1
|
Torres-Rico M, García-Calvo V, Gironda-Martínez A, Pascual-Guerra J, García AG, Maneu V. Targeting calciumopathy for neuroprotection: focus on calcium channels Cav1, Orai1 and P2X7. Cell Calcium 2024; 123:102928. [PMID: 39003871 DOI: 10.1016/j.ceca.2024.102928] [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: 05/30/2024] [Revised: 07/02/2024] [Accepted: 07/05/2024] [Indexed: 07/16/2024]
Abstract
As the uncontrolled entry of calcium ions (Ca2+) through plasmalemmal calcium channels is a cell death trigger, the conjecture is here raised that mitigating such an excess of Ca2+ entry should rescue from death the vulnerable neurons in neurodegenerative diseases (NDDs). However, this supposition has failed in some clinical trials (CTs). Thus, a recent CT tested whether isradipine, a blocker of the Cav1 subtype of voltage-operated calcium channels (VOCCs), exerted a benefit in patients with Parkinson's disease (PD); however, outcomes were negative. This is one more of the hundreds of CTs done under the principle of one-drug-one-target, that have failed in Alzheimer's disease (AD) and other NDDs during the last three decades. As there are myriad calcium channels to let Ca2+ ions gain the cell cytosol, it seems reasonable to predict that blockade of Ca2+ entry through a single channel may not be capable of preventing the Ca2+ flood of cells by the uncontrolled Ca2+ entry. Furthermore, as Ca2+ signaling is involved in the regulation of myriad functions in different cell types, it seems also reasonable to guess that a therapy should be more efficient by targeting different cells with various drugs. Here, we propose to mitigate Ca2+ entry by the simultaneous partial blockade of three quite different subtypes of plasmalemmal calcium channels that is, the Cav1 subtype of VOCCs, the Orai1 store-operated calcium channel (SOCC), and the purinergic P2X7 calcium channel. All three channels are expressed in both microglia and neurons. Thus, by targeting the three channels with a combination of three drug blockers we expect favorable changes in some of the pathogenic features of NDDs, namely (i) to mitigate Ca2+ entry into microglia; (ii) to decrease the Ca2+-dependent microglia activation; (iii) to decrease the sustained neuroinflammation; (iv) to decrease the uncontrolled Ca2+ entry into neurons; (v) to rescue vulnerable neurons from death; and (vi) to delay disease progression. In this review we discuss the arguments underlying our triad hypothesis in the sense that the combination of three repositioned medicines targeting Cav1, Orai1, and P2X7 calcium channels could boost neuroprotection and delay the progression of AD and other NDDs.
Collapse
Affiliation(s)
| | | | - Adrián Gironda-Martínez
- Instituto Fundación Teófilo Hernando, Madrid, Spain; Departamento de Farmacología y Terapéutica, Universidad Autónoma de Madrid, Madrid, Spain
| | | | - Antonio G García
- Instituto Fundación Teófilo Hernando, Madrid, Spain; Departamento de Farmacología y Terapéutica, Universidad Autónoma de Madrid, Madrid, Spain; Facultad de Medicina, Instituto de Investigación Sanitaria del Hospital Universitario La Princesa, Universidad Autónoma de Madrid, Madrid, Spain.
| | - Victoria Maneu
- Departamento de Óptica, Farmacología y Anatomía, Universidad de Alicante, Alicante, Spain.
| |
Collapse
|
2
|
Mignen O, Vannier JP, Schneider P, Renaudineau Y, Abdoul-Azize S. Orai1 Ca 2+ channel modulators as therapeutic tools for treating cancer: Emerging evidence! Biochem Pharmacol 2024; 219:115955. [PMID: 38040093 DOI: 10.1016/j.bcp.2023.115955] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 11/25/2023] [Accepted: 11/28/2023] [Indexed: 12/03/2023]
Abstract
In non-excitable cells, Orai proteins represent the main channel for Store-Operated Calcium Entry (SOCE), and also mediate various store-independent Calcium Entry (SICE) pathways. Deregulation of these pathways contribute to increased tumor cell proliferation, migration, metastasis, and angiogenesis. Among Orais, Orai1 is an attractive therapeutic target explaining the development of specific modulators. Therapeutic trials using Orai1 channel inhibitors have been evaluated for treating diverse diseases such as psoriasis and acute pancreatitis, and emerging data suggest that Orai1 channel modulators may be beneficial for cancer treatment. This review discusses herein the importance of Orai1 channel modulators as potential therapeutic tools and the added value of these modulators for treating cancer.
Collapse
Affiliation(s)
| | | | | | - Yves Renaudineau
- Laboratory of Immunology, CHU Purpan Toulouse, INSERM U1291, CNRS U5051, University Toulouse III, 31062 Toulouse, France
| | - Souleymane Abdoul-Azize
- LBAI, UMR1227, Univ Brest, Inserm, Brest, France; Normandie Univ., UNIROUEN, INSERM, U1234, Rouen 76000, France.
| |
Collapse
|
3
|
Kong X, Wang F, Chen Y, Liang X, Yin Y, Liu H, Luo G, Li Y, Liang S, Wang Y, Liu Z, Tang C. Molecular action mechanisms of two novel and selective calcium release-activated calcium channel antagonists. Int J Biol Macromol 2023; 253:126937. [PMID: 37722647 DOI: 10.1016/j.ijbiomac.2023.126937] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 09/11/2023] [Accepted: 09/15/2023] [Indexed: 09/20/2023]
Abstract
The prototypical calcium release-activated calcium (CRAC) channel, composed of STIM1 and Orai1, is a sought-after drug target for treating autoimmune disorders. Herein, we identified two novel and selective CRAC channel inhibitors, the indole-like compound C63368 and pyrazole core-containing compound C79413, potently and reversibly inhibiting the CRAC channel with low micromolar IC50s and sparing various off-target ion channels. These two compounds did not inhibit STIM1 activation or its coupling with Orai1, nor did they affect the channel's calcium-dependent fast inactivation. Instead, they directly acted on the Orai1 protein, with the channel's pore geometry profoundly affecting their potencies. In vitro, C63368 and C79413 effectively inhibited Jurkat cell proliferation and cytokines production in human T lymphocytes. Intragastric administration of C63368 and C79413 to mice yielded great therapeutic benefits in psoriasis and colitis animal models of autoimmune disorders, reducing serum cytokines production and significantly relieving pathological symptoms. It's worth noting, that this study provided the first insight into the characterization and mechanistic investigation of an indole-like CRAC channel antagonist. Altogether, the identification of these two highly selective CRAC channel antagonists, coupled with the elucidation of their action mechanisms, not only provides valuable template molecules but also offers profound insights for drug development targeting the CRAC channel.
Collapse
Affiliation(s)
- Xiangjin Kong
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha 410081, China; Peptide and Small Molecule Drug R&D Platform, Furong Laboratory, Changsha 40081, China
| | - Feifan Wang
- Beijing Key Laboratory of Gene Resource and Molecular Development, College of Life Sciences, Beijing Normal University, Beijing 100875, China
| | - Yan Chen
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha 410081, China
| | - Xinyao Liang
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha 410081, China
| | - Yuan Yin
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha 410081, China
| | - Hao Liu
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha 410081, China
| | - Guoqing Luo
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha 410081, China
| | - Yinping Li
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha 410081, China
| | - Songping Liang
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha 410081, China
| | - Youjun Wang
- Beijing Key Laboratory of Gene Resource and Molecular Development, College of Life Sciences, Beijing Normal University, Beijing 100875, China; Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, College of Life Sciences, Beijing Normal University, Beijing 100875, China.
| | - Zhonghua Liu
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha 410081, China; Peptide and Small Molecule Drug R&D Platform, Furong Laboratory, Changsha 40081, China.
| | - Cheng Tang
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha 410081, China; Peptide and Small Molecule Drug R&D Platform, Furong Laboratory, Changsha 40081, China.
| |
Collapse
|
4
|
Yao Y, Borkar NA, Zheng M, Wang S, Pabelick CM, Vogel ER, Prakash YS. Interactions between calcium regulatory pathways and mechanosensitive channels in airways. Expert Rev Respir Med 2023; 17:903-917. [PMID: 37905552 PMCID: PMC10872943 DOI: 10.1080/17476348.2023.2276732] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 10/25/2023] [Indexed: 11/02/2023]
Abstract
INTRODUCTION Asthma is a chronic lung disease influenced by environmental and inflammatory triggers and involving complex signaling pathways across resident airway cells such as epithelium, airway smooth muscle, fibroblasts, and immune cells. While our understanding of asthma pathophysiology is continually progressing, there is a growing realization that cellular microdomains play critical roles in mediating signaling relevant to asthma in the context of contractility and remodeling. Mechanosensitive pathways are increasingly recognized as important to microdomain signaling, with Piezo and transient receptor protein (TRP) channels at the plasma membrane considered important for converting mechanical stimuli into cellular behavior. Given their ion channel properties, particularly Ca2+ conduction, a question becomes whether and how mechanosensitive channels contribute to Ca2+ microdomains in airway cells relevant to asthma. AREAS COVERED Mechanosensitive TRP and Piezo channels regulate key Ca2+ regulatory proteins such as store operated calcium entry (SOCE) involving STIM and Orai channels, and sarcoendoplasmic (SR) mechanisms such as IP3 receptor channels (IP3Rs), and SR Ca2+ ATPase (SERCA) that are important in asthma pathophysiology including airway hyperreactivity and remodeling. EXPERT OPINION Physical and/or functional interactions between Ca2+ regulatory proteins and mechanosensitive channels such as TRP and Piezo can toward understanding asthma pathophysiology and identifying novel therapeutic approaches.
Collapse
Affiliation(s)
- Yang Yao
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi’an Medical University, Xi’an, Shaanxi, China
- Department of Anesthesiology, Mayo Clinic, Rochester, MN, USA
| | - Niyati A Borkar
- Department of Anesthesiology, Mayo Clinic, Rochester, MN, USA
| | - Mengning Zheng
- Department of Anesthesiology, Mayo Clinic, Rochester, MN, USA
- Department of Respiratory and Critical Care Medicine, Guizhou Province People’s Hospital, Guiyang, Guizhou, China
| | - Shengyu Wang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi’an Medical University, Xi’an, Shaanxi, China
| | - Christina M Pabelick
- Department of Anesthesiology, Mayo Clinic, Rochester, MN, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Elizabeth R Vogel
- Department of Anesthesiology, Mayo Clinic, Rochester, MN, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - YS Prakash
- Department of Anesthesiology, Mayo Clinic, Rochester, MN, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
5
|
Rubaiy HN. ORAI Calcium Channels: Regulation, Function, Pharmacology, and Therapeutic Targets. Pharmaceuticals (Basel) 2023; 16:162. [PMID: 37259313 PMCID: PMC9967976 DOI: 10.3390/ph16020162] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 01/15/2023] [Accepted: 01/18/2023] [Indexed: 11/25/2023] Open
Abstract
The changes in intracellular free calcium (Ca2+) levels are one of the most widely regulators of cell function; therefore, calcium as a universal intracellular mediator is involved in very important human diseases and disorders. In many cells, Ca2+ inflow is mediated by store-operated calcium channels, and it is recognized that the store-operated calcium entry (SOCE) is mediated by the two partners: the pore-forming proteins Orai (Orai1-3) and the calcium store sensor, stromal interaction molecule (STIM1-2). Importantly, the Orai/STIM channels are involved in crucial cell signalling processes such as growth factors, neurotransmitters, and cytokines via interaction with protein tyrosine kinase coupled receptors and G protein-coupled receptors. Therefore, in recent years, the issue of Orai/STIM channels as a drug target in human diseases has received considerable attention. This review summarizes and highlights our current knowledge of the Orai/STIM channels in human diseases and disorders, including immunodeficiency, myopathy, tubular aggregate, Stormorken syndrome, York platelet syndrome, cardiovascular and metabolic disorders, and cancers, as well as suggesting these channels as drug targets for pharmacological therapeutic intervention. Moreover, this work will also focus on the pharmacological modulators of Orai/STIM channel complexes. Together, our thoughtful of the biology and physiology of the Orai/STIM channels have grown remarkably during the past three decades, and the next important milestone in the field of store-operated calcium entry will be to identify potent and selective small molecules as a therapeutic agent with the purpose to target human diseases and disorders for patient benefit.
Collapse
Affiliation(s)
- Hussein N Rubaiy
- Department of Laboratory Medicine, Division of Clinical Pharmacology, Karolinska Institute and Karolinska University Hospital, C1:68, 141 86 Stockholm, Sweden
| |
Collapse
|
6
|
Johnson MT, Xin P, Benson JC, Pathak T, Walter V, Emrich SM, Yoast RE, Zhang X, Cao G, Panettieri RA, Trebak M. STIM1 is a core trigger of airway smooth muscle remodeling and hyperresponsiveness in asthma. Proc Natl Acad Sci U S A 2022; 119:e2114557118. [PMID: 34949717 PMCID: PMC8740694 DOI: 10.1073/pnas.2114557118] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/19/2021] [Indexed: 12/20/2022] Open
Abstract
Airway remodeling and airway hyperresponsiveness are central drivers of asthma severity. Airway remodeling is a structural change involving the dedifferentiation of airway smooth muscle (ASM) cells from a quiescent to a proliferative and secretory phenotype. Here, we show up-regulation of the endoplasmic reticulum Ca2+ sensor stromal-interacting molecule 1 (STIM1) in ASM of asthmatic mice. STIM1 is required for metabolic and transcriptional reprogramming that supports airway remodeling, including ASM proliferation, migration, secretion of cytokines and extracellular matrix, enhanced mitochondrial mass, and increased oxidative phosphorylation and glycolytic flux. Mechanistically, STIM1-mediated Ca2+ influx is critical for the activation of nuclear factor of activated T cells 4 and subsequent interleukin-6 secretion and transcription of pro-remodeling transcription factors, growth factors, surface receptors, and asthma-associated proteins. STIM1 drives airway hyperresponsiveness in asthmatic mice through enhanced frequency and amplitude of ASM cytosolic Ca2+ oscillations. Our data advocates for ASM STIM1 as a target for asthma therapy.
Collapse
Affiliation(s)
- Martin T Johnson
- Department of Cellular and Molecular Physiology, The Pennsylvania State University College of Medicine, Hershey, PA 17033
| | - Ping Xin
- Department of Cellular and Molecular Physiology, The Pennsylvania State University College of Medicine, Hershey, PA 17033
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15231
- Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA 15231
| | - J Cory Benson
- Department of Cellular and Molecular Physiology, The Pennsylvania State University College of Medicine, Hershey, PA 17033
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15231
- Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA 15231
| | - Trayambak Pathak
- Department of Cellular and Molecular Physiology, The Pennsylvania State University College of Medicine, Hershey, PA 17033
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15231
- Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA 15231
| | - Vonn Walter
- Department of Public Health Sciences, The Pennsylvania State University College of Medicine, Hershey, PA 17033
| | - Scott M Emrich
- Department of Cellular and Molecular Physiology, The Pennsylvania State University College of Medicine, Hershey, PA 17033
| | - Ryan E Yoast
- Department of Cellular and Molecular Physiology, The Pennsylvania State University College of Medicine, Hershey, PA 17033
| | - Xuexin Zhang
- Department of Cellular and Molecular Physiology, The Pennsylvania State University College of Medicine, Hershey, PA 17033
| | - Gaoyuan Cao
- Rutgers Institute for Translational Medicine and Science, New Brunswick, NJ 08901
| | - Reynold A Panettieri
- Rutgers Institute for Translational Medicine and Science, New Brunswick, NJ 08901
| | - Mohamed Trebak
- Department of Cellular and Molecular Physiology, The Pennsylvania State University College of Medicine, Hershey, PA 17033;
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15231
- Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA 15231
| |
Collapse
|
7
|
Wang YM, Xu WJ, Xiang LL, Ding M, Zhang JJ, Lu JY, Xie BJ, Gao YD. Store-operated Calcium Entry-associated Regulatory Factor Regulates Airway Inflammation and Airway Remodeling in Asthma Mice Models. Am J Physiol Lung Cell Mol Physiol 2021; 321:L533-L544. [PMID: 34231388 DOI: 10.1152/ajplung.00079.2020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Store-operated calcium entry (SOCE) is involved in the pathogenesis of airway inflammation and remodeling in asthma. Store-operated calcium entry-associated regulatory factor (SARAF) can down-regulate SOCE. OBJECTIVE We sought to investigate the role of SARAF in the regulation of airway inflammation and remodeling in asthma mice models, as well as in the functional regulation of human airway smooth muscle cells (hASMCs). METHODS Balb/c mice were sensitized and challenged with ovalbumin to establish the asthma mice models. Mice were transfected with lentivirus, which expressed the SARAF gene + GFP or the negative control gene + GFP. Airway resistance was measured with the animal pulmonary function system. Airway inflammation and remodeling were evaluated via histological staining. In vitro cultured hASMCs were transfected with scrambled small interfering RNA(siRNA) or SARAF-specific siRNA respecitvely. The proliferation, migration rate, hypertrophy and SOCE activity of hASMCs were examined with cell counting kit 8, wound healing test, bright field imaging and Ca2+ fluorescence imaging, respectively. SARAF expression was measured by quantitative real-time-PCR. RESULTS Asthma mice models showed decreased SARAF mRNA expression in the lungs. SARAF overexpression attenuated airway inflammation, resistance and also remodeling. Downregulation of SARAF expression with siRNA promoted the proliferation, migration, hypertrophy and SOCE activity in hASMCs. CONCLUSIONS SARAF plays a protective role against airway inflammation and remodeling in asthma mice models by blunting SOCE; SARAF may also be a functional regulating factor of hASMCs.
Collapse
Affiliation(s)
- Yi-Min Wang
- Department of Respiratory Medicine, Zhongnan Hospital of Wuhan University, China.,Department of Allergology, Zhongnan Hospital of Wuhan University, China
| | - Wen-Juan Xu
- Department of Respiratory Medicine, Zhongnan Hospital of Wuhan University, China
| | - Lin-Li Xiang
- Department of Respiratory Medicine, Zhongnan Hospital of Wuhan University, China
| | - Mei Ding
- Department of Allergology, Zhongnan Hospital of Wuhan University, China
| | - Jin-Jin Zhang
- Department of Allergology, Zhongnan Hospital of Wuhan University, China
| | - Jing-Ya Lu
- Department of Epidemiology and Biostatistics, Ministry of Education Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Bao-Juan Xie
- Department of Respiratory Medicine, Zhongnan Hospital of Wuhan University, China
| | - Ya-Dong Gao
- Department of Allergology, Zhongnan Hospital of Wuhan University, China
| |
Collapse
|
8
|
Shawer H, Norman K, Cheng CW, Foster R, Beech DJ, Bailey MA. ORAI1 Ca 2+ Channel as a Therapeutic Target in Pathological Vascular Remodelling. Front Cell Dev Biol 2021; 9:653812. [PMID: 33937254 PMCID: PMC8083964 DOI: 10.3389/fcell.2021.653812] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 03/08/2021] [Indexed: 12/21/2022] Open
Abstract
In the adult, vascular smooth muscle cells (VSMC) are normally physiologically quiescent, arranged circumferentially in one or more layers within blood vessel walls. Remodelling of native VSMC to a proliferative state for vascular development, adaptation or repair is driven by platelet-derived growth factor (PDGF). A key effector downstream of PDGF receptors is store-operated calcium entry (SOCE) mediated through the plasma membrane calcium ion channel, ORAI1, which is activated by the endoplasmic reticulum (ER) calcium store sensor, stromal interaction molecule-1 (STIM1). This SOCE was shown to play fundamental roles in the pathological remodelling of VSMC. Exciting transgenic lineage-tracing studies have revealed that the contribution of the phenotypically-modulated VSMC in atherosclerotic plaque formation is more significant than previously appreciated, and growing evidence supports the relevance of ORAI1 signalling in this pathologic remodelling. ORAI1 has also emerged as an attractive potential therapeutic target as it is accessible to extracellular compound inhibition. This is further supported by the progression of several ORAI1 inhibitors into clinical trials. Here we discuss the current knowledge of ORAI1-mediated signalling in pathologic vascular remodelling, particularly in the settings of atherosclerotic cardiovascular diseases (CVDs) and neointimal hyperplasia, and the recent developments in our understanding of the mechanisms by which ORAI1 coordinates VSMC phenotypic remodelling, through the activation of key transcription factor, nuclear factor of activated T-cell (NFAT). In addition, we discuss advances in therapeutic strategies aimed at the ORAI1 target.
Collapse
Affiliation(s)
- Heba Shawer
- School of Medicine, The Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, United Kingdom
| | - Katherine Norman
- School of Medicine, The Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, United Kingdom.,School of Chemistry, University of Leeds, Leeds, United Kingdom
| | - Chew W Cheng
- School of Medicine, The Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, United Kingdom
| | - Richard Foster
- School of Medicine, The Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, United Kingdom.,School of Chemistry, University of Leeds, Leeds, United Kingdom
| | - David J Beech
- School of Medicine, The Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, United Kingdom
| | - Marc A Bailey
- School of Medicine, The Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, United Kingdom
| |
Collapse
|
9
|
Šutovská M, Kocmálová M, Kazimierová I, Forsberg CIN, Jošková M, Adamkov M, Fraňová S. Effects of Inhalation of STIM-Orai Antagonist SKF 96365 on Ovalbumin-Induced Airway Remodeling in Guinea Pigs. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1335:87-101. [PMID: 33742420 DOI: 10.1007/5584_2021_633] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Airway remodeling (AR) consists of wall thickening and hyperreactivity. STIM (stromal interaction molecule) and Orai protein pathways mediate extracellular Ca2+ signals involved in AR. This study aims to define the effects on AR of the STIM-Orai antagonist SKF 96365 given by inhalation in three increasing doses in ovalbumin-induced AR. In the control group, the antiasthmatic budesonide and salbutamol were given in the same model. The airway structure was evaluated by histological and immunohistochemistry and reactivity by specific airway resistance, contraction strength of isolated airway smooth muscles, and mucociliary clearance expressed by ciliary beating frequency. The immuno-biochemical markers of chronic inflammation were evaluated by BioPlex and ELISA assays. The AR was mediated by inflammatory cytokines and growth factors. The findings show significant anti-remodeling effects of SKF 96365, which were associated with a decrease in airway hyperreactivity. The anti-remodeling effect of SKF 96365 was mediated via the suppression of IL-4, IL-5, and IL-13 synthesis, and IL-12-INF-γ-TGF-β pathway. The budesonide-related AR suppression had to do with a decrease in proinflammatory cytokines and an increase in the anti-inflammatory IL-10, with negligible influence on growth factors synthesis and mucous glands activity.
Collapse
Affiliation(s)
- Martina Šutovská
- Department of Pharmacology, Jessenius Faculty of Medicine, Comenius University, Mala Hora, Martin, Slovakia
| | - Michaela Kocmálová
- Department of Pharmacology, Jessenius Faculty of Medicine, Comenius University, Mala Hora, Martin, Slovakia. .,Martin's Biomedical Center (BioMed), Jessenius Faculty of Medicine, Comenius University, Martin, Slovakia.
| | - Ivana Kazimierová
- Department of Pharmacology, Jessenius Faculty of Medicine, Comenius University, Mala Hora, Martin, Slovakia.,Martin's Biomedical Center (BioMed), Jessenius Faculty of Medicine, Comenius University, Martin, Slovakia
| | | | - Marta Jošková
- Department of Pharmacology, Jessenius Faculty of Medicine, Comenius University, Mala Hora, Martin, Slovakia
| | - Marian Adamkov
- Institute of Histology and Embryology Jessenius Faculty of Medicine Comenius University, Martin, Slovakia
| | - Soňa Fraňová
- Department of Pharmacology, Jessenius Faculty of Medicine, Comenius University, Mala Hora, Martin, Slovakia
| |
Collapse
|
10
|
Barde PJ, Viswanadha S, Veeraraghavan S, Vakkalanka SV, Nair A. A first-in-human study to evaluate the safety, tolerability and pharmacokinetics of RP3128, an oral calcium release-activated calcium (CRAC) channel modulator in healthy volunteers. J Clin Pharm Ther 2020; 46:677-687. [PMID: 33314326 DOI: 10.1111/jcpt.13322] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 11/09/2020] [Accepted: 11/16/2020] [Indexed: 12/21/2022]
Abstract
WHAT IS KNOWN AND OBJECTIVE RP3128, a novel, orally available modulator of calcium released activated calcium (CRAC) channel, is being developed for the potential treatment of autoimmune and inflammatory diseases. RP3128 showed nano-molar potency and activity in a range of in vitro and in vivo models of inflammation. We report a first-in-human study investigating the safety, tolerability, pharmacokinetics (PK) and pharmacodynamics (PD) of RP3128 in healthy subjects. METHODS A randomized, double-blind, placebo-controlled trial of single (25, 50, 100, 200 and 400 mg) and multiple (7 days: 25, 100 and 400 mg once daily) doses of RP3128 were performed. Thirty-two and 24 subjects were randomized in the single ascending dose (SAD) and multiple ascending dose (MAD) parts, respectively. RESULTS AND DISCUSSION RP3128 was well tolerated, with no dose-limiting toxicity at single and multiple doses. Incidence of treatment emergent adverse events (TEAEs) did not increase with ascending RP3128 doses. No changes were seen in cognitive function and ECG parameters. RP3128 was rapidly absorbed. Elimination was slow with a half-life of more than 80 h. Exposures increased with increasing doses. Accumulation was seen on repeated dosing. PD response, as evidenced by lower plasma levels of tumour necrosis factor-alfa (TNFα) and interleukin-4 (IL-4), was seen when compared to pre-dose values or placebo. WHAT IS NEW AND CONCLUSION The safety, tolerability and PK/PD profile of RP3128 demonstrates its potential to be developed in inflammatory disorders and support further clinical development (ClinicalTrials.gov number: NCT02958982).
Collapse
Affiliation(s)
- Prajak J Barde
- Rhizen Pharmaceuticals SA, La Chaux-de-Fonds, Switzerland
| | | | | | | | - Ajit Nair
- Rhizen Pharmaceuticals SA, La Chaux-de-Fonds, Switzerland
| |
Collapse
|
11
|
Back to the future: re-establishing guinea pig in vivo asthma models. Clin Sci (Lond) 2020; 134:1219-1242. [PMID: 32501497 DOI: 10.1042/cs20200394] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 05/13/2020] [Accepted: 05/20/2020] [Indexed: 12/23/2022]
Abstract
Research using animal models of asthma is currently dominated by mouse models. This has been driven by the comprehensive knowledge on inflammatory and immune reactions in mice, as well as tools to produce genetically modified mice. Many of the identified therapeutic targets influencing airway hyper-responsiveness and inflammation in mouse models, have however been disappointing when tested clinically in asthma. It is therefore a great need for new animal models that more closely resemble human asthma. The guinea pig has for decades been used in asthma research and a comprehensive table of different protocols for asthma models is presented. The studies have primarily been focused on the pharmacological aspects of the disease, where the guinea pig undoubtedly is superior to mice. Further reasons are the anatomical and physiological similarities between human and guinea pig airways compared with that of the mouse, especially with respect to airway branching, neurophysiology, pulmonary circulation and smooth muscle distribution, as well as mast cell localization and mediator secretion. Lack of reagents and specific molecular tools to study inflammatory and immunological reactions in the guinea pig has however greatly diminished its use in asthma research. The aim in this position paper is to review and summarize what we know about different aspects of the use of guinea pig in vivo models for asthma research. The associated aim is to highlight the unmet needs that have to be addressed in the future.
Collapse
|
12
|
Bakowski D, Murray F, Parekh AB. Store-Operated Ca 2+ Channels: Mechanism, Function, Pharmacology, and Therapeutic Targets. Annu Rev Pharmacol Toxicol 2020; 61:629-654. [PMID: 32966177 DOI: 10.1146/annurev-pharmtox-031620-105135] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Calcium (Ca2+) release-activated Ca2+ (CRAC) channels are a major route for Ca2+ entry in eukaryotic cells. These channels are store operated, opening when the endoplasmic reticulum (ER) is depleted of Ca2+, and are composed of the ER Ca2+ sensor protein STIM and the pore-forming plasma membrane subunit Orai. Recent years have heralded major strides in our understanding of the structure, gating, and function of the channels. Loss-of-function and gain-of-function mutants combined with RNAi knockdown strategies have revealed important roles for the channel in numerous human diseases, making the channel a clinically relevant target. Drugs targeting the channels generally lack specificity or exhibit poor efficacy in animal models. However, the landscape is changing, and CRAC channel blockers are now entering clinical trials. Here, we describe the key molecular and biological features of CRAC channels, consider various diseases associated with aberrant channel activity, and discuss targeting of the channels from a therapeutic perspective.
Collapse
Affiliation(s)
| | - Fraser Murray
- Pandeia Therapeutics, Oxford OX4 4GP, United Kingdom
| | - Anant B Parekh
- Department of Physiology, Anatomy and Genetics, Oxford University, Oxford OX1 3PT, United Kingdom; , .,Current affiliation: National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina 27709, USA
| |
Collapse
|
13
|
Lin YP, Nelson C, Kramer H, Parekh AB. The Allergen Der p3 from House Dust Mite Stimulates Store-Operated Ca 2+ Channels and Mast Cell Migration through PAR4 Receptors. Mol Cell 2019; 70:228-241.e5. [PMID: 29677491 DOI: 10.1016/j.molcel.2018.03.025] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Revised: 02/05/2018] [Accepted: 03/22/2018] [Indexed: 11/17/2022]
Abstract
The house dust mite is the principal source of perennial aeroallergens in man. How these allergens activate innate and adaptive immunity is unclear, and therefore, there are no therapies targeting mite allergens. Here, we show that house dust mite extract activates store-operated Ca2+ channels, a common signaling module in numerous cell types in the lung. Activation of channel pore-forming Orai1 subunits by mite extract requires gating by STIM1 proteins. Although mite extract stimulates both protease-activated receptor type 2 (PAR2) and PAR4 receptors, Ca2+ influx is more tightly coupled to the PAR4 pathway. We identify a major role for the serine protease allergen Der p3 in stimulating Orai1 channels and show that a therapy involving sub-maximal inhibition of both Der p3 and Orai1 channels suppresses mast cell activation to house dust mite. Our results reveal Der p3 as an important aeroallergen that activates Ca2+ channels and suggest a therapeutic strategy for treating mite-induced asthma.
Collapse
Affiliation(s)
- Yu-Ping Lin
- Department of Physiology, Anatomy and Genetics, University of Oxford, Parks Road, Oxford OX1 3PT, UK
| | - Charmaine Nelson
- Department of Physiology, Anatomy and Genetics, University of Oxford, Parks Road, Oxford OX1 3PT, UK
| | - Holger Kramer
- Department of Physiology, Anatomy and Genetics, University of Oxford, Parks Road, Oxford OX1 3PT, UK
| | - Anant B Parekh
- Department of Physiology, Anatomy and Genetics, University of Oxford, Parks Road, Oxford OX1 3PT, UK.
| |
Collapse
|
14
|
CRAC channels as targets for drug discovery and development. Cell Calcium 2018; 74:147-159. [PMID: 30075400 DOI: 10.1016/j.ceca.2018.07.005] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 07/16/2018] [Accepted: 07/16/2018] [Indexed: 12/13/2022]
Abstract
Calcium release-activated calcium (CRAC) channels have been the target of drug discovery for many years. The identification of STIM and Orai proteins as key components of CRAC channels greatly facilitated this process because their co-expression in cell lines produced electrophysiological currents (ICRAC) much larger than those in native cells, making it easier to confirm and characterize the effects of modulatory compounds. A driving force in the quest for CRAC channel drugs has been the immunocompromised phenotype displayed by humans and mice with null or loss-of-function mutations in STIM1 or Orai1, suggesting that CRAC channel inhibitors could be useful therapeutics for autoimmune or inflammatory conditions. Emerging data also suggests that other therapeutic conditions may benefit from CRAC channel inhibition. However, only recently have CRAC channel inhibitors reached clinical trials. This review discusses the challenges associated with drug discovery and development on CRAC channels and the approaches employed to date, as well as the results, starting from initial high-throughput screens for CRAC channel modulators and progressing through target selection and justification, descriptions of pharmacological, safety and toxicological profiles of compounds, and finally the entry of CRAC channel inhibitors into clinical trials.
Collapse
|
15
|
Samanta K, Parekh AB. Store-operated Ca2+ channels in airway epithelial cell function and implications for asthma. Philos Trans R Soc Lond B Biol Sci 2017; 371:rstb.2015.0424. [PMID: 27377718 PMCID: PMC4938024 DOI: 10.1098/rstb.2015.0424] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/30/2016] [Indexed: 12/18/2022] Open
Abstract
The epithelial cells of the lung are at the interface of a host and its environment and are therefore directly exposed to the inhaled air-borne particles. Rather than serving as a simple physical barrier, airway epithelia detect allergens and other irritants and then help organize the subsequent immune response through release of a plethora of secreted signals. Many of these signals are generated in response to opening of store-operated Ca2+ channels in the plasma membrane. In this review, we describe the properties of airway store-operated channels and their role in regulating airway epithelial cell function. This article is part of the themed issue ‘Evolution brings Ca2+ and ATP together to control life and death’.
Collapse
Affiliation(s)
- Krishna Samanta
- Department of Physiology, Anatomy and Genetics, Sherrington Building, Parks Road, Oxford OX1 3PT, UK
| | - Anant B Parekh
- Department of Physiology, Anatomy and Genetics, Sherrington Building, Parks Road, Oxford OX1 3PT, UK
| |
Collapse
|