-like channel co-expressed with TRPV1 on sensory neurons.
Collapse
Affiliation(s)
- Riccardo Patacchini
- Department of Pharmacology, Menarini Ricerche SpA, via Rismondo 12/A, 50131, Florence, Italy.
| | | | | | | |
Collapse
|
37
|
Abstract
Abstract: Sodium azulene sulfonate is a water‐soluble derivative of azulene which is an antiinflammatory component of chamomile of the family of Asteraceae. Sodium azulene sulfonate is clinically used as a therapeutic agent in the treatment of pharyngitis as well as other inflammatory diseases such as tonsillitis, stomatitis and conjunctivitis. There has been no documentation on the effect of sodium azulene sulfonate on pharyngitis in laboratory models, probably because of no availability of such models. We recently established a pharyngitis model using capsaicin application on pharyngeal mucosa in rats. The present study investigated the antipharyngitis activity of sodium azulene sulfonate comparing with those of ruthenium red (vanilloid receptor antagonist, 8.5 and 85 mg/ml), ascorbic acid (antioxidative compound, 100 μg/ml), povidone iodine (gargle as disinfectant, oxidative compound, 5 and 20 mg/ml) and diclofenac sodium (cyclooxygenase inhibitor, 0.1 and 1 mg/ml). As an antipharyngeal effect, the capsaicin‐induced plasma exudation in the pharyngeal mucosa of the rat was evaluated. The capsaicin‐induced plasma exudation in the pharyngeal mucosa was inhibited by sodium azulene sulfonate (100 and 200 μg/ml) as well as ruthenium red and ascorbic acid, but not by povidone iodine and dicrofenac sodium; povidone iodine rather promoted the plasma exudation. In conclusion, the antipharyngitis effect of sodium azulene sulfonate was demonstrated for the first time in a laboratory model. Although the mechanism by which sodium azulene sulfonate inhibited the capsaicin‐induced pharyngitis is not yet unraveled, antioxidative effect, but not inhibitory effect on cyclooxygenase pathway, might be involved.
Collapse
Affiliation(s)
- Hiroyasu Sakai
- Department of Pharmacology, School of Pharmacy, Hoshi University, 2-4-41 Ebara, Shinagawa-ku, Tokyo, Japan.
| | | |
Collapse
|
38
|
Langhorst MF, Schwarzmann N, Guse AH. Ca2+ release via ryanodine receptors and Ca2+ entry: major mechanisms in NAADP-mediated Ca2+ signaling in T-lymphocytes. Cell Signal 2005; 16:1283-9. [PMID: 15337527 DOI: 10.1016/j.cellsig.2004.03.013] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2004] [Revised: 03/23/2004] [Accepted: 03/23/2004] [Indexed: 11/15/2022]
Abstract
Nicotinic acid adenine dinucleotide phosphate (NAADP) is a potent Ca2+ mobilizing nucleotide essentially involved in T cell activation. Using combined microinjection and single cell calcium imaging, we demonstrate that co-injection of NAADP and the D-myo-inositol 1,4,5-trisphosphate antagonist heparin did not inhibit Ca2+ mobilization. In contrast, co-injection of the ryanodine receptor antagonist ruthenium red efficiently blocked NAADP induced Ca2+ signalling. This pharmacological approach was confirmed using T cell clones stably transfected with plasmids expressing antisense mRNA targeted specifically against ryanodine receptors. NAADP induced Ca2+ signaling was strongly reduced in these clones. In addition, inhibition of Ca2+ entry by SK&F 96365 resulted in a dramatically decreased Ca2+ signal upon NAADP injection. Gd3+, a known blocker of Ca2+ release activated Ca2+ entry, only partially inhibited NAADP mediated Ca2+ signaling. These data indicate that in T cells (i) ryanodine receptor are the major intracellular Ca2+ release channels involved in NAADP induced Ca2+ signals, and that (ii) such Ca2+ release events are largely amplified by Ca2+ entry.
Collapse
Affiliation(s)
- Matthias F Langhorst
- Institute of Biochemistry and Molecular Biology I: Cellular Signal Transduction, University Hospital Hamburg-Eppendorf, Center of Experimental Medicine, Martinistr. 52, D-20246, Germany
| | | | | |
Collapse
|
39
|
Bulmer DCE, Jiang W, Hicks GA, Davis JB, Winchester WJ, Grundy D. Vagal selective effects of ruthenium red on the jejunal afferent fibre response to ischaemia in the rat. Neurogastroenterol Motil 2005; 17:102-11. [PMID: 15670270 DOI: 10.1111/j.1365-2982.2004.00586.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
A variety of inflammatory mediators and local metabolites, have been implicated in the sensitivity of intestinal afferent fibres to brief periods of ischaemia and reperfusion. As yet, the contribution of the vanilloid transient receptor potential (TRPV)1 receptor to the response to intestinal ischaemia remains undetermined. In the present study, the effect of pretreatment with the competitive TRPV1 antagonist capsazepine and the non-selective TRPV channel antagonist ruthenium red, on the mesenteric afferent fibre response to ischaemia was examined. In control animals there was a reproducible biphasic increase in whole nerve afferent fibre activity during two brief periods of ischaemia. Treatment with ruthenium red significantly attenuated the early phase increase in afferent fibre activity during ischaemia. However, capsazepine treatment did not significantly alter the afferent fibre response to either ischaemia or reperfusion. Further experiments in chronically vagotomized animals indicated that the early phase response to ischaemia was mediated via vagal afferent fibres. The mechanism via which ruthenium red selectively inhibited vagal afferent fibres during ischaemia is unknown, but it does not appear to involve blockade of the TRPV1 receptor.
Collapse
Affiliation(s)
- D C E Bulmer
- Department of Biomedical Sciences, University of Sheffield, Sheffield, UK
| | | | | | | | | | | |
Collapse
|
40
|
Cheng FHM, Andrews PLR, Moreaux B, Ngan MP, Rudd JA, Sam TSW, Wai MK, Wan C. Evaluation of the anti-emetic potential of anti-migraine drugs to prevent resiniferatoxin-induced emesis in Suncus murinus (house musk shrew). Eur J Pharmacol 2005; 508:231-8. [PMID: 15680276 DOI: 10.1016/j.ejphar.2004.12.022] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2004] [Revised: 12/07/2004] [Accepted: 12/10/2004] [Indexed: 11/18/2022]
Abstract
Activation of vanilloid receptors has commonly been used to facilitate neurogenic inflammation and plasma exudation to model components of the pathogenesis of migraine; however, these studies have been performed mainly in species lacking the emetic reflex. In the present studies, therefore, we used Suncus murinus, a species of insectivore capable of emesis, to investigate if the vanilloid receptor agonist resiniferatoxin is capable of modeling the emesis associated with migraine. Resiniferatoxin (100 nmol/kg, s.c.) induced an emetic response that was antagonized significantly (P<0.05) by ruthenium red (1-3 micromol), (2R-trans)-4-[1-[3,5-bis(trifluromethyl)benzoyl]-2-(phenylmethyl)-4-piperidinyl]-N-(2,6-dimethylphenyl)-1-acetamide (S)-hydroxybutanedioate (R116301; 10-100 micromol/kg), and scopolamine (1 micromol/kg), but not by dihydroergotamine (0.3-3 micromol/kg), sumatriptan (1-10 micromol/kg), methysergide (1-10 micromol/kg), tropanyl 3,5-dichlorobenzoate (MDL72222; 3-30 micromol/kg), ondansetron (0.3-3 micromol/kg), metoclopramide (3-30 micromol/kg), domperidone (3-30 micromol/kg), diphenhydramine (1-10 micromol/kg), or indomethacin (3-30 micromol/kg). The failure of a wide range of representative anti-migraine drugs to reduce retching and vomiting limits the use of this model to identify/investigate novel treatments for the emesis (and nausea) associated with migraine attacks in humans. However, the results provide further evidence for the involvement of a novel vanilloid receptor in resiniferatoxin-induced emesis and implicate both tachykinins and acetylcholine in the pathway(s) activated by resiniferatoxin in S. murinus.
Collapse
Affiliation(s)
- Frankie H M Cheng
- Emesis Research Group, Department of Pharmacology, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Kadowaki M, Kuramoto H, Takaki M. Combined determination with functional and morphological studies of origin of nerve fibers expressing transient receptor potential vanilloid 1 in the myenteric plexus of the rat jejunum. Auton Neurosci 2004; 116:11-8. [PMID: 15556833 DOI: 10.1016/j.autneu.2004.08.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2004] [Revised: 07/30/2004] [Accepted: 08/21/2004] [Indexed: 11/20/2022]
Abstract
The aim of this study was to determine the action of capsaicin in isolated rat intestine and the origin of nerve fibers expressing transient receptor potential vanilloid 1 (TRPV1: capsaicin receptor) in the rat jejunum by combination of functional and immunohistochemical experiments. Capsaicin (1 microM) produced a prolonged relaxation response (52. +/-15.3% of the relaxation response to papaverine, mean +/- S.D., n=27) of the isolated jejunum in the presence of atropine and guanethidine. Pretreatment with the TRPV1 antagonist, capsazepine (10 microM) and ruthenium red (3 microM) significantly reduced the relaxation response to capsaicin by 78% (P<0.01) and 38% (P<0.05), respectively. Tetrodotoxin and calcitonin gene-related peptide (CGRP)-desensitization significantly reduced the response to capsaicin by 72% (P<0.01) and 42% (P<0.01), respectively. Therefore, we investigated the distribution of TRPV1-immunoreactivity (IR) in the myenteric plexus of the rat jejunum. Using antisera raised against either the N-terminal or C-terminal domains of rat TRPV1, TRPV1-IR was present in the nerve fibers, but not in the cell bodies of myenteric neurons. These TRPV1-immunoreactive nerve fibers were running in myenteric ganglia and their interconnecting strands. Most TRPV1-immunoreactive nerve fibers showed CGRP-IR, whereas few VR1-immunoreactive nerve fibers showed substance P-IR. After chronic denervation of the extrinsic nerve supply to the jejunum, both the relaxation response to capsaicin and TRPV1-immunoreactive nerve fibers completely disappeared. These findings indicate that these TRPV1-immunoreactive nerve fibers in the rat jejunum derive from extrinsic neurons and that activation of TRPV1 produces the relaxation response in the rat jejunum, at least in part, through the release of CGRP from nerve fibers expressing TRPV1.
Collapse
Affiliation(s)
- Makoto Kadowaki
- Division of Gastrointestinal Pathophysiology, Department of Bioscience, Institute of Natural Medicine, Toyama Medical and Pharmaceutical University, 2630 Sugitani, Toyama 930-0194, Japan.
| | | | | |
Collapse
|
42
|
Eguchi S, Tezuka S, Hobara N, Akiyama S, Kurosaki Y, Kawasaki H. Vanilloid receptors mediate adrenergic nerve- and CGRP-containing nerve-dependent vasodilation induced by nicotine in rat mesenteric resistance arteries. Br J Pharmacol 2004; 142:1137-46. [PMID: 15249421 PMCID: PMC1575167 DOI: 10.1038/sj.bjp.0705773] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Previous studies showed that nicotine induces adrenergic nerve-dependent vasodilation that is mediated by endogenous calcitonin gene-related peptide (CGRP) released from CGRP-containing (CGRPergic) nerves. The mechanisms underlying the nicotine-induced vasodilation were further studied. Rat mesenteric vascular beds without endothelium were contracted by perfusion with Krebs solution containing methoxamine, and the perfusion pressure was measured with a pressure transducer. Perfusion of nicotine (1-100 microm) for 1 min caused concentration-dependent vasodilation. Capsazepine (vanilloid receptor-1 antagonist; 1-10 microm) and ruthenium red (inhibitor of vanilloid response; 1-30 microm) concentration-dependently inhibited the nicotine-induced vasodilation without affecting the vasodilator response to exogenous CGRP. Nicotine-induced vasodilation was not inhibited by treatment with 3,4-dihydroxyphenylalanine (DOPA) receptor antagonist (l-DOPA cyclohexyl ester; 0.001-10 microm), dopamine D1 receptor-selective antagonist (SCH23390; 1-10 microm), dopamine D2 receptor antagonist (haloperidol; 0.1-0.5 microm), ATP P2x receptor-desensitizing agonist (alpha,beta-methylene ATP; 1-10 microm), adenosine A2 receptor antagonist (8(p-sulfophenyl)theophylline; 10-50 microm) or neuropeptide Y (NPY)-Y1 receptor antagonist (BIBP3226; 0.1-0.5 microm). Immunohistochemical staining of the mesenteric artery showed dense innervation of CGRP- and vanilloid receptor-1-positive nerves, with both immunostainings appearing in the same neuron. The mesenteric artery was also densely innervated by NPY-positive nerves. Double immunostainings showed that both NPY and CGRP immunoreactivities appeared in the same neuron of the artery. These results suggest that nicotine acts on presynaptic nicotinic receptors to release adrenergic neurotransmitter(s) or related substance(s), which then stimulate vanilloid receptor-1 on CGRPergic nerves, resulting in CGRP release and vasodilation.
Collapse
MESH Headings
- Adrenergic Fibers/drug effects
- Adrenergic Fibers/physiology
- Animals
- Calcitonin Gene-Related Peptide/physiology
- Calcitonin Gene-Related Peptide Receptor Antagonists
- Dopamine Antagonists/pharmacology
- Dose-Response Relationship, Drug
- Electric Stimulation
- Male
- Mesenteric Arteries/drug effects
- Mesenteric Arteries/innervation
- Mesenteric Arteries/physiology
- Microscopy, Confocal
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/innervation
- Muscle, Smooth, Vascular/physiology
- Nicotine/administration & dosage
- Nicotine/pharmacology
- Purinergic P1 Receptor Antagonists
- Purinergic P2 Receptor Antagonists
- Rats
- Rats, Wistar
- Receptors, Calcitonin Gene-Related Peptide/metabolism
- Receptors, Drug/antagonists & inhibitors
- Receptors, Drug/physiology
- Receptors, Neuropeptide Y/antagonists & inhibitors
- Receptors, Neuropeptide Y/metabolism
- Receptors, Purinergic P1/metabolism
- Receptors, Purinergic P2/metabolism
- Ruthenium Red/administration & dosage
- Ruthenium Red/pharmacology
- Vasodilation
- Vasodilator Agents/pharmacology
Collapse
Affiliation(s)
- Shinji Eguchi
- Department of Clinical Pharmaceutical Science, Graduate School of Natural Science and Technology, Okayama University, 1-1-1 Tsushima-naka, Okayama 700-8530, Japan
| | - Satoko Tezuka
- Department of Clinical Pharmaceutical Science, Graduate School of Natural Science and Technology, Okayama University, 1-1-1 Tsushima-naka, Okayama 700-8530, Japan
| | - Narumi Hobara
- Department of Clinical Pharmaceutical Science, Graduate School of Natural Science and Technology, Okayama University, 1-1-1 Tsushima-naka, Okayama 700-8530, Japan
| | - Shinji Akiyama
- Department of Clinical Pharmaceutical Science, Graduate School of Natural Science and Technology, Okayama University, 1-1-1 Tsushima-naka, Okayama 700-8530, Japan
| | - Yuji Kurosaki
- Department of Clinical Pharmaceutical Science, Graduate School of Natural Science and Technology, Okayama University, 1-1-1 Tsushima-naka, Okayama 700-8530, Japan
| | - Hiromu Kawasaki
- Department of Clinical Pharmaceutical Science, Graduate School of Natural Science and Technology, Okayama University, 1-1-1 Tsushima-naka, Okayama 700-8530, Japan
- Author for correspondence:
| |
Collapse
|
43
|
Szallasi A, Appendino G. Vanilloid receptor TRPV1 antagonists as the next generation of painkillers. Are we putting the cart before the horse? J Med Chem 2004; 47:2717-23. [PMID: 15139748 DOI: 10.1021/jm030560j] [Citation(s) in RCA: 122] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Arpad Szallasi
- Department of Pathology and Laboratory Medicine, 6 Founders Building, Hospital of the University of Pennsylvania, 3400 Spruce Street, Philadelphia, Pennsylvania 19104, USA.
| | | |
Collapse
|
44
|
Chen SC, Chang TJ, Wu FS. Competitive inhibition of the capsaicin receptor-mediated current by dehydroepiandrosterone in rat dorsal root ganglion neurons. J Pharmacol Exp Ther 2004; 311:529-36. [PMID: 15201344 DOI: 10.1124/jpet.104.069096] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The effects of dehydroepiandrosterone (5-androsten-3beta-ol-17-one; DHEA) and related steroids on the capsaicin receptor-mediated current were studied in acutely dissociated rat dorsal root ganglion neurons using the whole-cell voltage-clamp technique. DHEA rapidly and reversibly inhibited the capsaicin-induced current in a concentration-dependent manner, with an EC(50) of 6.7 microM and a maximal inhibition of 100%. DHEA increased the capsaicin EC(50) with little effect on the capsaicin maximal response, suggesting that the blocking action of DHEA is competitive. Neither the capsaicin response nor inhibition of the capsaicin response by extracellularly applied DHEA was significantly affected by inclusion of a saturating concentration of DHEA in the electrode buffer, arguing that DHEA acted at the extracellular surface of the membrane. Moreover, DHEA did not act through protein phosphatases to inhibit the capsaicin-induced current. Furthermore, the stereoisomer of DHEA, 5-androsten-3alpha-ol-17-one, failed to inhibit the capsaicin-induced current, producing instead a potentiating effect on the capsaicin response, demonstrating that the interaction of steroids with the capsaicin receptor is stereospecific. The inhibitory action of DHEA on the capsaicin-induced current may provide a basis for reducing capsaicin receptor-mediated nociception.
Collapse
Affiliation(s)
- Shu-Cheng Chen
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, 1 Ta-Hsiue Road, Tainan 70101, Taiwan
| | | | | |
Collapse
|
45
|
Pethö G, Izydorczyk I, Reeh PW. Effects of TRPV1 receptor antagonists on stimulated iCGRP release from isolated skin of rats and TRPV1 mutant mice. Pain 2004; 109:284-290. [PMID: 15157689 DOI: 10.1016/j.pain.2004.01.038] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2003] [Accepted: 01/12/2004] [Indexed: 11/26/2022]
Abstract
Capsaicin antagonists including ruthenium red, capsazepine and iodo-resiniferatoxin (I-RTX) have recently been shown to inhibit the activation by noxious heat of the capsaicin receptor (TRPV1) expressed in non-neuronal host cells, and natively, in cultured dorsal root ganglion cells. Noxious heat has been shown to release immunoreactive calcitonin gene-related peptide (iCGRP) from the isolated rat skin. In this model, ruthenium red, I-RTX as well as capsazepine 10 microM caused no alteration in iCGRP release at 32 degrees C by themselves whereas capsazepine 100 microM doubled it reversibly. In wild-type mice 100 microM capsazepine also stimulated iCGRP release while it was without effect in TRPV1 knockout littermates. In the rat skin, both ruthenium red and capsazepine (10/100 microM) reduced and abolished, respectively, capsaicin-induced iCGRP release while I-RTX (1/10 microM) was ineffective. Only ruthenium red 100 microM showed an unspecific effect inhibiting iCGRP release induced by KCl. Ruthenium red and capsazepine (10/100 microM) caused no significant alteration of iCGRP release induced by heat stimulation at 47 degrees C. Employing 45 degrees C stimulation intensity, capsazepine and I-RTX (in the higher concentrations) showed a significant facilitatory effect on the heat response suggesting a partial agonistic action of the compounds. It is concluded that noxious heat-induced iCGRP release in the isolated rat skin occurs through a mechanism that is not inhibited by TRPV1 antagonism reflecting a different pharmacological profile of noxious heat transduction in terminals of sensory neurons compared to that in cultured cell bodies and TRPV1-transfected host cells.
Collapse
Affiliation(s)
- Gábor Pethö
- Institute of Physiology and Experimental Pathophysiology, University Erlangen/Nürnberg, Universitaetsstr. 17, D-91054 Erlangen, Germany
| | | | | |
Collapse
|
46
|
Kollarik M, Undem BJ. Activation of bronchopulmonary vagal afferent nerves with bradykinin, acid and vanilloid receptor agonists in wild-type and TRPV1-/- mice. J Physiol 2003; 555:115-23. [PMID: 14634201 PMCID: PMC1664811 DOI: 10.1113/jphysiol.2003.054890] [Citation(s) in RCA: 118] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
The vanilloid receptor TRPV1 (formerly VR1) has been implicated in the activation of nociceptive sensory nerves by capsaicin, noxious heat, protons, bradykinin, cannabinoids such as anandamide, and certain metabolites of arachidonic acid. Using TRPV1 knockout mouse (TRPV1-/-) we address the question of whether TRPV1 is obligatory for action potential discharge in vagal C-fibre terminals evoked by capsaicin, anandamide, acid and bradykinin. The response of a defined subtype of the vagal afferent bronchopulmonary C-fibres (conduction velocity < 0.7 ms(-1)) to the putative TRPV1 activators was studied in vitro in the mouse isolated/perfused lung-nerve preparation. Capsaicin (1 microm) evoked action potential discharge of approximately 90% (28/31) of C-fibres in the TRPV1+/+ mice, but failed to activate bronchopulmonary C-fibres in TRPV1-/- animals (n = 10). Anandamide (3-100 microm) induced concentration-dependent activation of capsaicin-sensitive TRPV1+/+ C-fibres with a threshold of 3-10 microm, but failed to evoke substantive discharge in TRPV1-/- C-fibres. In the TRPV1+/+ mice, the B2 receptor-mediated activation by bradykinin (1 microm) was restricted to the capsaicin-sensitive C-fibres. Bradykinin was effective in evoking B2 receptor-mediated action potential discharge in TRPV1-/- C-fibres, but the response was significantly (P < 0.05) less persistent than in TRPV1+/+ C-fibres. Exposing the tissue to acid (pH = 5) excited both TRPV1+/+ and TRPV1-/- C-fibres. We conclude that TRPV1 is obligatory for vagal C-fibre activation by capsaicin and anandamide. By contrast, whereas TRPV1 may have a modulatory role in bradykinin and acid-induced activation of bronchopulmonary C-fibres, it is not required for action potential discharge evoked by these stimuli.
Collapse
Affiliation(s)
- M Kollarik
- Johns Hopkins Asthma Center, Johns Hopkins School of Medicine, 5501 Hopkins Bayview Circle, Baltimore, MD 21224, USA
| | | |
Collapse
|
47
|
Pongpiriyadacha Y, Matsuda H, Morikawa T, Asao Y, Yoshikawa M. Protective effects of polygodial on gastric mucosal lesions induced by necrotizing agents in rats and the possible mechanisms of action. Biol Pharm Bull 2003; 26:651-7. [PMID: 12736506 DOI: 10.1248/bpb.26.651] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The effects of polygodial isolated from the leaves of Tasmannia lanceolata on necrotizing agents-induced gastric lesions in rats were compared with capsaicin. Polygodial markedly inhibited the gastric mucosal lesions induced by several necrotizing agents, such as ethanol (ED(50)=0.029 mg/kg, p.o.), 0.6 M HCl (ED(50)=0.26 mg/kg, p.o.), and aspirin (ED(50)=0.38 mg/kg, p.o.), and partly inhibited the gastric mucosal lesions induced by indomethacin, but showed no significant effect on acid output in pylorus-ligated rats at doses of 0.05-0.5 mg/kg. The gastroprotection of polygodial was attenuated by pretreatment with indomethacin (10 mg/kg, s.c.), N(G)-nitro-L-arginine methyl ester (70 mg/kg, i.p.), N-ethylmaleimide (10 mg/kg, s.c.) and ruthenium red (3.5 mg/kg, s.c.). Polygodial (0.2 mg/kg, p.o.) increased the amount of reduced glutathione in gastric mucosa of ethanol-treated group. These results suggested that endogenous prostaglandins, nitric oxide, sulfhydryl compounds and vanilloid receptor-mediated effects are involved in the protective effect of polygodial.
Collapse
|
48
|
Rigoni M, Trevisani M, Gazzieri D, Nadaletto R, Tognetto M, Creminon C, Davis JB, Campi B, Amadesi S, Geppetti P, Harrison S. Neurogenic responses mediated by vanilloid receptor-1 (TRPV1) are blocked by the high affinity antagonist, iodo-resiniferatoxin. Br J Pharmacol 2003; 138:977-85. [PMID: 12642400 PMCID: PMC1573721 DOI: 10.1038/sj.bjp.0705110] [Citation(s) in RCA: 85] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
(1) Stimulation of the vanilloid receptor-1 (TRPV1) results in the activation of nociceptive and neurogenic inflammatory responses. Poor specificity and potency of TRPV1 antagonists has, however, limited the clarification of the physiological role of TRPV1. (2) Recently, iodo-resiniferatoxin (I-RTX) has been reported to bind as a high affinity antagonist at the native and heterologously expressed rat TRPV1. Here we have studied the ability of I-RTX to block a series of TRPV1 mediated nociceptive and neurogenic inflammatory responses in different species (including transfected human TRPV1). (3) We have demonstrated that I-RTX inhibited capsaicin-induced mobilization of intracellular Ca(2+) in rat trigeminal neurons (IC(50) 0.87 nM) and in HEK293 cells transfected with the human TRPV1 (IC(50) 0.071 nM). (4) Furthermore, I-RTX significantly inhibited both capsaicin-induced CGRP release from slices of rat dorsal spinal cord (IC(50) 0.27 nM) and contraction of isolated guinea-pig and rat urinary bladder (pK(B) of 10.68 and 9.63, respectively), whilst I-RTX failed to alter the response to high KCl or SP. (5) Finally, in vivo I-RTX significantly inhibited acetic acid-induced writhing in mice (ED(50) 0.42 micro mol kg(-1)) and plasma extravasation in mouse urinary bladder (ED(50) 0.41 micro mol kg(-1)). (6) In in vitro and in vivo TRPV1 activated responses I-RTX was approximately 3 log units and approximately 20 times more potent than capsazepine, respectively. This high affinity antagonist, I-RTX, may be an important tool for future studies in pain and neurogenic inflammatory models.
Collapse
Affiliation(s)
- Michela Rigoni
- Department of Experimental & Clinical Medicine, Pharmacology Unit, University of Ferrara, Via Fossato di Mortara 19, 44100 Ferrara, Italy
| | - Marcello Trevisani
- Department of Experimental & Clinical Medicine, Pharmacology Unit, University of Ferrara, Via Fossato di Mortara 19, 44100 Ferrara, Italy
| | - David Gazzieri
- Department of Experimental & Clinical Medicine, Pharmacology Unit, University of Ferrara, Via Fossato di Mortara 19, 44100 Ferrara, Italy
| | - Riccardo Nadaletto
- Department of Experimental & Clinical Medicine, Pharmacology Unit, University of Ferrara, Via Fossato di Mortara 19, 44100 Ferrara, Italy
| | - Michele Tognetto
- Department of Experimental & Clinical Medicine, Pharmacology Unit, University of Ferrara, Via Fossato di Mortara 19, 44100 Ferrara, Italy
| | - Christophe Creminon
- CEA, Service de Pharmacologie et d'Immunologie, DRM, CEA-Saclay, Gif sur Yvette, France
| | - John B Davis
- Neurology-CEDD, GlaxoSmithKline, New Frontiers Science Park, Third Avenue, Harlow, Essex
| | - Barbara Campi
- Department of Experimental & Clinical Medicine, Pharmacology Unit, University of Ferrara, Via Fossato di Mortara 19, 44100 Ferrara, Italy
| | - Silvia Amadesi
- Department of Experimental & Clinical Medicine, Pharmacology Unit, University of Ferrara, Via Fossato di Mortara 19, 44100 Ferrara, Italy
| | - Pierangelo Geppetti
- Department of Experimental & Clinical Medicine, Pharmacology Unit, University of Ferrara, Via Fossato di Mortara 19, 44100 Ferrara, Italy
- Author for correspondence:
| | - Selena Harrison
- Department of Experimental & Clinical Medicine, Pharmacology Unit, University of Ferrara, Via Fossato di Mortara 19, 44100 Ferrara, Italy
| |
Collapse
|
49
|
|
50
|
Kyoi T, Oka M, Noda K, Ukai Y. Irsogladine Prevents Monochloramine-Induced Gastric Mucosal Lesions by Improving the Decrease in Mucosal Blood Flow Due to the Disturbance of Nitric Oxide Synthesis in Rats. J Pharmacol Sci 2003; 93:314-20. [PMID: 14646249 DOI: 10.1254/jphs.93.314] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
Abstract
The inhibitory effect of an anti-ulcer drug irsogladine [2,4-diamino-6-(2,5-dichlorophenyl)-s-triazine maleate] on monochloramine (NH(2)Cl)-induced gastric mucosal lesions and its mechanisms of action were clarified in rats. Irsogladine dose-dependently prevented the formation of gastric mucosal lesions induced by 60 mM NH(2)Cl. The mucosal protective effect of irsogladine was not influenced by capsaicin-sensitive sensory defunctionalization. On the other hand, its protective effect was diminished by the inhibitor of nitric oxide synthase N(G)-nitro-L-arginine methylester (L-NAME), but not by the inducible nitric oxide synthase selective inhibitor aminoguanidine. Irsogladine restored the NH(2)Cl-induced decrease in the gastric cGMP formation as an index of nitric oxide synthesis, while it alone had no influence on the cGMP formation in intact tissues. Pretreatment with L-NAME abolished the recovery of cGMP by irsogladine. Furthermore, irsogladine ameliorated the NH(2)Cl-induced decrease in gastric mucosal blood flow, which was also reversed by pretreatment with L-NAME. These findings suggest that the improvement of the decrease in mucosal blood flow subsequent to the disturbance of gastric nitric oxide synthesis is involved in the protective effect of irsogladine on gastric mucosal lesions caused by NH(2)Cl.
Collapse
Affiliation(s)
- Takashi Kyoi
- Research Laboratories, Nippon Shinyaku Co., Ltd., Kyoto, Japan.
| | | | | | | |
Collapse
|