1
|
Liardo E, Pham AT, Ghilardi AF, Zhelay T, Szteyn K, Gandi NL, Ekkati A, Koerner S, Kozak JA, Sun L. Discovery of selective Orai channel blockers bearing an indazole or a pyrazole scaffold. Eur J Med Chem 2024; 278:116805. [PMID: 39232360 DOI: 10.1016/j.ejmech.2024.116805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 08/19/2024] [Accepted: 08/26/2024] [Indexed: 09/06/2024]
Abstract
The calcium release activated calcium (CRAC) channel is highly expressed in T lymphocytes and plays a critical role in regulating T cell proliferation and functions including activation of the transcription factor nuclear factor of activated T cells (NFAT), cytokine production and cytotoxicity. The CRAC channel consists of the Orai pore subunit and STIM (stromal interacting molecule) endoplasmic reticulum calcium sensor. Loss of CRAC channel mediated calcium signaling has been identified as an underlying cause of severe combined immunodeficiency (SCID), leading to drastically weakened immunity against infections. Gain-of-function mutations in Orai and STIM have been associated with tubular aggregated myopathy (TAM), a skeletal muscle disease. While a number of small molecules have shown activity in inhibiting the CRAC signaling pathway, the usefulness of those tool compounds is limited by their off-target activity against TRPM4 and TRPM7 ion channels, high lipophilicity, and a lack of understanding of their mechanism of action. We report structure-activity relationship (SAR) studies that resulted in the characterization of compound 4k [1-(cyclopropylmethyl)-N-(3-fluoropyridin-4-yl)-1H-indazole-3-carboxamie] as a fast onset, reversible, and selective CRAC channel blocker. 4k fully blocked the CRAC current (IC50: 4.9 μM) and the nuclear translocation of NFAT at 30 and 10 μM, respectively, without affecting the electrophysiological function of TRPM4 and TRPM7 channels. Computational modeling appears to support its direction binding to Orai proteins that form the transmembrane CRACchannel.
Collapse
Affiliation(s)
- Elisa Liardo
- Center for Drug Discovery and Translational Research, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA
| | - Anh-Tuan Pham
- Center for Drug Discovery and Translational Research, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA
| | - Amanda F Ghilardi
- Center for Drug Discovery and Translational Research, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA
| | - Tetyana Zhelay
- Department of Neuroscience, Cell Biology and Physiology, Boonshoft School of Medicine and College of Science and Mathematics, Wright State University, Dayton, OH, 45435, USA
| | - Kalina Szteyn
- Department of Neuroscience, Cell Biology and Physiology, Boonshoft School of Medicine and College of Science and Mathematics, Wright State University, Dayton, OH, 45435, USA
| | - Naga Lakshmi Gandi
- Department of Neuroscience, Cell Biology and Physiology, Boonshoft School of Medicine and College of Science and Mathematics, Wright State University, Dayton, OH, 45435, USA
| | - Anil Ekkati
- Center for Drug Discovery and Translational Research, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA
| | - Steffi Koerner
- Center for Drug Discovery and Translational Research, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA
| | - J Ashot Kozak
- Department of Neuroscience, Cell Biology and Physiology, Boonshoft School of Medicine and College of Science and Mathematics, Wright State University, Dayton, OH, 45435, USA.
| | - Lijun Sun
- Center for Drug Discovery and Translational Research, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA.
| |
Collapse
|
2
|
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
|
3
|
Ji R, Chang L, An C, Zhang J. Proton-sensing ion channels, GPCRs and calcium signaling regulated by them: implications for cancer. Front Cell Dev Biol 2024; 12:1326231. [PMID: 38505262 PMCID: PMC10949864 DOI: 10.3389/fcell.2024.1326231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 02/14/2024] [Indexed: 03/21/2024] Open
Abstract
Extracellular acidification of tumors is common. Through proton-sensing ion channels or proton-sensing G protein-coupled receptors (GPCRs), tumor cells sense extracellular acidification to stimulate a variety of intracellular signaling pathways including the calcium signaling, which consequently exerts global impacts on tumor cells. Proton-sensing ion channels, and proton-sensing GPCRs have natural advantages as drug targets of anticancer therapy. However, they and the calcium signaling regulated by them attracted limited attention as potential targets of anticancer drugs. In the present review, we discuss the progress in studies on proton-sensing ion channels, and proton-sensing GPCRs, especially emphasizing the effects of calcium signaling activated by them on the characteristics of tumors, including proliferation, migration, invasion, metastasis, drug resistance, angiogenesis. In addition, we review the drugs targeting proton-sensing channels or GPCRs that are currently in clinical trials, as well as the relevant potential drugs for cancer treatments, and discuss their future prospects. The present review aims to elucidate the important role of proton-sensing ion channels, GPCRs and calcium signaling regulated by them in cancer initiation and development. This review will promote the development of drugs targeting proton-sensing channels or GPCRs for cancer treatments, effectively taking their unique advantage as anti-cancer drug targets.
Collapse
Affiliation(s)
- Renhui Ji
- Foundational and Translational Medical Research Center, Department of Allergy and General Surgery, Hohhot First Hospital, Hohhot, China
- Department of Pathophysiology, Basic Medicine College of Inner Mongolia Medical University, Hohhot, China
| | - Li Chang
- Foundational and Translational Medical Research Center, Department of Allergy and General Surgery, Hohhot First Hospital, Hohhot, China
- Department of Pathophysiology, Basic Medicine College of Inner Mongolia Medical University, Hohhot, China
| | - Caiyan An
- Foundational and Translational Medical Research Center, Department of Allergy and General Surgery, Hohhot First Hospital, Hohhot, China
| | - Junjing Zhang
- Foundational and Translational Medical Research Center, Department of Allergy and General Surgery, Hohhot First Hospital, Hohhot, China
| |
Collapse
|
4
|
Norman K, Hemmings KE, Shawer H, Appleby HL, Burnett AJ, Hamzah N, Gosain R, Woodhouse EM, Beech DJ, Foster R, Bailey MA. Side-by-side comparison of published small molecule inhibitors against thapsigargin-induced store-operated Ca2+ entry in HEK293 cells. PLoS One 2024; 19:e0296065. [PMID: 38261554 PMCID: PMC10805320 DOI: 10.1371/journal.pone.0296065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 12/05/2023] [Indexed: 01/25/2024] Open
Abstract
Calcium (Ca2+) is a key second messenger in eukaryotes, with store-operated Ca2+ entry (SOCE) being the main source of Ca2+ influx into non-excitable cells. ORAI1 is a highly Ca2+-selective plasma membrane channel that encodes SOCE. It is ubiquitously expressed in mammals and has been implicated in numerous diseases, including cardiovascular disease and cancer. A number of small molecules have been identified as inhibitors of SOCE with a variety of potential therapeutic uses proposed and validated in vitro and in vivo. These encompass both nonselective Ca2+ channel inhibitors and targeted selective inhibitors of SOCE. Inhibition of SOCE can be quantified both directly and indirectly with a variety of assay setups, making an accurate comparison of the activity of different SOCE inhibitors challenging. We have used a fluorescence based Ca2+ addback assay in native HEK293 cells to generate dose-response data for many published SOCE inhibitors. We were able to directly compare potency. Most compounds were validated with only minor and expected variations in potency, but some were not. This could be due to differences in assay setup relating to the mechanism of action of the inhibitors and highlights the value of a singular approach to compare these compounds, as well as the general need for biorthogonal validation of novel bioactive compounds. The compounds observed to be the most potent against SOCE in our study were: 7-azaindole 14d (12), JPIII (17), Synta-66 (6), Pyr 3 (5), GSK5503A (8), CM4620 (14) and RO2959 (7). These represent the most promising candidates for future development of SOCE inhibitors for therapeutic use.
Collapse
Affiliation(s)
- Katherine Norman
- School of Chemistry, University of Leeds, Leeds, West Yorkshire, United Kingdom
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds, West Yorkshire, United Kingdom
| | - Karen E. Hemmings
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds, West Yorkshire, United Kingdom
| | - Heba Shawer
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds, West Yorkshire, United Kingdom
| | - Hollie L. Appleby
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds, West Yorkshire, United Kingdom
| | - Alan J. Burnett
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds, West Yorkshire, United Kingdom
| | - Nurasyikin Hamzah
- School of Chemistry, University of Leeds, Leeds, West Yorkshire, United Kingdom
| | - Rajendra Gosain
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds, West Yorkshire, United Kingdom
| | - Emily M. Woodhouse
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds, West Yorkshire, United Kingdom
| | - David J. Beech
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds, West Yorkshire, United Kingdom
| | - Richard Foster
- School of Chemistry, University of Leeds, Leeds, West Yorkshire, United Kingdom
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds, West Yorkshire, United Kingdom
| | - Marc A. Bailey
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds, West Yorkshire, United Kingdom
| |
Collapse
|
5
|
Tscherrig D, Bhardwaj R, Biner D, Dernič J, Ross-Kaschitza D, Peinelt C, Hediger MA, Lochner M. Development of chemical tools based on GSK-7975A to study store-operated calcium entry in cells. Cell Calcium 2024; 117:102834. [PMID: 38006628 DOI: 10.1016/j.ceca.2023.102834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 11/11/2023] [Accepted: 11/13/2023] [Indexed: 11/27/2023]
Abstract
Many physiological functions, such as cell differentiation, proliferation, muscle contraction, neurotransmission and fertilisation, are regulated by changes of Ca2+ levels. The major Ca2+ store in cells is the endoplasmic reticulum (ER). Certain cellular processes induce ER store depletion, e.g. by activating IP3 receptors, that in turn induces a store refilling process known as store-operated calcium entry (SOCE). This refilling process entails protein-protein interactions between Ca2+ sensing stromal interaction molecules (STIM) in the ER membrane and Orai proteins in the plasma membrane. Fully assembled STIM/Orai complexes then form highly selective Ca2+ channels called Ca2+ release-activated Ca2+ Channels (CRAC) through which Ca2+ ions flow into the cytosol and subsequently are pumped into the ER by the sarcoplasmic/endoplasmic reticulum calcium ATPase (SERCA). Abnormal SOCE has been associated with numerous human diseases and cancers, and therefore key players STIM and Orai have attracted significant therapeutic interest. Several potent experimental and clinical candidate compounds have been developed and have helped to study SOCE in various cell types. We have synthesized multiple novel small-molecule probes based on the known SOCE inhibitor GSK-7975A. Here we present GSK-7975A derivatives, which feature photo-caging, photo-crosslinking, biotin and clickable moieties, and also contain deuterium labels. Evaluation of these GSK-7975A probes using a fluorometric imaging plate reader (FLIPR)-Tetra-based Ca2+ imaging assay showed that most synthetic modifications did not have a detrimental impact on the SOCE inhibitory activity. The photo-caged GSK-7975A was also used in patch-clamp electrophysiology experiments. In summary, we have developed a number of active, GSK-7975A-based molecular probes that have interesting properties and therefore are useful experimental tools to study SOCE in various cells and settings.
Collapse
Affiliation(s)
- Dominic Tscherrig
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bühlstrasse 28, 3012 Bern, Switzerland
| | - Rajesh Bhardwaj
- Department of BioMedical Research, University of Bern and Department of Nephrology and Hypertension, Inselspital, Bern University Hospital, Freiburgstrasse 15, 3010 Bern, Switzerland.
| | - Daniel Biner
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Freiestrasse 3, 3012 Bern, Switzerland
| | - Jan Dernič
- Department of BioMedical Research, University of Bern and Department of Nephrology and Hypertension, Inselspital, Bern University Hospital, Freiburgstrasse 15, 3010 Bern, Switzerland
| | - Daniela Ross-Kaschitza
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bühlstrasse 28, 3012 Bern, Switzerland
| | - Christine Peinelt
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bühlstrasse 28, 3012 Bern, Switzerland
| | - Matthias A Hediger
- Department of BioMedical Research, University of Bern and Department of Nephrology and Hypertension, Inselspital, Bern University Hospital, Freiburgstrasse 15, 3010 Bern, Switzerland.
| | - Martin Lochner
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bühlstrasse 28, 3012 Bern, Switzerland.
| |
Collapse
|
6
|
Park JS, Perl A. Endosome Traffic Modulates Pro-Inflammatory Signal Transduction in CD4 + T Cells-Implications for the Pathogenesis of Systemic Lupus Erythematosus. Int J Mol Sci 2023; 24:10749. [PMID: 37445926 DOI: 10.3390/ijms241310749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 06/10/2023] [Accepted: 06/26/2023] [Indexed: 07/15/2023] Open
Abstract
Endocytic recycling regulates the cell surface receptor composition of the plasma membrane. The surface expression levels of the T cell receptor (TCR), in concert with signal transducing co-receptors, regulate T cell responses, such as proliferation, differentiation, and cytokine production. Altered TCR expression contributes to pro-inflammatory skewing, which is a hallmark of autoimmune diseases, such as systemic lupus erythematosus (SLE), defined by a reduced function of regulatory T cells (Tregs) and the expansion of CD4+ helper T (Th) cells. The ensuing secretion of inflammatory cytokines, such as interferon-γ and interleukin (IL)-4, IL-17, IL-21, and IL-23, trigger autoantibody production and tissue infiltration by cells of the adaptive and innate immune system that induce organ damage. Endocytic recycling influences immunological synapse formation by CD4+ T lymphocytes, signal transduction from crosslinked surface receptors through recruitment of adaptor molecules, intracellular traffic of organelles, and the generation of metabolites to support growth, cytokine production, and epigenetic control of DNA replication and gene expression in the cell nucleus. This review will delineate checkpoints of endosome traffic that can be targeted for therapeutic interventions in autoimmune and other disease conditions.
Collapse
Affiliation(s)
- Joy S Park
- Department of Medicine, Norton College of Medicine, State University of New York, Upstate Medical University, Syracuse, NY 13210, USA
- Department of Biochemistry and Molecular Biology, Norton College of Medicine, State University of New York, Upstate Medical University, Syracuse, NY 13210, USA
| | - Andras Perl
- Department of Medicine, Norton College of Medicine, State University of New York, Upstate Medical University, Syracuse, NY 13210, USA
- Department of Biochemistry and Molecular Biology, Norton College of Medicine, State University of New York, Upstate Medical University, Syracuse, NY 13210, USA
- Department of Microbiology and Immunology, Norton College of Medicine, State University of New York, Upstate Medical University, Syracuse, NY 13210, USA
| |
Collapse
|
7
|
Protasi F, Girolami B, Roccabianca S, Rossi D. Store-operated calcium entry: From physiology to tubular aggregate myopathy. Curr Opin Pharmacol 2023; 68:102347. [PMID: 36608411 DOI: 10.1016/j.coph.2022.102347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 11/30/2022] [Accepted: 12/04/2022] [Indexed: 01/06/2023]
Abstract
Store-Operated Ca2+ entry (SOCE) is recognized as a key mechanism in muscle physiology necessary to refill intracellular Ca2+ stores during sustained muscle activity. For many years the cell structures expected to mediate SOCE in skeletal muscle fibres remained unknown. Recently, the identification of Ca2+ Entry Units (CEUs) in exercised muscle fibres opened new insights into the role of extracellular Ca2+ in muscle contraction and, more generally, in intracellular Ca2+ homeostasis. Accordingly, intracellular Ca2+ unbalance due to alterations in SOCE strictly correlates with muscle disfunction and disease. Mutations in proteins involved in SOCE (STIM1, ORAI1, and CASQ1) have been linked to tubular aggregate myopathy (TAM), a disease that causes muscle weakness and myalgia and is characterized by a typical accumulation of highly ordered and packed membrane tubules originated from the sarcoplasmic reticulum (SR). Achieving a full understanding of the molecular pathways activated by alterations in Ca2+ entry mechanisms is a necessary step to design effective therapies for human SOCE-related disorders.
Collapse
Affiliation(s)
- Feliciano Protasi
- CAST, Center for Advanced Studies and Technology; University G. d'Annunzio of Chieti-Pescara, I-66100, Italy; DMSI, Department of Medicine and Aging Sciences; University G. d'Annunzio of Chieti-Pescara, I-66100, Italy
| | - Barbara Girolami
- CAST, Center for Advanced Studies and Technology; University G. d'Annunzio of Chieti-Pescara, I-66100, Italy; DMSI, Department of Medicine and Aging Sciences; University G. d'Annunzio of Chieti-Pescara, I-66100, Italy
| | - Sara Roccabianca
- DMMS, Department of Molecular and Developmental Medicine; University of Siena, I-53100, Siena Italy
| | - Daniela Rossi
- DMMS, Department of Molecular and Developmental Medicine; University of Siena, I-53100, Siena Italy.
| |
Collapse
|
8
|
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: 6] [Impact Index Per Article: 6.0] [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
|
9
|
The role of adipose tissue-derived hydrogen sulfide in inhibiting atherosclerosis. Nitric Oxide 2022; 127:18-25. [PMID: 35839994 DOI: 10.1016/j.niox.2022.07.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 07/08/2022] [Indexed: 02/06/2023]
Abstract
Hydrogen sulfide (H2S) is the third gaseous signaling molecule discovered in the body after NO and CO and plays an important organismal protective role in various diseases. Within adipose tissue, related catalytic enzymes (cystathionine-β-synthetase, cystathionine-γ-lyase, and 3-mercaptopyruvate transsulfuration enzyme) can produce and release endogenous H2S. Atherosclerosis (As) is a pathological change in arterial vessels that is closely related to abnormal glucose and lipid metabolism and a chronic inflammatory response. Previous studies have shown that H2S can act on the cardiovascular system, exerting effects such as improving disorders of glycolipid metabolism, alleviating insulin resistance, protecting the function of vascular endothelial cells, inhibiting vascular smooth muscle cell proliferation and migration, regulating vascular tone, inhibiting the inflammatory response, and antagonizing the occurrence and development of As.
Collapse
|
10
|
Souza Bomfim GH, Niemeyer BA, Lacruz RS, Lis A. On the Connections between TRPM Channels and SOCE. Cells 2022; 11:1190. [PMID: 35406753 PMCID: PMC8997886 DOI: 10.3390/cells11071190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 03/23/2022] [Accepted: 03/30/2022] [Indexed: 12/02/2022] Open
Abstract
Plasma membrane protein channels provide a passageway for ions to access the intracellular milieu. Rapid entry of calcium ions into cells is controlled mostly by ion channels, while Ca2+-ATPases and Ca2+ exchangers ensure that cytosolic Ca2+ levels ([Ca2+]cyt) are maintained at low (~100 nM) concentrations. Some channels, such as the Ca2+-release-activated Ca2+ (CRAC) channels and voltage-dependent Ca2+ channels (CACNAs), are highly Ca2+-selective, while others, including the Transient Receptor Potential Melastatin (TRPM) family, have broader selectivity and are mostly permeable to monovalent and divalent cations. Activation of CRAC channels involves the coupling between ORAI1-3 channels with the endoplasmic reticulum (ER) located Ca2+ store sensor, Stromal Interaction Molecules 1-2 (STIM1/2), a pathway also termed store-operated Ca2+ entry (SOCE). The TRPM family is formed by 8 members (TRPM1-8) permeable to Mg2+, Ca2+, Zn2+ and Na+ cations, and is activated by multiple stimuli. Recent studies indicated that SOCE and TRPM structure-function are interlinked in some instances, although the molecular details of this interaction are only emerging. Here we review the role of TRPM and SOCE in Ca2+ handling and highlight the available evidence for this interaction.
Collapse
Affiliation(s)
- Guilherme H. Souza Bomfim
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY 10010, USA;
| | - Barbara A. Niemeyer
- Department of Molecular Biophysics, Center for Integrative Physiology and Molecular Medicine, School of Medicine, Saarland University, 66421 Homburg, Germany;
| | - Rodrigo S. Lacruz
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY 10010, USA;
| | - Annette Lis
- Department of Biophysics, Center for Integrative Physiology and Molecular Medicine, School of Medicine, Saarland University, 66421 Homburg, Germany
| |
Collapse
|
11
|
Cañas CA, Castaño-Valencia S, Castro-Herrera F. Pharmacological blockade of KV1.3 channel as a promising treatment in autoimmune diseases. J Transl Autoimmun 2022; 5:100146. [PMID: 35146402 PMCID: PMC8818563 DOI: 10.1016/j.jtauto.2022.100146] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 01/20/2022] [Accepted: 01/22/2022] [Indexed: 11/27/2022] Open
Abstract
There are more than 100 autoimmune diseases (AD), which have a high prevalence that ranges between 5% and 8% of the general population. Type I diabetes mellitus, multiple sclerosis, systemic lupus erythematosus and rheumatoid arthritis remain the health problem of highest concern among people worldwide due to its high morbidity and mortality. The development of new treatment strategies has become a research hotspot. In recent years, the study of the ion channels presents in the cells of the immune system, regarding their functional role, the consequences of mutations in their genes and the different ways of blocking them are the subject of intense research. Pharmacological blockade of KV1.3 channel inhibits Ca2+ signaling, T cell proliferation, and pro-inflammatory interleukins production in human CD4+ effector memory T cells. These cells mediated most of the AD and their inhibition is a promising therapeutic target. In this review, we will highlight the biological function of KV1.3 channel in T cells, consequence of the pharmacological inhibition (through anemone and scorpion toxins, synthetic peptides, nanoparticles, or monoclonal antibodies) as well as the possible therapeutical application in AD.
Collapse
Affiliation(s)
- Carlos A. Cañas
- Universidad Icesi, CIRAT, Centro de Investigación en Reumatología, Autoinmunidad y Medicina Traslacional, Cali, Colombia
- Unit of Rheumatology, Fundación Valle del Lili, Cali, Colombia
- Corresponding author. Rheumatology Unit, Fundación Valle del Lili, Cra. 98 18-49, Cali, 760032, Colombia.
| | - Santiago Castaño-Valencia
- Department of Physiological Sciences, Department of Health Sciences, Universidad del Valle, Cali, Colombia
| | - Fernando Castro-Herrera
- Department of Physiological Sciences, Department of Health Sciences, Universidad del Valle, Cali, Colombia
| |
Collapse
|
12
|
Li Y, Yang X, Shen Y. Structural Insights into Ca 2+ Permeation through Orai Channels. Cells 2021; 10:cells10113062. [PMID: 34831285 PMCID: PMC8619096 DOI: 10.3390/cells10113062] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 11/03/2021] [Accepted: 11/04/2021] [Indexed: 11/16/2022] Open
Abstract
Orai channels belong to the calcium release-activated calcium (CRAC) channel family. Orai channels are responsible for the influx of extracellular Ca2+ that is triggered by Ca2+ depletion from the endoplasmic reticulum (ER); this function is essential for many types of non-excitable cells. Extensive structural and functional studies have advanced the knowledge of the molecular mechanism by which Orai channels are activated. However, the gating mechanism that allows Ca2+ permeation through Orai channels is less well explained. Here, we reviewed and summarized the existing structural studies of Orai channels. We detailed the structural features of Orai channels, described structural comparisons of their closed and open states, and finally proposed a "push-pull" model of Ca2+ permeation.
Collapse
|
13
|
Zhu X, Zhan Y, Gu Y, Huang Q, Wang T, Deng Z, Xie J. Cigarette Smoke Promotes Interleukin-8 Production in Alveolar Macrophages Through the Reactive Oxygen Species/Stromal Interaction Molecule 1/Ca 2+ Axis. Front Physiol 2021; 12:733650. [PMID: 34690806 PMCID: PMC8531208 DOI: 10.3389/fphys.2021.733650] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 09/22/2021] [Indexed: 11/13/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD), primarily attributed to cigarette smoke (CS), is characterized by multiple pathophysiological changes, including oxidative stress and inflammation. Stromal interaction molecule 1 (STIM1) is a Ca2+ sensor that regulates Ca2+ entry in different types of cells. The present study aimed to explore the relationship between CS-induced oxidative stress and inflammation, as well as the functional role of STIM1 thereinto. Our results showed that the reactive oxygen species (ROS)/STIM1/Ca2+ axis played a critical role in CS-induced secretion of interleukin (IL)-8 in human alveolar macrophages. Specifically, smokers with COPD (SC) showed higher levels of ROS in the lung tissues compared with healthy non-smokers (HN). STIM1 was upregulated in the lung tissues of COPD patients. The expression of STIM1 was positively associated with ROS levels and negatively correlated with pulmonary function. The expression of STIM1 was also increased in the bronchoalveolar lavage fluid (BALF) macrophages of COPD patients and PMA-differentiated THP-1 macrophages stimulated by cigarette smoke extract (CSE). Additionally, CSE-induced upregulation of STIM1 in PMA-differentiated THP-1 macrophages was inhibited by pretreatment with N-acetylcysteine (NAC), a ROS scavenger. Transfection with small interfering RNA (siRNA) targeting STIM1 and pretreatment with NAC alleviated CSE-induced increase in intracellular Ca2+ levels and IL-8 expression. Furthermore, pretreatment with SKF-96365 and 2-APB, the inhibitors of Ca2+ influx, suppressed CSE-induced secretion of IL-8. In conclusion, our study demonstrates that CSE-induced ROS production may increase the expression of STIM1 in macrophages, which further promotes the release of IL-8 by regulating Ca2+ entry. These data suggest that STIM1 may play a crucial role in CSE-induced ROS production and inflammation, and participate in the pathogenesis of COPD.
Collapse
Affiliation(s)
- Xianying Zhu
- Department of Respiratory and Critical Care Medicine, National Clinical Research Center for Respiratory Disease, Key Laboratory of Pulmonary Diseases of Health Ministry, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Department of Intensive Care Unit, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Yuan Zhan
- Department of Respiratory and Critical Care Medicine, National Clinical Research Center for Respiratory Disease, Key Laboratory of Pulmonary Diseases of Health Ministry, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yiya Gu
- Department of Respiratory and Critical Care Medicine, National Clinical Research Center for Respiratory Disease, Key Laboratory of Pulmonary Diseases of Health Ministry, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qian Huang
- Department of Respiratory and Critical Care Medicine, National Clinical Research Center for Respiratory Disease, Key Laboratory of Pulmonary Diseases of Health Ministry, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ting Wang
- Department of Respiratory and Critical Care Medicine, National Clinical Research Center for Respiratory Disease, Key Laboratory of Pulmonary Diseases of Health Ministry, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhesong Deng
- Department of Respiratory and Critical Care Medicine, National Clinical Research Center for Respiratory Disease, Key Laboratory of Pulmonary Diseases of Health Ministry, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jungang Xie
- Department of Respiratory and Critical Care Medicine, National Clinical Research Center for Respiratory Disease, Key Laboratory of Pulmonary Diseases of Health Ministry, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
14
|
Tiffner A, Derler I. Isoform-Specific Properties of Orai Homologues in Activation, Downstream Signaling, Physiology and Pathophysiology. Int J Mol Sci 2021; 22:8020. [PMID: 34360783 PMCID: PMC8347056 DOI: 10.3390/ijms22158020] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/21/2021] [Accepted: 07/23/2021] [Indexed: 11/21/2022] Open
Abstract
Ca2+ ion channels are critical in a variety of physiological events, including cell growth, differentiation, gene transcription and apoptosis. One such essential entry pathway for calcium into the cell is the Ca2+ release-activated Ca2+ (CRAC) channel. It consists of the Ca2+ sensing protein, stromal interaction molecule 1 (STIM1) located in the endoplasmic reticulum (ER) and a Ca2+ ion channel Orai in the plasma membrane. The Orai channel family includes three homologues Orai1, Orai2 and Orai3. While Orai1 is the "classical" Ca2+ ion channel within the CRAC channel complex and plays a universal role in the human body, there is increasing evidence that Orai2 and Orai3 are important in specific physiological and pathophysiological processes. This makes them an attractive target in drug discovery, but requires a detailed understanding of the three Orai channels and, in particular, their differences. Orai channel activation is initiated via Ca2+ store depletion, which is sensed by STIM1 proteins, and induces their conformational change and oligomerization. Upon STIM1 coupling, Orai channels activate to allow Ca2+ permeation into the cell. While this activation mechanism is comparable among the isoforms, they differ by a number of functional and structural properties due to non-conserved regions in their sequences. In this review, we summarize the knowledge as well as open questions in our current understanding of the three isoforms in terms of their structure/function relationship, downstream signaling and physiology as well as pathophysiology.
Collapse
Affiliation(s)
| | - Isabella Derler
- Institute of Biophysics, JKU Life Science Center, Johannes Kepler University Linz, A-4020 Linz, Austria;
| |
Collapse
|
15
|
Liang X, Zhang N, Pan H, Xie J, Han W. Development of Store-Operated Calcium Entry-Targeted Compounds in Cancer. Front Pharmacol 2021; 12:688244. [PMID: 34122115 PMCID: PMC8194303 DOI: 10.3389/fphar.2021.688244] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 05/17/2021] [Indexed: 11/17/2022] Open
Abstract
Store-operated Ca2+ entry (SOCE) is the major pathway of Ca2+ entry in mammalian cells, and regulates a variety of cellular functions including proliferation, motility, apoptosis, and death. Accumulating evidence has indicated that augmented SOCE is related to the generation and development of cancer, including tumor formation, proliferation, angiogenesis, metastasis, and antitumor immunity. Therefore, the development of compounds targeting SOCE has been proposed as a potential and effective strategy for use in cancer therapy. In this review, we summarize the current research on SOCE inhibitors and blockers, discuss their effects and possible mechanisms of action in cancer therapy, and induce a new perspective on the treatment of cancer.
Collapse
Affiliation(s)
- Xiaojing Liang
- Department of Medical Oncology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Ningxia Zhang
- Department of Medical Oncology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Hongming Pan
- Department of Medical Oncology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Jiansheng Xie
- Department of Medical Oncology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, China.,Laboratory of Cancer Biology, Institute of Clinical Science, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Weidong Han
- Department of Medical Oncology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
16
|
Chang Y, Roy S, Pan Z. Store-Operated Calcium Channels as Drug Target in Gastroesophageal Cancers. Front Pharmacol 2021; 12:668730. [PMID: 34012400 PMCID: PMC8126661 DOI: 10.3389/fphar.2021.668730] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 04/12/2021] [Indexed: 12/24/2022] Open
Abstract
Gastroesophageal cancers, including tumors occurring in esophagus and stomach, usually have poor prognosis and lack effective chemotherapeutic drugs for treatment. The association between dysregulated store-operated calcium entry (SOCE), a key intracellular Ca2+ signaling pathway and gastroesophageal cancers are emerging. This review summarizes the recent advances in understanding the contribution of SOCE-mediated intracellular Ca2+ signaling to gastroesophageal cancers. It assesses the pathophysiological role of each component in SOCE machinery, such as Orais and STIMs in the cancer cell proliferation, migration, and invasion as well as stemness maintenance. Lastly, it discusses efforts towards development of more specific and potent SOCE inhibitors, which may be a new set of chemotherapeutic drugs appearing at the horizon, to provide either targeted therapy or adjuvant treatment to overcome drug resistance for gastroesophageal cancers.
Collapse
Affiliation(s)
- Yan Chang
- College of Nursing and Health Innovation, The University of Texas at Arlington, Arlington, TX, United States
| | - Souvik Roy
- Department of Mathematics, The University of Texas at Arlington, Arlington, TX, United States
| | - Zui Pan
- College of Nursing and Health Innovation, The University of Texas at Arlington, Arlington, TX, United States
| |
Collapse
|
17
|
Aprile S, Riva B, Bhela IP, Cordero-Sanchez C, Avino G, Genazzani AA, Serafini M, Pirali T. 1,2,4-Oxadiazole-Bearing Pyrazoles as Metabolically Stable Modulators of Store-Operated Calcium Entry. ACS Med Chem Lett 2021; 12:640-646. [PMID: 33854704 PMCID: PMC8040252 DOI: 10.1021/acsmedchemlett.1c00034] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 03/08/2021] [Indexed: 11/28/2022] Open
Abstract
![]()
Store-operated calcium
entry (SOCE) is a pivotal mechanism in calcium
homeostasis, and, despite still being under investigation, its dysregulation
is known to be associated with severe human disorders. SOCE modulators
are therefore needed both as chemical probes and as therapeutic agents.
While many small molecules have been described so far, their poor
properties in terms of drug-likeness have limited their translation
into the clinical practice. In this work, we describe the bioisosteric
replacement of the ester moiety in pyrazole derivatives with a 1,2,4-oxadiazole
ring as a means to afford a class of modulators with high metabolic
stability. Moreover, among our derivatives, a compound able to increase
the calcium entry was identified, further enriching the library of
available SOCE activators.
Collapse
Affiliation(s)
- Silvio Aprile
- Department of Pharmaceutical Sciences, Università degli Studi del Piemonte Orientale, Largo Donegani 2, 28100 Novara, Italy
| | - Beatrice Riva
- Department of Pharmaceutical Sciences, Università degli Studi del Piemonte Orientale, Largo Donegani 2, 28100 Novara, Italy
- ChemICare S.r.l., Enne3, Corso Trieste 15/A, 28100 Novara, Italy
| | - Irene Preet Bhela
- Department of Pharmaceutical Sciences, Università degli Studi del Piemonte Orientale, Largo Donegani 2, 28100 Novara, Italy
| | - Celia Cordero-Sanchez
- Department of Pharmaceutical Sciences, Università degli Studi del Piemonte Orientale, Largo Donegani 2, 28100 Novara, Italy
| | - Giulia Avino
- Department of Pharmaceutical Sciences, Università degli Studi di Trieste, Via Giorgieri 1, 34127 Trieste, Italy
| | - Armando A. Genazzani
- Department of Pharmaceutical Sciences, Università degli Studi del Piemonte Orientale, Largo Donegani 2, 28100 Novara, Italy
| | - Marta Serafini
- Department of Pharmaceutical Sciences, Università degli Studi del Piemonte Orientale, Largo Donegani 2, 28100 Novara, Italy
| | - Tracey Pirali
- Department of Pharmaceutical Sciences, Università degli Studi del Piemonte Orientale, Largo Donegani 2, 28100 Novara, Italy
- ChemICare S.r.l., Enne3, Corso Trieste 15/A, 28100 Novara, Italy
| |
Collapse
|
18
|
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: 7.0] [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
|
19
|
Tiffner A, Derler I. Molecular Choreography and Structure of Ca 2+ Release-Activated Ca 2+ (CRAC) and K Ca2+ Channels and Their Relevance in Disease with Special Focus on Cancer. MEMBRANES 2020; 10:E425. [PMID: 33333945 PMCID: PMC7765462 DOI: 10.3390/membranes10120425] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 12/04/2020] [Accepted: 12/07/2020] [Indexed: 12/16/2022]
Abstract
Ca2+ ions play a variety of roles in the human body as well as within a single cell. Cellular Ca2+ signal transduction processes are governed by Ca2+ sensing and Ca2+ transporting proteins. In this review, we discuss the Ca2+ and the Ca2+-sensing ion channels with particular focus on the structure-function relationship of the Ca2+ release-activated Ca2+ (CRAC) ion channel, the Ca2+-activated K+ (KCa2+) ion channels, and their modulation via other cellular components. Moreover, we highlight their roles in healthy signaling processes as well as in disease with a special focus on cancer. As KCa2+ channels are activated via elevations of intracellular Ca2+ levels, we summarize the current knowledge on the action mechanisms of the interplay of CRAC and KCa2+ ion channels and their role in cancer cell development.
Collapse
Affiliation(s)
| | - Isabella Derler
- Institute of Biophysics, JKU Life Science Center, Johannes Kepler University Linz, A-4020 Linz, Austria;
| |
Collapse
|
20
|
Zhang I, Hu H. Store-Operated Calcium Channels in Physiological and Pathological States of the Nervous System. Front Cell Neurosci 2020; 14:600758. [PMID: 33328896 PMCID: PMC7732603 DOI: 10.3389/fncel.2020.600758] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 11/03/2020] [Indexed: 12/13/2022] Open
Abstract
Store-operated calcium channels (SOCs) are widely expressed in excitatory and non-excitatory cells where they mediate significant store-operated calcium entry (SOCE), an important pathway for calcium signaling throughout the body. While the activity of SOCs has been well studied in non-excitable cells, attention has turned to their role in neurons and glia in recent years. In particular, the role of SOCs in the nervous system has been extensively investigated, with links to their dysregulation found in a wide variety of neurological diseases from Alzheimer’s disease (AD) to pain. In this review, we provide an overview of their molecular components, expression, and physiological role in the nervous system and describe how the dysregulation of those roles could potentially lead to various neurological disorders. Although further studies are still needed to understand how SOCs are activated under physiological conditions and how they are linked to pathological states, growing evidence indicates that SOCs are important players in neurological disorders and could be potential new targets for therapies. While the role of SOCE in the nervous system continues to be multifaceted and controversial, the study of SOCs provides a potentially fruitful avenue into better understanding the nervous system and its pathologies.
Collapse
Affiliation(s)
- Isis Zhang
- Department of Anesthesiology, Rutgers New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, United States
| | - Huijuan Hu
- Department of Anesthesiology, Rutgers New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, United States
| |
Collapse
|
21
|
Targeting Calcium Release-activated Calcium Channel Is Not Sufficient to Prevent Rejection in Nonhuman Primate Kidney Transplantation. Transplantation 2020; 104:970-980. [PMID: 32317615 DOI: 10.1097/tp.0000000000003078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Calcineurin inhibitors successfully control rejection of transplanted organs but also cause nephrotoxicity. This study, using a rhesus monkey renal transplantation model, sought to determine the applicability of a new immunomodulatory drug inhibiting the store-operated calcium release-activated calcium channel of lymphocytes to control transplant rejection without nephrotoxicity. METHODS Animals underwent kidney transplantation and were treated with tacrolimus alone (n = 3), a CRACM1 inhibitor (PRCL-02) (n = 6) alone, or with initial tacrolimus monotherapy followed by gradual conversion at 3 weeks to PRCL-02 alone (n = 3). PRCL-02 was administered via a surgically inserted gastrostomy tube BID. RESULTS Dose-related drug exposure in monkeys was established and renal transplants were then performed using PRCL-02 monotherapy. Oral dosing of PRCL-02 was well tolerated and resulted in suppressed T-cell proliferation in in vitro MLR comparable to animals in the tacrolimus control arm. Animals receiving tacrolimus monotherapy were e on day 100 without rejection. PRCL-02 monotherapy only marginally prolonged graft survival (MST = 13.16 d; group 2) compared with untreated controls. Animals treated initially with tacrolimus and converted to PRCL-02 monotherapy had a mean graft survival of 35.3 days which was prolonged compared with PRCL-02 monotherapy but not compared with the tacrolimus-treated group. Pharmacokinetic studies showed inconsistent drug exposures despite attempts to adjust dose and exposure which may have contributed to the rejections. CONCLUSIONS We conclude that, in this nonhuman primate model of kidney transplantation, PRCL-02 demonstrated evidence of in vivo immunosuppressive activity but was inferior to tacrolimus treatment with respect to suppressing immune transplant rejection.
Collapse
|
22
|
Waldherr L, Tiffner A, Mishra D, Sallinger M, Schober R, Frischauf I, Schmidt T, Handl V, Sagmeister P, Köckinger M, Derler I, Üçal M, Bonhenry D, Patz S, Schindl R. Blockage of Store-Operated Ca 2+ Influx by Synta66 is Mediated by Direct Inhibition of the Ca 2+ Selective Orai1 Pore. Cancers (Basel) 2020; 12:E2876. [PMID: 33036292 PMCID: PMC7600887 DOI: 10.3390/cancers12102876] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 09/29/2020] [Accepted: 09/30/2020] [Indexed: 12/14/2022] Open
Abstract
The Ca2+ sensor STIM1 and the Ca2+ channel Orai1 that form the store-operated Ca2+ (SOC) channel complex are key targets for drug development. Selective SOC inhibitors are currently undergoing clinical evaluation for the treatment of auto-immune and inflammatory responses and are also deemed promising anti-neoplastic agents since SOC channels are linked with enhanced cancer cell progression. Here, we describe an investigation of the site of binding of the selective inhibitor Synta66 to the SOC channel Orai1 using docking and molecular dynamics simulations, and live cell recordings. Synta66 binding was localized to the extracellular site close to the transmembrane (TM)1 and TM3 helices and the extracellular loop segments, which, importantly, are adjacent to the Orai1-selectivity filter. Synta66-sensitivity of the Orai1 pore was, in fact, diminished by both Orai1 mutations affecting Ca2+ selectivity and permeation of Na+ in the absence of Ca2+. Synta66 also efficiently blocked SOC in three glioblastoma cell lines but failed to interfere with cell viability, division and migration. These experiments provide new structural and functional insights into selective drug inhibition of the Orai1 Ca2+ channel by a high-affinity pore blocker.
Collapse
Affiliation(s)
- Linda Waldherr
- Gottfried Schatz Research Centre, Medical University of Graz, A-8010 Graz, Austria; (L.W.); (R.S.); (T.S.)
| | - Adela Tiffner
- Institute of Biophysics, JKU Life Science Centre, Johannes Kepler University Linz, A-4020 Linz, Austria; (A.T.); (M.S.); (I.F.); (I.D.)
| | - Deepti Mishra
- Centre for Nanobiology and Structural Biology, Academy of Sciences of the Czech Republic, 373 33 Nové Hrady, Czech Republic;
| | - Matthias Sallinger
- Institute of Biophysics, JKU Life Science Centre, Johannes Kepler University Linz, A-4020 Linz, Austria; (A.T.); (M.S.); (I.F.); (I.D.)
| | - Romana Schober
- Gottfried Schatz Research Centre, Medical University of Graz, A-8010 Graz, Austria; (L.W.); (R.S.); (T.S.)
- Institute of Biophysics, JKU Life Science Centre, Johannes Kepler University Linz, A-4020 Linz, Austria; (A.T.); (M.S.); (I.F.); (I.D.)
| | - Irene Frischauf
- Institute of Biophysics, JKU Life Science Centre, Johannes Kepler University Linz, A-4020 Linz, Austria; (A.T.); (M.S.); (I.F.); (I.D.)
| | - Tony Schmidt
- Gottfried Schatz Research Centre, Medical University of Graz, A-8010 Graz, Austria; (L.W.); (R.S.); (T.S.)
| | - Verena Handl
- Department of Neurosurgery, Medical University of Graz, A-8010 Graz, Austria; (V.H.); (M.Ü.)
| | - Peter Sagmeister
- Institute of Chemistry, University of Graz, Heinrichstraße 28, A-8010 Graz, Austria; (P.S.); (M.K.)
| | - Manuel Köckinger
- Institute of Chemistry, University of Graz, Heinrichstraße 28, A-8010 Graz, Austria; (P.S.); (M.K.)
| | - Isabella Derler
- Institute of Biophysics, JKU Life Science Centre, Johannes Kepler University Linz, A-4020 Linz, Austria; (A.T.); (M.S.); (I.F.); (I.D.)
| | - Muammer Üçal
- Department of Neurosurgery, Medical University of Graz, A-8010 Graz, Austria; (V.H.); (M.Ü.)
| | - Daniel Bonhenry
- Centre for Nanobiology and Structural Biology, Academy of Sciences of the Czech Republic, 373 33 Nové Hrady, Czech Republic;
| | - Silke Patz
- Department of Neurosurgery, Medical University of Graz, A-8010 Graz, Austria; (V.H.); (M.Ü.)
| | - Rainer Schindl
- Gottfried Schatz Research Centre, Medical University of Graz, A-8010 Graz, Austria; (L.W.); (R.S.); (T.S.)
- Institute of Biophysics, JKU Life Science Centre, Johannes Kepler University Linz, A-4020 Linz, Austria; (A.T.); (M.S.); (I.F.); (I.D.)
| |
Collapse
|
23
|
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: 9.0] [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
|
24
|
Synthesis and Pharmacological Characterization of 2-Aminoethyl Diphenylborinate (2-APB) Derivatives for Inhibition of Store-Operated Calcium Entry (SOCE) in MDA-MB-231 Breast Cancer Cells. Int J Mol Sci 2020; 21:ijms21165604. [PMID: 32764353 PMCID: PMC7460636 DOI: 10.3390/ijms21165604] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 07/31/2020] [Accepted: 08/03/2020] [Indexed: 12/15/2022] Open
Abstract
Calcium ions regulate a wide array of physiological functions including cell differentiation, proliferation, muscle contraction, neurotransmission, and fertilization. The endoplasmic reticulum (ER) is the major intracellular Ca2+ store and cellular events that induce ER store depletion (e.g., activation of inositol 1,4,5-triphosphate (IP3) receptors) trigger a refilling process known as store-operated calcium entry (SOCE). It requires the intricate interaction between the Ca2+ sensing stromal interaction molecules (STIM) located in the ER membrane and the channel forming Orai proteins in the plasma membrane (PM). The resulting active STIM/Orai complexes form highly selective Ca2+ channels that facilitate a measurable Ca2+ influx into the cytosol followed by successive refilling of the ER by the sarcoplasmic/endoplasmic reticulum calcium ATPase (SERCA). STIM and Orai have attracted significant therapeutic interest, as enhanced SOCE has been associated with several cancers, and mutations in STIM and Orai have been linked to immunodeficiency, autoimmune, and muscular diseases. 2-Aminoethyl diphenylborinate (2-APB) is a known modulator and depending on its concentration can inhibit or enhance SOCE. We have synthesized several novel derivatives of 2-APB, introducing halogen and other small substituents systematically on each position of one of the phenyl rings. Using a fluorometric imaging plate reader (FLIPR) Tetra-based calcium imaging assay we have studied how these structural changes of 2-APB affect the SOCE modulation activity at different compound concentrations in MDA-MB-231 breast cancer cells. We have discovered 2-APB derivatives that block SOCE at low concentrations, at which 2-APB usually enhances SOCE.
Collapse
|
25
|
Tajada S, Villalobos C. Calcium Permeable Channels in Cancer Hallmarks. Front Pharmacol 2020; 11:968. [PMID: 32733237 PMCID: PMC7358640 DOI: 10.3389/fphar.2020.00968] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 06/15/2020] [Indexed: 12/17/2022] Open
Abstract
Cancer, the second cause of death worldwide, is characterized by several common criteria, known as the “cancer hallmarks” such as unrestrained cell proliferation, cell death resistance, angiogenesis, invasion and metastasis. Calcium permeable channels are proteins present in external and internal biological membranes, diffusing Ca2+ ions down their electrochemical gradient. Numerous physiological functions are mediated by calcium channels, ranging from intracellular calcium homeostasis to sensory transduction. Consequently, calcium channels play important roles in human physiology and it is not a surprise the increasing number of evidences connecting calcium channels disorders with tumor cells growth, survival and migration. Multiple studies suggest that calcium signals are augmented in various cancer cell types, contributing to cancer hallmarks. This review focuses in the role of calcium permeable channels signaling in cancer with special attention to the mechanisms behind the remodeling of the calcium signals. Transient Receptor Potential (TRP) channels and Store Operated Channels (SOC) are the main extracellular Ca2+ source in the plasma membrane of non-excitable cells, while inositol trisphosphate receptors (IP3R) are the main channels releasing Ca2+ from the endoplasmic reticulum (ER). Alterations in the function and/or expression of these calcium channels, as wells as, the calcium buffering by mitochondria affect intracellular calcium homeostasis and signaling, contributing to the transformation of normal cells into their tumor counterparts. Several compounds reported to counteract several cancer hallmarks also modulate the activity and/or the expression of these channels including non-steroidal anti-inflammatory drugs (NSAIDs) like sulindac and aspirin, and inhibitors of polyamine biosynthesis, like difluoromethylornithine (DFMO). The possible role of the calcium permeable channels targeted by these compounds in cancer and their action mechanism will be discussed also in the review.
Collapse
Affiliation(s)
- Sendoa Tajada
- Instituto de Biología y Genética Molecular (IBGM), Universidad de Valladolid and Consejo Superior de Investigaciones Científicas (CSIC), Valladolid, Spain
| | - Carlos Villalobos
- Instituto de Biología y Genética Molecular (IBGM), Universidad de Valladolid and Consejo Superior de Investigaciones Científicas (CSIC), Valladolid, Spain
| |
Collapse
|
26
|
Nam JH, Kim WK. The Role of TRP Channels in Allergic Inflammation and its Clinical Relevance. Curr Med Chem 2020; 27:1446-1468. [PMID: 30474526 DOI: 10.2174/0929867326666181126113015] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 09/03/2018] [Accepted: 11/07/2018] [Indexed: 12/24/2022]
Abstract
Allergy refers to an abnormal adaptive immune response to non-infectious environmental substances (allergen) that can induce various diseases such as asthma, atopic dermatitis, and allergic rhinitis. In this allergic inflammation, various immune cells, such as B cells, T cells, and mast cells, are involved and undergo complex interactions that cause a variety of pathophysiological conditions. In immune cells, calcium ions play a crucial role in controlling intracellular Ca2+ signaling pathways. Cations, such as Na+, indirectly modulate the calcium signal generation by regulating cell membrane potential. This intracellular Ca2+ signaling is mediated by various cation channels; among them, the Transient Receptor Potential (TRP) family is present in almost all immune cell types, and each channel has a unique function in regulating Ca2+ signals. In this review, we focus on the role of TRP ion channels in allergic inflammatory responses in T cells and mast cells. In addition, the TRP ion channels, which are attracting attention in clinical practice in relation to allergic diseases, and the current status of the development of therapeutic agents that target TRP channels are discussed.
Collapse
Affiliation(s)
- Joo Hyun Nam
- Department of Physiology, Dongguk University College of Medicine, 123 Dongdae-ro, Gyeongju 38066, Korea.,Channelopathy Research Center (CRC), Dongguk University College of Medicine, 32 Dongguk-ro, Ilsan Dong-gu, Goyang, Gyeonggi-do 10326, Korea
| | - Woo Kyung Kim
- Channelopathy Research Center (CRC), Dongguk University College of Medicine, 32 Dongguk-ro, Ilsan Dong-gu, Goyang, Gyeonggi-do 10326, Korea.,Department of Internal Medicine Graduate School of Medicine, Dongguk University, 27 Dongguk-ro, Ilsan Dong-gu, Goyang, Gyeonggi-do 10326, Korea
| |
Collapse
|
27
|
Häusler D, Torke S, Weber MS. High-Dose Vitamin D-Mediated Hypercalcemia as a Potential Risk Factor in Central Nervous System Demyelinating Disease. Front Immunol 2020; 11:301. [PMID: 32161591 PMCID: PMC7053380 DOI: 10.3389/fimmu.2020.00301] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 02/06/2020] [Indexed: 12/28/2022] Open
Abstract
The exact cause of multiple sclerosis (MS) is unknown; however, it is considered to be an inflammatory disease of the central nervous system (CNS) triggered by a combination of both environmental and genetic factors. Vitamin D deficiency is also discussed as a possible disease-promoting factor in MS, as low vitamin D status is associated with increased formation of CNS lesions, elevated number of relapses and accelerated disease progression. However, it remains unclear whether this association is causal and related and most importantly, whether vitamin D supplementation in MS is of direct therapeutic benefit. Recently, we could show that in a murine model of MS, administration of a moderate vitamin D dose was of clinical benefit, while excessive vitamin D supplementation had a negative effect on disease severity. Of note, disease exacerbation was associated with high-dose vitamin D caused secondary hypercalcemia. Mechanistically dissecting this outcome, we found that hypercalcemia independent of vitamin D similarly triggered activation of disease-perpetuating T cells. These findings caution that vitamin D should be supplemented in a controlled and moderate manner in patients with MS and concomitantly highlight calcium as a novel potential MS risk factor by itself. In this review, we will summarize the current evidence from animal and clinical studies aiming to assess whether vitamin D may be of benefit in patients with MS. Furthermore, we will discuss any possible secondary effects of vitamin D with a particular focus on the role of calcium on immune cells and in the pathogenesis of CNS demyelinating disease.
Collapse
Affiliation(s)
- Darius Häusler
- Department of Neuropathology, Institute of Neuropathology, University Medical Center, Göttingen, Germany
| | - Sebastian Torke
- Department of Neuropathology, Institute of Neuropathology, University Medical Center, Göttingen, Germany
| | - Martin S Weber
- Department of Neuropathology, Institute of Neuropathology, University Medical Center, Göttingen, Germany.,Department of Neurology, University Medical Center, Göttingen, Germany
| |
Collapse
|
28
|
The regulatory roles of calcium channels in tumors. Biochem Pharmacol 2019; 169:113603. [DOI: 10.1016/j.bcp.2019.08.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 08/08/2019] [Indexed: 02/06/2023]
|
29
|
Bhuvaneshwari S, Sankaranarayanan K. Structural and Mechanistic Insights of CRAC Channel as a Drug Target in Autoimmune Disorder. Curr Drug Targets 2019; 21:55-75. [PMID: 31556856 DOI: 10.2174/1389450120666190926150258] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 08/20/2019] [Accepted: 08/20/2019] [Indexed: 01/17/2023]
Abstract
BACKGROUND Calcium (Ca2+) ion is a major intracellular signaling messenger, controlling a diverse array of cellular functions like gene expression, secretion, cell growth, proliferation, and apoptosis. The major mechanism controlling this Ca2+ homeostasis is store-operated Ca2+ release-activated Ca2+ (CRAC) channels. CRAC channels are integral membrane protein majorly constituted via two proteins, the stromal interaction molecule (STIM) and ORAI. Following Ca2+ depletion in the Endoplasmic reticulum (ER) store, STIM1 interacts with ORAI1 and leads to the opening of the CRAC channel gate and consequently allows the influx of Ca2+ ions. A plethora of studies report that aberrant CRAC channel activity due to Loss- or gain-of-function mutations in ORAI1 and STIM1 disturbs this Ca2+ homeostasis and causes several autoimmune disorders. Hence, it clearly indicates that the therapeutic target of CRAC channels provides the space for a new approach to treat autoimmune disorders. OBJECTIVE This review aims to provide the key structural and mechanical insights of STIM1, ORAI1 and other molecular modulators involved in CRAC channel regulation. RESULTS AND CONCLUSION Understanding the structure and function of the protein is the foremost step towards improving the effective target specificity by limiting their potential side effects. Herein, the review mainly focusses on the structural underpinnings of the CRAC channel gating mechanism along with its biophysical properties that would provide the solid foundation to aid the development of novel targeted drugs for an autoimmune disorder. Finally, the immune deficiencies caused due to mutations in CRAC channel and currently used pharmacological blockers with their limitation are briefly summarized.
Collapse
Affiliation(s)
- Sampath Bhuvaneshwari
- Ion Channel Biology Laboratory, AU-KBC Research Centre, Madras Institute of Technology, Anna University, Chrompet, Chennai -600 044, India
| | - Kavitha Sankaranarayanan
- Ion Channel Biology Laboratory, AU-KBC Research Centre, Madras Institute of Technology, Anna University, Chrompet, Chennai -600 044, India
| |
Collapse
|
30
|
Tellechea A, Bai S, Dangwal S, Theocharidis G, Nagai M, Koerner S, Cheong JE, Bhasin S, Shih TY, Zheng Y, Zhao W, Zhang C, Li X, Kounas K, Panagiotidou S, Theoharides T, Mooney D, Bhasin M, Sun L, Veves A. Topical Application of a Mast Cell Stabilizer Improves Impaired Diabetic Wound Healing. J Invest Dermatol 2019; 140:901-911.e11. [PMID: 31568772 DOI: 10.1016/j.jid.2019.08.449] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 08/19/2019] [Accepted: 08/21/2019] [Indexed: 02/09/2023]
Abstract
Impaired wound healing in the diabetic foot is a major problem often leading to amputation. Mast cells have been shown to regulate wound healing in diabetes. We developed an indole-carboxamide type mast cell stabilizer, MCS-01, which proved to be an effective mast cell degranulation inhibitor in vitro and can be delivered topically for prolonged periods through controlled release by specifically designed alginate bandages. In diabetic mice, both pre- and post-wounding, topical MCS-01 application accelerated wound healing comparable to that achieved with systemic mast cell stabilization. Moreover, MCS-01 altered the macrophage phenotype, promoting classically activated polarization. Bulk transcriptome analysis from wounds treated with MCS-01 or placebo showed that MCS-01 significantly modulated the mRNA and microRNA profile of diabetic wounds, stimulated upregulation of pathways linked to acute inflammation and immune cell migration, and activated the NF-κB complex along with other master regulators of inflammation. Single-cell RNA sequencing analysis of 6,154 cells from wounded and unwounded mouse skin revealed that MCS-01 primarily altered the gene expression of mast cells, monocytes, and keratinocytes. Taken together, these findings offer insights into the process of diabetic wound healing and suggest topical mast cell stabilization as a potentially successful treatment for diabetic foot ulceration.
Collapse
Affiliation(s)
- Ana Tellechea
- Joslin-Beth Israel Deaconess Foot Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA; The Rongxiang Xu, MD, Center for Regenerative Therapeutics, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Sha Bai
- Center for Drug Discovery and Translational Research, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Seema Dangwal
- Joslin-Beth Israel Deaconess Foot Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA; The Rongxiang Xu, MD, Center for Regenerative Therapeutics, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA; Institute for Translational and Therapeutics Strategies, Hannover Medical School, Hannover, Germany
| | - Georgios Theocharidis
- Joslin-Beth Israel Deaconess Foot Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA; The Rongxiang Xu, MD, Center for Regenerative Therapeutics, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Masa Nagai
- Center for Drug Discovery and Translational Research, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Steffi Koerner
- Center for Drug Discovery and Translational Research, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Jae Eun Cheong
- Center for Drug Discovery and Translational Research, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Swati Bhasin
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA; Division of Interdisciplinary Medicine and Biotechnology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA; Genomics, Proteomics, Bioinformatics and Systems Biology Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Ting-Yu Shih
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts, USA; Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, Massachusetts, USA
| | - YongJun Zheng
- Joslin-Beth Israel Deaconess Foot Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA; The Rongxiang Xu, MD, Center for Regenerative Therapeutics, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Wanni Zhao
- Joslin-Beth Israel Deaconess Foot Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA; The Rongxiang Xu, MD, Center for Regenerative Therapeutics, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Cuiping Zhang
- Joslin-Beth Israel Deaconess Foot Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA; The Rongxiang Xu, MD, Center for Regenerative Therapeutics, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Xiaoli Li
- Joslin-Beth Israel Deaconess Foot Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA; The Rongxiang Xu, MD, Center for Regenerative Therapeutics, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Konstantinos Kounas
- Joslin-Beth Israel Deaconess Foot Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA; The Rongxiang Xu, MD, Center for Regenerative Therapeutics, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Smaro Panagiotidou
- Laboratory of Immunopharmacology and Drug, Discovery Department of Immunology Tufts University School of Medicine, Boston, Massachusetts, USA
| | - Theoharis Theoharides
- Laboratory of Immunopharmacology and Drug, Discovery Department of Immunology Tufts University School of Medicine, Boston, Massachusetts, USA
| | - David Mooney
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts, USA; Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, Massachusetts, USA
| | - Manoj Bhasin
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA; Division of Interdisciplinary Medicine and Biotechnology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA; Genomics, Proteomics, Bioinformatics and Systems Biology Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA.
| | - Lijun Sun
- Center for Drug Discovery and Translational Research, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA.
| | - Aristidis Veves
- Joslin-Beth Israel Deaconess Foot Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA; The Rongxiang Xu, MD, Center for Regenerative Therapeutics, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA.
| |
Collapse
|
31
|
Monaco S, Jahraus B, Samstag Y, Bading H. Conditions of limited calcium influx (CLCI) inhibits IL2 induction and favors expression of anergy-related genes in TCR/CD3 and CD28 costimulated primary human T cells. Mol Immunol 2019; 114:81-87. [PMID: 31344552 DOI: 10.1016/j.molimm.2019.07.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 07/08/2019] [Accepted: 07/09/2019] [Indexed: 12/31/2022]
Abstract
Calcium is a key regulator of the T cell immune response. Depending on the spatial properties (nucleus versus cytoplasm) of the calcium signals generated after CD3xCD28 stimulation, primary human T cells either mount a productive immune response or develop tolerance. Nuclear calcium acts as a genomic decision maker: during T cell activation, it drives expression of genes associated with a productive immune response while in its absence, stimulated T cells acquire an anergy-like gene profile. Selective inhibition of nuclear calcium signaling in stimulated T cells blocks the productive immune response and directs the cells towards an anergy-like state. Here we show that the two transcriptional programs that include, respectively, the 'activation gene', interleukin 2 (IL2) and 'anergy-related genes', EGR2, EGR3, and CREM have different requirements for transmembrane calcium flux. By either lowering extracellular calcium concentrations with EGTA or using low concentrations of the ORAI blockers, BTP2 or RO2959, we reduced transmembrane calcium flux in human primary T cells stimulated with CD3xCD28. These 'conditions of limited calcium influx' (CLCI) blocked CD3xCD28-induced IL2 expression but only moderately affected induction of the anergy-related genes EGR2, EGR3, and CREM. We observed no difference in NFAT2 nuclear translocation after CD3xCD28 stimulation between normal conditions and CLCI. These results indicate that CLCI favors expression of anergy-related genes in activated human T cells. CLCI may be used to develop novel means for pro-tolerance immunosuppressive treatments.
Collapse
Affiliation(s)
- Sara Monaco
- Department of Neurobiology, Interdisciplinary Center for Neurosciences, Heidelberg University, 69120, Heidelberg, Germany.
| | - Beate Jahraus
- Department of Immunology, Heidelberg University, 69120, Heidelberg, Germany.
| | - Yvonne Samstag
- Department of Immunology, Heidelberg University, 69120, Heidelberg, Germany.
| | - Hilmar Bading
- Department of Neurobiology, Interdisciplinary Center for Neurosciences, Heidelberg University, 69120, Heidelberg, Germany.
| |
Collapse
|
32
|
Rosenberg P, Katz D, Bryson V. SOCE and STIM1 signaling in the heart: Timing and location matter. Cell Calcium 2018; 77:20-28. [PMID: 30508734 DOI: 10.1016/j.ceca.2018.11.008] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 11/26/2018] [Accepted: 11/26/2018] [Indexed: 01/11/2023]
Abstract
Store operated Ca2+ entry (SOCE) is an ancient and ubiquitous Ca2+ signaling pathway discovered decades ago, but the function of SOCE in human physiology is only now being revealed. The relevance of this pathway to striated muscle was solidified with the description of skeletal myopathies that result from mutations in STIM1 and Orai1, the two SOCE components. Here, we consider the evidence for STIM1 and SOCE in cardiac muscle and the sinoatrial node. We highlight recent studies revealing a role for STIM1 in cardiac growth in response to developmental and pathologic cues. We also review the role of STIM1 in the regulation of SOCE and Ca2+ store refilling in a non-Orai dependent manner. Finally, we discuss the importance of this pathway in ventricular cardiomyocytes where SOCE contribute to developmental growth and in pacemaker cells where SOCE likely has a fundamental to generating the cardiac rhythm.
Collapse
Affiliation(s)
- Paul Rosenberg
- Department of Medicine, Duke University School of Medicine, Durham, NC, United States.
| | - Danielle Katz
- Department of Medicine, Duke University School of Medicine, Durham, NC, United States
| | - Victoria Bryson
- Department of Medicine, Duke University School of Medicine, Durham, NC, United States
| |
Collapse
|
33
|
Riva B, Griglio A, Serafini M, Cordero-Sanchez C, Aprile S, Di Paola R, Gugliandolo E, Alansary D, Biocotino I, Lim D, Grosa G, Galli U, Niemeyer B, Sorba G, Canonico PL, Cuzzocrea S, Genazzani AA, Pirali T. Pyrtriazoles, a Novel Class of Store-Operated Calcium Entry Modulators: Discovery, Biological Profiling, and in Vivo Proof-of-Concept Efficacy in Acute Pancreatitis. J Med Chem 2018; 61:9756-9783. [PMID: 30347159 DOI: 10.1021/acs.jmedchem.8b01512] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
In recent years, channels that mediate store-operated calcium entry (SOCE, i.e., the ability of cells to sense a decrease in endoplasmic reticulum luminal calcium and induce calcium entry across the plasma membrane) have been associated with a number of disorders, spanning from immune disorders to acute pancreatitis and have been suggested to be druggable targets. In the present contribution, we exploited the click chemistry approach to synthesize a class of SOCE modulators where the arylamide substructure that characterizes most inhibitors so far described is substituted by a 1,4-disubstituted 1,2,3-triazole ring. Within this series, inhibitors of SOCE were identified and the best compound proved effective in an animal model of acute pancreatitis, a disease characterized by a hyperactivation of SOCE. Strikingly, two enhancers of the process were discovered, affording invaluable research tools to further explore the (patho)physiological role of capacitative calcium entry.
Collapse
Affiliation(s)
- Beatrice Riva
- Department of Pharmaceutical Sciences , Università del Piemonte Orientale , Novara 28100 , Italy.,ChemICare Srl , Enne3 , Novara 28100 , Italy
| | - Alessia Griglio
- Department of Pharmaceutical Sciences , Università del Piemonte Orientale , Novara 28100 , Italy
| | - Marta Serafini
- Department of Pharmaceutical Sciences , Università del Piemonte Orientale , Novara 28100 , Italy
| | - Celia Cordero-Sanchez
- Department of Pharmaceutical Sciences , Università del Piemonte Orientale , Novara 28100 , Italy
| | - Silvio Aprile
- Department of Pharmaceutical Sciences , Università del Piemonte Orientale , Novara 28100 , Italy
| | - Rosanna Di Paola
- Department of Chemical, Biological, Pharmaceutical, and Enviromental Sciences , Università di Messina , Messina 98166 , Italy
| | - Enrico Gugliandolo
- Department of Chemical, Biological, Pharmaceutical, and Enviromental Sciences , Università di Messina , Messina 98166 , Italy
| | - Dalia Alansary
- Department of Molecular Biophysics , Saarland University CIPMM , Homburg 66421 , Germany
| | - Isabella Biocotino
- Department of Pharmaceutical Sciences , Università del Piemonte Orientale , Novara 28100 , Italy
| | - Dmitry Lim
- Department of Pharmaceutical Sciences , Università del Piemonte Orientale , Novara 28100 , Italy
| | - Giorgio Grosa
- Department of Pharmaceutical Sciences , Università del Piemonte Orientale , Novara 28100 , Italy
| | - Ubaldina Galli
- Department of Pharmaceutical Sciences , Università del Piemonte Orientale , Novara 28100 , Italy
| | - Barbara Niemeyer
- Department of Molecular Biophysics , Saarland University CIPMM , Homburg 66421 , Germany
| | - Giovanni Sorba
- Department of Pharmaceutical Sciences , Università del Piemonte Orientale , Novara 28100 , Italy
| | - Pier Luigi Canonico
- Department of Pharmaceutical Sciences , Università del Piemonte Orientale , Novara 28100 , Italy
| | - Salvatore Cuzzocrea
- Department of Chemical, Biological, Pharmaceutical, and Enviromental Sciences , Università di Messina , Messina 98166 , Italy
| | - Armando A Genazzani
- Department of Pharmaceutical Sciences , Università del Piemonte Orientale , Novara 28100 , Italy
| | - Tracey Pirali
- Department of Pharmaceutical Sciences , Università del Piemonte Orientale , Novara 28100 , Italy.,ChemICare Srl , Enne3 , Novara 28100 , Italy
| |
Collapse
|
34
|
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: 11.0] [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
|
35
|
Stevenson RJ, Azimi I, Flanagan JU, Inserra M, Vetter I, Monteith GR, Denny WA. An SAR study of hydroxy-trifluoromethylpyrazolines as inhibitors of Orai1-mediated store operated Ca 2+ entry in MDA-MB-231 breast cancer cells using a convenient Fluorescence Imaging Plate Reader assay. Bioorg Med Chem 2018; 26:3406-3413. [PMID: 29776832 DOI: 10.1016/j.bmc.2018.05.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 05/01/2018] [Accepted: 05/08/2018] [Indexed: 12/30/2022]
Abstract
The proteins Orai1 and STIM1 control store-operated Ca2+ entry (SOCE) into cells. SOCE is important for migration, invasion and metastasis of MDA-MB-231 human triple negative breast cancer (TNBC) cells and has been proposed as a target for cancer drug discovery. Two hit compounds from a medium throughput screen, displayed encouraging inhibition of SOCE in MDA-MB-231 cells, as measured by a Fluorescence Imaging Plate Reader (FLIPR) Ca2+ assay. Following NMR spectroscopic analysis of these hits and reassignment of their structures as 5-hydroxy-5-trifluoromethylpyrazolines, a series of analogues was prepared via thermal condensation reactions between substituted acylhydrazones and trifluoromethyl 1,3-dicarbonyl arenes. Structure-activity relationship (SAR) studies showed that small lipophilic substituents at the 2- and 3-positions of the RHS and 2-, 3- and 4-postions of the LHS terminal benzene rings improved activity, resulting in a novel class of potent and selective inhibitors of SOCE.
Collapse
Affiliation(s)
- Ralph J Stevenson
- Auckland Cancer Society Research Centre, School of Medical Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand.
| | - Iman Azimi
- The School of Pharmacy, The University of Queensland, Brisbane, Queensland, Australia; Mater Research Institute, The University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia; Division of Pharmacy, College of Health and Medicine, University of Tasmania, Hobart, Tasmania, Australia
| | - Jack U Flanagan
- Auckland Cancer Society Research Centre, School of Medical Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand; Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Marco Inserra
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland 4072, Australia; School of Pharmacy, The University of Queensland, Woolloongabba, Queensland 4102, Australia
| | - Irina Vetter
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland 4072, Australia; School of Pharmacy, The University of Queensland, Woolloongabba, Queensland 4102, Australia
| | - Gregory R Monteith
- The School of Pharmacy, The University of Queensland, Brisbane, Queensland, Australia; Mater Research Institute, The University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia
| | - William A Denny
- Auckland Cancer Society Research Centre, School of Medical Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand; Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand.
| |
Collapse
|
36
|
A gene module associated with dysregulated TCR signaling pathways in CD4 + T cell subsets in rheumatoid arthritis. J Autoimmun 2018; 89:21-29. [DOI: 10.1016/j.jaut.2017.11.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Revised: 11/01/2017] [Accepted: 11/05/2017] [Indexed: 01/27/2023]
|
37
|
Zheng C, Zhong M, Qi Z, Shen F, Zhao Q, Wu L, Huang Y, Tsang SY, Yao X. Histone Deacetylase Inhibitors Relax Mouse Aorta Partly through Their Inhibitory Action on L-Type Ca 2+ Channels. J Pharmacol Exp Ther 2017; 363:211-220. [PMID: 28860353 DOI: 10.1124/jpet.117.242685] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2017] [Accepted: 08/21/2017] [Indexed: 12/21/2022] Open
Abstract
Histone deacetylase (HDAC) inhibitors modulate acetylation/deacetylation of histone and nonhistone proteins. They have been widely used for cancer treatment. However, there have been only a few studies investigating the effect of HDAC inhibitors on vascular tone regulation, most of which employed chronic treatment with HDAC inhibitors. In the present study, we found that two hydroxamate-based pan-HDAC inhibitors, suberoylanilide hydroxamic acid (SAHA) and trichostatin A (TSA), could partially but acutely relax high extracellular K+-contracted mouse aortas. SAHA and TSA also attenuated the high extracellular K+-induced cytosolic Ca2+ rise and inhibited L-type Ca2+ channel current in whole-cell patch-clamp. These data demonstrate that SAHA could inhibit L-type Ca2+ channels to cause vascular relaxation. In addition, SAHA and TSA dose dependently relaxed the arteries precontracted with phenylephrine. The relaxant effect of SAHA and TSA was greater in phenylephrine-precontracted arteries than in high K+-contracted arteries. Although part of the relaxant effect of SAHA and TSA on phenylephrine-precontracted arteries was related to L-type Ca2+ channels, both agents could also induce relaxation via a mechanism independent of L-type Ca2+ channels. Taken together, HDAC inhibitors SAHA and TSA can acutely relax blood vessels via their inhibitory action on L-type Ca2+ channels and via another L-type Ca2+ channel-independent mechanism.
Collapse
Affiliation(s)
- Changbo Zheng
- Department of Physiology, Anhui Medical University, Hefei, China (M.Z., F.S.); School of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, Yunnan, China (C.Z.); and School of Biomedical Sciences and Li Ka Shing Institute of Health Science (C.Z., Q.Z., L.W., Y.H., X.Y.), School of Life Sciences (S.-Y.T, Z.Q.), Shenzhen Research Institute (C.Z., Q.Z., L.W., X.Y.), The Chinese University of Hong Kong, Shenzhen, China
| | - Mingkui Zhong
- Department of Physiology, Anhui Medical University, Hefei, China (M.Z., F.S.); School of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, Yunnan, China (C.Z.); and School of Biomedical Sciences and Li Ka Shing Institute of Health Science (C.Z., Q.Z., L.W., Y.H., X.Y.), School of Life Sciences (S.-Y.T, Z.Q.), Shenzhen Research Institute (C.Z., Q.Z., L.W., X.Y.), The Chinese University of Hong Kong, Shenzhen, China.
| | - Zenghua Qi
- Department of Physiology, Anhui Medical University, Hefei, China (M.Z., F.S.); School of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, Yunnan, China (C.Z.); and School of Biomedical Sciences and Li Ka Shing Institute of Health Science (C.Z., Q.Z., L.W., Y.H., X.Y.), School of Life Sciences (S.-Y.T, Z.Q.), Shenzhen Research Institute (C.Z., Q.Z., L.W., X.Y.), The Chinese University of Hong Kong, Shenzhen, China
| | - Fan Shen
- Department of Physiology, Anhui Medical University, Hefei, China (M.Z., F.S.); School of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, Yunnan, China (C.Z.); and School of Biomedical Sciences and Li Ka Shing Institute of Health Science (C.Z., Q.Z., L.W., Y.H., X.Y.), School of Life Sciences (S.-Y.T, Z.Q.), Shenzhen Research Institute (C.Z., Q.Z., L.W., X.Y.), The Chinese University of Hong Kong, Shenzhen, China
| | - Qiannan Zhao
- Department of Physiology, Anhui Medical University, Hefei, China (M.Z., F.S.); School of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, Yunnan, China (C.Z.); and School of Biomedical Sciences and Li Ka Shing Institute of Health Science (C.Z., Q.Z., L.W., Y.H., X.Y.), School of Life Sciences (S.-Y.T, Z.Q.), Shenzhen Research Institute (C.Z., Q.Z., L.W., X.Y.), The Chinese University of Hong Kong, Shenzhen, China
| | - Lulu Wu
- Department of Physiology, Anhui Medical University, Hefei, China (M.Z., F.S.); School of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, Yunnan, China (C.Z.); and School of Biomedical Sciences and Li Ka Shing Institute of Health Science (C.Z., Q.Z., L.W., Y.H., X.Y.), School of Life Sciences (S.-Y.T, Z.Q.), Shenzhen Research Institute (C.Z., Q.Z., L.W., X.Y.), The Chinese University of Hong Kong, Shenzhen, China
| | - Yu Huang
- Department of Physiology, Anhui Medical University, Hefei, China (M.Z., F.S.); School of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, Yunnan, China (C.Z.); and School of Biomedical Sciences and Li Ka Shing Institute of Health Science (C.Z., Q.Z., L.W., Y.H., X.Y.), School of Life Sciences (S.-Y.T, Z.Q.), Shenzhen Research Institute (C.Z., Q.Z., L.W., X.Y.), The Chinese University of Hong Kong, Shenzhen, China
| | - Suk-Ying Tsang
- Department of Physiology, Anhui Medical University, Hefei, China (M.Z., F.S.); School of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, Yunnan, China (C.Z.); and School of Biomedical Sciences and Li Ka Shing Institute of Health Science (C.Z., Q.Z., L.W., Y.H., X.Y.), School of Life Sciences (S.-Y.T, Z.Q.), Shenzhen Research Institute (C.Z., Q.Z., L.W., X.Y.), The Chinese University of Hong Kong, Shenzhen, China
| | - Xiaoqiang Yao
- Department of Physiology, Anhui Medical University, Hefei, China (M.Z., F.S.); School of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, Yunnan, China (C.Z.); and School of Biomedical Sciences and Li Ka Shing Institute of Health Science (C.Z., Q.Z., L.W., Y.H., X.Y.), School of Life Sciences (S.-Y.T, Z.Q.), Shenzhen Research Institute (C.Z., Q.Z., L.W., X.Y.), The Chinese University of Hong Kong, Shenzhen, China.
| |
Collapse
|
38
|
Zheng C, Lo CY, Meng Z, Li Z, Zhong M, Zhang P, Lu J, Yang Z, Yan F, Zhang Y, Huang Y, Yao X. Gastrodin Inhibits Store-Operated Ca 2+ Entry and Alleviates Cardiac Hypertrophy. Front Pharmacol 2017; 8:222. [PMID: 28487655 PMCID: PMC5404510 DOI: 10.3389/fphar.2017.00222] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Accepted: 04/10/2017] [Indexed: 11/13/2022] Open
Abstract
Cardiac hypertrophy is a major risk factor for heart failure, which are among the leading causes of human death. Gastrodin is a small molecule that has been used clinically to treat neurological and vascular diseases for many years without safety issues. In the present study, we examined protective effect of gastrodin against cardiac hypertrophy and explored the underlying mechanism. Phenylephrine and angiotensin II were used to induce cardiac hypertrophy in a mouse model and a cultured cardiomyocyte model. Gastrodin was found to alleviate the cardiac hypertrophy in both models. Mechanistically, gastrodin attenuated the store-operated Ca2+ entry (SOCE) by reducing the expression of STIM1 and Orai1, two key proteins in SOCE, in animal models as well as in cultured cardiomyocyte model. Furthermore, suppressing SOCE by RO2959, Orai1-siRNAs or STIM1-siRNAs markedly attenuated the phenylephrine-induced hypertrophy in cultured cardiomyocyte model. Together, these results showed that gastrodin inhibited cardiac hypertrophy and it also reduced the SOCE via its action on the expression of STIM1 and Orai1. Furthermore, suppression of SOCE could reduce the phenylephrine-induced cardiomyocyte hypertrophy, suggesting that SOCE-STIM1-Orai1 is located upstream of hypertrophy.
Collapse
Affiliation(s)
- Changbo Zheng
- Li Ka Shing Institute of Health Sciences and School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong KongHong Kong, China.,Shenzhen Research Institute, The Chinese University of Hong KongShenzhen, China
| | - Chun-Yin Lo
- Li Ka Shing Institute of Health Sciences and School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong KongHong Kong, China.,Shenzhen Research Institute, The Chinese University of Hong KongShenzhen, China
| | - Zhaoyue Meng
- Li Ka Shing Institute of Health Sciences and School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong KongHong Kong, China.,Shenzhen Research Institute, The Chinese University of Hong KongShenzhen, China.,School of Life Sciences, The Chinese University of Hong KongHong Kong, China
| | - Zhichao Li
- Li Ka Shing Institute of Health Sciences and School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong KongHong Kong, China.,Shenzhen Research Institute, The Chinese University of Hong KongShenzhen, China
| | - Mingkui Zhong
- Li Ka Shing Institute of Health Sciences and School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong KongHong Kong, China
| | - Peng Zhang
- Li Ka Shing Institute of Health Sciences and School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong KongHong Kong, China
| | - Jun Lu
- Li Ka Shing Institute of Health Sciences and School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong KongHong Kong, China.,Shenzhen Research Institute, The Chinese University of Hong KongShenzhen, China
| | - Zhaoxiang Yang
- Institute for Drug Research and Development, Kunming Pharmaceutical CorporationKunming, China
| | - Fuman Yan
- Li Ka Shing Institute of Health Sciences and School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong KongHong Kong, China
| | - Yunting Zhang
- Li Ka Shing Institute of Health Sciences and School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong KongHong Kong, China
| | - Yu Huang
- Li Ka Shing Institute of Health Sciences and School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong KongHong Kong, China
| | - Xiaoqiang Yao
- Li Ka Shing Institute of Health Sciences and School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong KongHong Kong, China.,Shenzhen Research Institute, The Chinese University of Hong KongShenzhen, China
| |
Collapse
|
39
|
Chiang EY, Li T, Jeet S, Peng I, Zhang J, Lee WP, DeVoss J, Caplazi P, Chen J, Warming S, Hackos DH, Mukund S, Koth CM, Grogan JL. Potassium channels Kv1.3 and KCa3.1 cooperatively and compensatorily regulate antigen-specific memory T cell functions. Nat Commun 2017; 8:14644. [PMID: 28248292 PMCID: PMC5337993 DOI: 10.1038/ncomms14644] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Accepted: 01/18/2017] [Indexed: 01/01/2023] Open
Abstract
Voltage-gated Kv1.3 and Ca2+-dependent KCa3.1 are the most prevalent K+ channels expressed by human and rat T cells. Despite the preferential upregulation of Kv1.3 over KCa3.1 on autoantigen-experienced effector memory T cells, whether Kv1.3 is required for their induction and function is unclear. Here we show, using Kv1.3-deficient rats, that Kv1.3 is involved in the development of chronically activated antigen-specific T cells. Several immune responses are normal in Kv1.3 knockout (KO) rats, suggesting that KCa3.1 can compensate for the absence of Kv1.3 under these specific settings. However, experiments with Kv1.3 KO rats and Kv1.3 siRNA knockdown or channel-specific inhibition of human T cells show that maximal T-cell responses against autoantigen or repeated tetanus toxoid stimulations require both Kv1.3 and KCa3.1. Finally, our data also suggest that T-cell dependency on Kv1.3 or KCa3.1 might be irreversibly modulated by antigen exposure.
Collapse
Affiliation(s)
- Eugene Y Chiang
- Department of Immunology, Genentech Inc., 1 DNA Way, South San Francisco, California 94080, USA
| | - Tianbo Li
- Department of Biochemical and Cellular Pharmacology, Genentech Inc., 1 DNA Way, South San Francisco, California 94080, USA
| | - Surinder Jeet
- Department of Translational Immunology, Genentech Inc., 1 DNA Way, South San Francisco, California 94080, USA
| | - Ivan Peng
- Department of Translational Immunology, Genentech Inc., 1 DNA Way, South San Francisco, California 94080, USA
| | - Juan Zhang
- Department of Translational Immunology, Genentech Inc., 1 DNA Way, South San Francisco, California 94080, USA
| | - Wyne P Lee
- Department of Translational Immunology, Genentech Inc., 1 DNA Way, South San Francisco, California 94080, USA
| | - Jason DeVoss
- Department of Translational Immunology, Genentech Inc., 1 DNA Way, South San Francisco, California 94080, USA
| | - Patrick Caplazi
- Department of Pathology, Genentech Inc., 1 DNA Way, South San Francisco, California 94080, USA
| | - Jun Chen
- Department of Biochemical and Cellular Pharmacology, Genentech Inc., 1 DNA Way, South San Francisco, California 94080, USA
| | - Søren Warming
- Department of Molecular Biology, Genentech Inc., 1 DNA Way, South San Francisco, California 94080, USA
| | - David H Hackos
- Department of Neurobiology, Genentech Inc., 1 DNA Way, South San Francisco, California 94080, USA
| | - Susmith Mukund
- Department of Structural Biology, Genentech Inc., 1 DNA Way, South San Francisco, California 94080, USA
| | - Christopher M Koth
- Department of Structural Biology, Genentech Inc., 1 DNA Way, South San Francisco, California 94080, USA
| | - Jane L Grogan
- Department of Immunology, Genentech Inc., 1 DNA Way, South San Francisco, California 94080, USA
| |
Collapse
|
40
|
Bai S, Nagai M, Koerner SK, Veves A, Sun L. Structure-activity relationship study and discovery of indazole 3-carboxamides as calcium-release activated calcium channel blockers. Bioorg Med Chem Lett 2017; 27:393-397. [PMID: 28057422 PMCID: PMC5271583 DOI: 10.1016/j.bmcl.2016.12.062] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 12/23/2016] [Accepted: 12/24/2016] [Indexed: 02/04/2023]
Abstract
Aberrant activation of mast cells contributes to the development of numerous diseases including cancer, autoimmune disorders, as well as diabetes and its complications. The influx of extracellular calcium via the highly calcium selective calcium-release activated calcium (CRAC) channel controls mast cell functions. Intracellular calcium homeostasis in mast cells can be maintained via the modulation of the CRAC channel, representing a critical point for therapeutic interventions. We describe the structure-activity relationship study (SAR) of indazole-3-carboxamides as potent CRAC channel blockers and their ability to stabilize mast cells. Our SAR results show that the unique regiochemistry of the amide linker is critical for the inhibition of calcium influx, the release of the pro-inflammatory mediators β-hexosaminidase and tumor necrosis factor α by activated mast cells. Thus, the indazole-3-carboxamide 12d actively inhibits calcium influx and stabilizes mast cells with sub-μM IC50. In contrast, its reverse amide isomer 9c is inactive in the calcium influx assay even at 100μM concentration. This requirement of the specific 3-carboxamide regiochemistry in indazoles is unprecedented in known CRAC channel blockers. The new structural scaffolds described in this report expand the structural diversity of the CRAC channel blockers and may lead to the discovery of novel immune modulators for the treatment of human diseases.
Collapse
Affiliation(s)
- Sha Bai
- Center for Drug Discovery and Translational Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Masazumi Nagai
- Center for Drug Discovery and Translational Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Steffi K Koerner
- Center for Drug Discovery and Translational Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Aristidis Veves
- The Rongxiang Xu, MD Center for Regenerative Therapeutics, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Lijun Sun
- Center for Drug Discovery and Translational Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.
| |
Collapse
|
41
|
Cui C, Merritt R, Fu L, Pan Z. Targeting calcium signaling in cancer therapy. Acta Pharm Sin B 2017; 7:3-17. [PMID: 28119804 PMCID: PMC5237760 DOI: 10.1016/j.apsb.2016.11.001] [Citation(s) in RCA: 383] [Impact Index Per Article: 54.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Accepted: 10/28/2016] [Indexed: 12/15/2022] Open
Abstract
The intracellular calcium ions (Ca2+) act as second messenger to regulate gene transcription, cell proliferation, migration and death. Accumulating evidences have demonstrated that intracellular Ca2+ homeostasis is altered in cancer cells and the alteration is involved in tumor initiation, angiogenesis, progression and metastasis. Targeting derailed Ca2+ signaling for cancer therapy has become an emerging research area. This review summarizes some important Ca2+ channels, transporters and Ca2+-ATPases, which have been reported to be altered in human cancer patients. It discusses the current research effort toward evaluation of the blockers, inhibitors or regulators for Ca2+ channels/transporters or Ca2+-ATPase pumps as anti-cancer drugs. This review is also aimed to stimulate interest in, and support for research into the understanding of cellular mechanisms underlying the regulation of Ca2+ signaling in different cancer cells, and to search for novel therapies to cure these malignancies by targeting Ca2+ channels or transporters.
Collapse
Key Words
- 20-GPPD, 20-O-β-D-glucopyranosyl-20(S)-protopanaxadiol
- Apoptosis
- CBD, cannabidiol
- CBG, cannabigerol
- CPZ, capsazepine
- CRAC, Ca2+ release-activated Ca2+ channel
- CTL, cytotoxic T cells
- CYP3A4, cytochrome P450 3A4
- Ca2+ channels
- CaM, calmodulin
- CaMKII, calmodulin-dependent protein kinase II
- Cancer therapy
- Cell proliferation
- Channel blockers;
- ER/SR, endoplasmic/sarcoplasmic reticulum
- HCX, H+/Ca2+ exchangers
- IP3, inositol 1,4,5-trisphosphate
- IP3R (1, 2, 3), IP3 receptor (type 1, type 2, type 3)
- MCU, mitochondrial Ca2+ uniporter
- MCUR1, MCU uniporter regulator 1
- MICU (1, 2, 3), mitochondrial calcium uptake (type 1, type 2, type 3)
- MLCK, myosin light-chain kinase
- Migration
- NCX, Na+/Ca2+ exchanger
- NF-κB, nuclear factor-κB
- NFAT, nuclear factor of activated T cells
- NSCLC, non-small cell lung cancer
- OSCC, oral squamous cell carcinoma cells
- PKC, protein kinase C
- PM, plasma membrane
- PMCA, plasma membrane Ca2+-ATPase
- PTP, permeability transition pore
- ROS, reactive oxygen species
- RyR, ryanodine receptor
- SERCA, SR/ER Ca2+-ATPase
- SOCE, store-operated Ca2+ entry
- SPCA, secretory pathway Ca2+-ATPase
- Store-operated Ca2+ entry
- TEA, tetraethylammonium
- TG, thapsigargin
- TPC2, two-pore channel 2
- TRIM, 1-(2-(trifluoromethyl) phenyl) imidazole
- TRP (A, C, M, ML, N, P, V), transient receptor potential (ankyrin, canonical, melastatin, mucolipin, no mechanoreceptor potential C, polycystic, vanilloid)
- VGCC, voltage-gated Ca2+ channel
- mAb, monoclonal antibody
Collapse
Affiliation(s)
- Chaochu Cui
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
- Department of Surgery, Division of Thoracic Surgery, The Ohio State University, Columbus, OH 43210, USA
| | - Robert Merritt
- Department of Surgery, Division of Thoracic Surgery, The Ohio State University, Columbus, OH 43210, USA
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| | - Liwu Fu
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Zui Pan
- Department of Surgery, Division of Thoracic Surgery, The Ohio State University, Columbus, OH 43210, USA
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
- College of Nursing and Health Innovation, The University of Texas at Arlington, Arlington, TX 76019, USA
| |
Collapse
|
42
|
Abstract
Aberrant Ca(2+) release-activated Ca(2+) (CRAC) channel activity has been implicated in a number of human disorders, including immunodeficiency, autoimmunity, occlusive vascular diseases and cancer, thus placing CRAC channels among the important targets for the treatment of these disorders. We briefly summarize herein the molecular basis and activation mechanism of CRAC channel and focus on discussing several pharmacological inhibitors of CRAC channels with respect to their biological activity, mechanisms of action and selectivity over other types of Ca(2+) channel in different types of cells.
Collapse
|
43
|
Velmurugan GV, Huang H, Sun H, Candela J, Jaiswal MK, Beaman KD, Yamashita M, Prakriya M, White C. Depletion of H2S during obesity enhances store-operated Ca2+ entry in adipose tissue macrophages to increase cytokine production. Sci Signal 2015; 8:ra128. [PMID: 26671149 DOI: 10.1126/scisignal.aac7135] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The increased production of proinflammatory cytokines by adipose tissue macrophages (ATMs) contributes to chronic, low-level inflammation during obesity. We found that obesity in mice reduced the bioavailability of the gaseous signaling molecule hydrogen sulfide (H2S). Steady-state, intracellular concentrations of H2S were lower in ATMs isolated from mice with diet-induced obesity than in ATMs from lean mice. In addition, the intracellular concentration of H2S in the macrophage cell line RAW264.7 was reduced during an acute inflammatory response evoked by the microbial product lipopolysaccharide (LPS). Reduced intracellular concentrations of H2S led to increased Ca(2+) influx through the store-operated Ca(2+) entry (SOCE) pathway, which was prevented by the exogenous H2S donor GYY4137. Furthermore, GYY4137 inhibited the Orai3 channel, a key component of the SOCE machinery. The enhanced production of proinflammatory cytokines by RAW264.7 cells and ATMs from obese mice was reduced by exogenous H2S or by inhibition of SOCE. Together, these data suggest that the depletion of macrophage H2S that occurs during acute (LPS-induced) or chronic (obesity) inflammation increases SOCE through disinhibition of Orai3 and promotes the production of proinflammatory cytokines.
Collapse
Affiliation(s)
- Gopal V Velmurugan
- Department of Physiology and Biophysics, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, USA
| | - Huiya Huang
- Department of Physiology and Biophysics, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, USA
| | - Hongbin Sun
- Department of Physiology and Biophysics, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, USA
| | - Joseph Candela
- Department of Physiology and Biophysics, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, USA
| | - Mukesh K Jaiswal
- Department of Microbiology and Immunology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, USA
| | - Kenneth D Beaman
- Department of Microbiology and Immunology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, USA
| | - Megumi Yamashita
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Murali Prakriya
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Carl White
- Department of Physiology and Biophysics, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, USA.
| |
Collapse
|
44
|
Calcium Regulation of Hemorrhagic Fever Virus Budding: Mechanistic Implications for Host-Oriented Therapeutic Intervention. PLoS Pathog 2015; 11:e1005220. [PMID: 26513362 PMCID: PMC4634230 DOI: 10.1371/journal.ppat.1005220] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Accepted: 09/21/2015] [Indexed: 12/19/2022] Open
Abstract
Hemorrhagic fever viruses, including the filoviruses (Ebola and Marburg) and arenaviruses (Lassa and Junín viruses), are serious human pathogens for which there are currently no FDA approved therapeutics or vaccines. Importantly, transmission of these viruses, and specifically late steps of budding, critically depend upon host cell machinery. Consequently, strategies which target these mechanisms represent potential targets for broad spectrum host oriented therapeutics. An important cellular signal implicated previously in EBOV budding is calcium. Indeed, host cell calcium signals are increasingly being recognized to play a role in steps of entry, replication, and transmission for a range of viruses, but if and how filoviruses and arenaviruses mobilize calcium and the precise stage of virus transmission regulated by calcium have not been defined. Here we demonstrate that expression of matrix proteins from both filoviruses and arenaviruses triggers an increase in host cytoplasmic Ca2+ concentration by a mechanism that requires host Orai1 channels. Furthermore, we demonstrate that Orai1 regulates both VLP and infectious filovirus and arenavirus production and spread. Notably, suppression of the protein that triggers Orai activation (Stromal Interaction Molecule 1, STIM1) and genetic inactivation or pharmacological blockade of Orai1 channels inhibits VLP and infectious virus egress. These findings are highly significant as they expand our understanding of host mechanisms that may broadly control enveloped RNA virus budding, and they establish Orai and STIM1 as novel targets for broad-spectrum host-oriented therapeutics to combat these emerging BSL-4 pathogens and potentially other enveloped RNA viruses that bud via similar mechanisms. Filoviruses (Ebola and Marburg viruses) and arenaviruses (Lassa and Junín viruses) are high-priority pathogens that hijack host proteins and pathways to complete their replication cycles and spread from cell to cell. Here we provide genetic and pharmacological evidence to demonstrate that the host calcium channel protein Orai1 and ER calcium sensor protein STIM1 regulate efficient budding and spread of BSL-4 pathogens Ebola, Marburg, Lassa, and Junín viruses. Our findings are of broad significance as they provide new mechanistic insight into fundamental, immutable, and conserved mechanisms of hemorrhagic fever virus pathogenesis. Moreover, this strategy of targeting highly conserved host cellular protein(s) and mechanisms required by these viruses to complete their life cycle should elicit minimal drug resistance.
Collapse
|
45
|
Abstract
Store-operated calcium channels (SOCs) are a major pathway for calcium signaling in virtually all metozoan cells and serve a wide variety of functions ranging from gene expression, motility, and secretion to tissue and organ development and the immune response. SOCs are activated by the depletion of Ca(2+) from the endoplasmic reticulum (ER), triggered physiologically through stimulation of a diverse set of surface receptors. Over 15 years after the first characterization of SOCs through electrophysiology, the identification of the STIM proteins as ER Ca(2+) sensors and the Orai proteins as store-operated channels has enabled rapid progress in understanding the unique mechanism of store-operate calcium entry (SOCE). Depletion of Ca(2+) from the ER causes STIM to accumulate at ER-plasma membrane (PM) junctions where it traps and activates Orai channels diffusing in the closely apposed PM. Mutagenesis studies combined with recent structural insights about STIM and Orai proteins are now beginning to reveal the molecular underpinnings of these choreographic events. This review describes the major experimental advances underlying our current understanding of how ER Ca(2+) depletion is coupled to the activation of SOCs. Particular emphasis is placed on the molecular mechanisms of STIM and Orai activation, Orai channel properties, modulation of STIM and Orai function, pharmacological inhibitors of SOCE, and the functions of STIM and Orai in physiology and disease.
Collapse
Affiliation(s)
- Murali Prakriya
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois; and Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, California
| | - Richard S Lewis
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois; and Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, California
| |
Collapse
|
46
|
Xie J, Pan H, Yao J, Zhou Y, Han W. SOCE and cancer: Recent progress and new perspectives. Int J Cancer 2015; 138:2067-77. [PMID: 26355642 PMCID: PMC4764496 DOI: 10.1002/ijc.29840] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2015] [Accepted: 09/03/2015] [Indexed: 12/15/2022]
Abstract
Ca2+ acts as a universal and versatile second messenger in the regulation of a myriad of biological processes, including cell proliferation, differentiation, migration and apoptosis. Store‐operated Ca2+ entry (SOCE) mediated by ORAI and the stromal interaction molecule (STIM) constitutes one of the major routes of calcium entry in nonexcitable cells, in which the depletion of intracellular Ca2+ stores triggers activation of the endoplasmic reticulum (ER)‐resident Ca2+ sensor protein STIM to gate and open the ORAI Ca2+ channels in the plasma membrane (PM). Accumulating evidence indicates that SOCE plays critical roles in cancer cell proliferation, metastasis and tumor neovascularization, as well as in antitumor immunity. We summarize herein the recent advances in our understanding of the function of SOCE in various types of tumor cells, vascular endothelial cells and cells of the immune system. Finally, the therapeutic potential of SOCE inhibitors in the treatment of cancer is also discussed.
Collapse
Affiliation(s)
- Jiansheng Xie
- Laboratory of Cancer Biology, Institute of Clinical Science, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Hongming Pan
- Laboratory of Cancer Biology, Institute of Clinical Science, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.,Department of Medical Oncology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Junlin Yao
- Department of Medical Oncology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yubin Zhou
- Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M University Health Science Center, Houston, TX
| | - Weidong Han
- Laboratory of Cancer Biology, Institute of Clinical Science, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.,Department of Medical Oncology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|
47
|
The role of CRAC channel in asthma. Pulm Pharmacol Ther 2015; 35:67-74. [PMID: 26344428 DOI: 10.1016/j.pupt.2015.09.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Revised: 08/29/2015] [Accepted: 09/03/2015] [Indexed: 10/23/2022]
Abstract
Asthma is increasing globally and current treatments only manage a proportion of patients. There is an urgent need to develop new therapies. Lymphocytes are thought to play a central role in the pathophysiology of asthma through the production of inflammatory mediators. This is thought to be via the transcription factor NFAT which in turn can be activated through Ca(2+) release-activated Ca(2+) (CRAC) channels. The aim of this work was to investigate the role of CRAC in clinical and pre-clinical models of allergic asthma. Initial data demonstrated that the NFAT pathway is increased in stimulated lymphocytes from asthmatics. To confirm a role for the channel we showed that a selective inhibitor, Synta 66, blocked mediator production from lymphocytes. Synta 66 inhibited CD2/3/28 induced IL-2, IL-7, IL-13 & IFNΥ in a concentration-dependent manner in healthy and severe asthma donors, with over 60% inhibition observed for all cytokines. NFAT pathway was also increased in a pre-clinical asthma model. In this model we have demonstrated that CRAC played a central role in the airway inflammation and late asthmatic response (LAR). In conclusion, our data provides evidence that suggests targeting CRAC channels could be of therapeutic benefit for asthma sufferers.
Collapse
|
48
|
Jairaman A, Yamashita M, Schleimer RP, Prakriya M. Store-Operated Ca2+ Release-Activated Ca2+ Channels Regulate PAR2-Activated Ca2+ Signaling and Cytokine Production in Airway Epithelial Cells. THE JOURNAL OF IMMUNOLOGY 2015; 195:2122-33. [PMID: 26238490 DOI: 10.4049/jimmunol.1500396] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Accepted: 06/30/2015] [Indexed: 01/11/2023]
Abstract
The G-protein-coupled protease-activated receptor 2 (PAR2) plays an important role in the pathogenesis of various inflammatory and auto-immune disorders. In airway epithelial cells (AECs), stimulation of PAR2 by allergens and proteases triggers the release of a host of inflammatory mediators to regulate bronchomotor tone and immune cell recruitment. Activation of PAR2 turns on several cell signaling pathways of which the mobilization of cytosolic Ca(2+) is likely a critical but poorly understood event. In this study, we show that Ca(2+) release-activated Ca(2+) (CRAC) channels encoded by stromal interaction molecule 1 and Orai1 are a major route of Ca(2+) entry in primary human AECs and drive the Ca(2+) elevations seen in response to PAR2 activation. Activation of CRAC channels induces the production of several key inflammatory mediators from AECs including thymic stromal lymphopoietin, IL-6, and PGE2, in part through stimulation of gene expression via nuclear factor of activated T cells (NFAT). Furthermore, PAR2 stimulation induces the production of many key inflammatory mediators including PGE2, IL-6, IL-8, and GM-CSF in a CRAC channel-dependent manner. These findings indicate that CRAC channels are the primary mechanism for Ca(2+) influx in AECs and a vital checkpoint for the induction of PAR2-induced proinflammatory cytokines.
Collapse
Affiliation(s)
- Amit Jairaman
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611; and
| | - Megumi Yamashita
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611; and
| | - Robert P Schleimer
- Division of Allergy/Immunology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Murali Prakriya
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611; and
| |
Collapse
|
49
|
Cui R, Yan L, Luo Z, Guo X, Yan M. Blockade of store-operated calcium entry alleviates ethanol-induced hepatotoxicity via inhibiting apoptosis. Toxicol Appl Pharmacol 2015; 287:52-66. [DOI: 10.1016/j.taap.2015.05.014] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Revised: 05/16/2015] [Accepted: 05/26/2015] [Indexed: 11/15/2022]
|
50
|
Vacher P, Vacher AM, Pineau R, Latour S, Soubeyran I, Pangault C, Tarte K, Soubeyran P, Ducret T, Bresson-Bepoldin L. Localized Store-Operated Calcium Influx Represses CD95-Dependent Apoptotic Effects of Rituximab in Non-Hodgkin B Lymphomas. THE JOURNAL OF IMMUNOLOGY 2015. [PMID: 26202984 DOI: 10.4049/jimmunol.1402942] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
The anti-CD20 mAb, rituximab, is routinely used to treat B cell malignancies. However, a majority of patients relapse. An improvement in the complete response was obtained by combining rituximab with chemotherapy, at the cost of increased toxicity. We reported that rituximab induced the colocalization of both the Orai1 Ca(2+) release-activated Ca(2+) channel (CRAC) and the endoplasmic reticulum Ca(2+) sensor stromal interaction molecule 1 with CD20 and CD95 into a cluster, eliciting a polarized store-operated Ca(2+) entry (SOCE). We observed that blocking this Ca(2+) entry with downregulation of Orai1, pharmacological inhibitors, or reducing calcemia with hypocalcemic drugs sensitized human B lymphoma cell lines and primary human lymphoma cells to rituximab-induced apoptosis in vitro, and improved the antitumoral effect of rituximab in xenografted mice. This revealed that Ca(2+) entry exerted a negative feedback loop on rituximab-induced apoptosis, suggesting that associating CRAC channel inhibitors or hypocalcemic agents with rituximab may improve the treatment of patients with B cell malignancies. The calcium-dependent proteins involved in this process appear to vary according to the B lymphoma cell type, suggesting that CRAC-channel targeting is likely to be more efficient than calcium-dependent protein targeting.
Collapse
Affiliation(s)
- Pierre Vacher
- Institut Bergonié, Centre de Lutte contre le Cancer, F-33076 Bordeaux Cedex, France; Université de Bordeaux, F-33076 Bordeaux Cedex, France; INSERM U916 Validation et Identification de Nouvelles Cibles en Oncologie, F-33076 Bordeaux Cedex, France;
| | - Anne-Marie Vacher
- Institut Bergonié, Centre de Lutte contre le Cancer, F-33076 Bordeaux Cedex, France; Université de Bordeaux, F-33076 Bordeaux Cedex, France; INSERM U916 Validation et Identification de Nouvelles Cibles en Oncologie, F-33076 Bordeaux Cedex, France
| | - Raphael Pineau
- Animalerie Mutualisée, Université de Bordeaux, F33400 Talence, France
| | - Simon Latour
- Institut Bergonié, Centre de Lutte contre le Cancer, F-33076 Bordeaux Cedex, France; Université de Bordeaux, F-33076 Bordeaux Cedex, France; INSERM U916 Validation et Identification de Nouvelles Cibles en Oncologie, F-33076 Bordeaux Cedex, France
| | - Isabelle Soubeyran
- Institut Bergonié, Centre de Lutte contre le Cancer, F-33076 Bordeaux Cedex, France; Université de Bordeaux, F-33076 Bordeaux Cedex, France; INSERM U916 Validation et Identification de Nouvelles Cibles en Oncologie, F-33076 Bordeaux Cedex, France
| | - Celine Pangault
- INSERM, Unité Mixte de Recherche 917, F-35043 Rennes, France; Université de Rennes 1, F-35043 Rennes, France; Centre Hospitalier Universitaire Pontchaillou, F-35033 Rennes, France
| | - Karin Tarte
- INSERM, Unité Mixte de Recherche 917, F-35043 Rennes, France; Université de Rennes 1, F-35043 Rennes, France; Centre Hospitalier Universitaire Pontchaillou, F-35033 Rennes, France
| | - Pierre Soubeyran
- Institut Bergonié, Centre de Lutte contre le Cancer, F-33076 Bordeaux Cedex, France; Université de Bordeaux, F-33076 Bordeaux Cedex, France; INSERM U916 Validation et Identification de Nouvelles Cibles en Oncologie, F-33076 Bordeaux Cedex, France
| | - Thomas Ducret
- Université de Bordeaux, F-33076 Bordeaux Cedex, France; Centre de Recherche Cardio-Thoracique de Bordeaux, F-33076 Bordeaux Cedex, France; and INSERM, U1045, F-33076 Bordeaux Cedex, France
| | - Laurence Bresson-Bepoldin
- Institut Bergonié, Centre de Lutte contre le Cancer, F-33076 Bordeaux Cedex, France; Université de Bordeaux, F-33076 Bordeaux Cedex, France; INSERM U916 Validation et Identification de Nouvelles Cibles en Oncologie, F-33076 Bordeaux Cedex, France;
| |
Collapse
|