1
|
Lin YP, Scappini E, Mirams G, Tucker CJ, Parekh AB. CRAC channel activity pulsates during cytosolic Ca 2+ oscillations. J Biol Chem 2025:108519. [PMID: 40280418 DOI: 10.1016/j.jbc.2025.108519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Revised: 04/12/2025] [Accepted: 04/15/2025] [Indexed: 04/29/2025] Open
Abstract
Intracellular Ca2+ ions are used as second messengers throughout the phylogenetic tree. They are indispensable for diverse biological processes ranging from fertilization to cell death. In Metazoans, signaling information is conveyed via the amplitude, frequency and spatial profile of cytosolic Ca2+ oscillations. In non-excitable cells, these oscillations generally arise from regenerative release of Ca2+ from inositol 1,4,5-trisphosphate (InsP3)-sensitive intracellular stores, which are refilled by entry of Ca2+ through Ca2+ release-activated Ca2+ (CRAC) channels in the plasma membrane. However, the precise contribution of these store-operated CRAC channels to Ca2+ oscillations has remained controversial for decades. One view proposes that CRAC channels remain open throughout stimulation, functioning as the pacemaker in setting Ca2+ oscillation frequency. An alternative hypothesis is that channel activity oscillates in parallel with InsP3-driven regenerative Ca2+ release. Here, by tethering a genetically encoded Ca2+ indicator to the pore- forming subunit of the CRAC channel, Orai1, we distinguish between these hypotheses and demonstrate that CRAC channel activity fluctuates in phase with cytosolic Ca2+ oscillations during physiological levels of stimulation. We also find that spatially distinct CRAC channel clusters fire in a coordinated manner, revealing that CRAC channels are not independent units but might function in a synchronized manner to provide pulses of Ca2+ signal at the same time.
Collapse
Affiliation(s)
- Yu-Ping Lin
- Laboratory of Signal Transduction, National Institute of Environmental Health Sciences, National Institutes of Health, 111 TW Alexander Drive, Durham NC 27709 USA
| | - Erica Scappini
- Laboratory of Signal Transduction, National Institute of Environmental Health Sciences, National Institutes of Health, 111 TW Alexander Drive, Durham NC 27709 USA
| | - Gary Mirams
- Centre for Mathematical Medicine and Biology, School of Mathematical Sciences, University Park, University of Nottingham, NG7 2RD, UK
| | - Charles J Tucker
- Laboratory of Signal Transduction, National Institute of Environmental Health Sciences, National Institutes of Health, 111 TW Alexander Drive, Durham NC 27709 USA
| | - Anant B Parekh
- Laboratory of Signal Transduction, National Institute of Environmental Health Sciences, National Institutes of Health, 111 TW Alexander Drive, Durham NC 27709 USA
| |
Collapse
|
2
|
Zaccolo M, Kovanich D. Nanodomain cAMP signaling in cardiac pathophysiology: potential for developing targeted therapeutic interventions. Physiol Rev 2025; 105:541-591. [PMID: 39115424 PMCID: PMC7617275 DOI: 10.1152/physrev.00013.2024] [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: 03/29/2024] [Revised: 07/22/2024] [Accepted: 08/03/2024] [Indexed: 08/20/2024] Open
Abstract
The 3',5'-cyclic adenosine monophosphate (cAMP) mediates the effects of sympathetic stimulation on the rate and strength of cardiac contraction. Beyond this pivotal role, in cardiac myocytes cAMP also orchestrates a diverse array of reactions to various stimuli. To ensure specificity of response, the cAMP signaling pathway is intricately organized into multiple, spatially confined, subcellular domains, each governing a distinct cellular function. In this review, we describe the molecular components of the cAMP signaling pathway with a specific focus on adenylyl cyclases, A-kinase anchoring proteins, and phosphodiesterases. We discuss how they are organized inside the intracellular space and how they achieve exquisite regulation of signaling within nanometer-size domains. We delineate the key experimental findings that lead to the current model of compartmentalized cAMP signaling, and we offer an overview of our present understanding of how cAMP nanodomains are structured and regulated within cardiac myocytes. Furthermore, we discuss how compartmentalized cAMP signaling is affected in cardiac disease and consider the potential therapeutic opportunities arising from understanding such organization. By exploiting the nuances of compartmentalized cAMP signaling, novel and more effective therapeutic strategies for managing cardiac conditions may emerge. Finally, we highlight the unresolved questions and hurdles that must be addressed to translate these insights into interventions that may benefit patients.
Collapse
Affiliation(s)
- Manuela Zaccolo
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Duangnapa Kovanich
- Center for Vaccine Development, Institute of Molecular Biosciences, Mahidol University, Nakhon Pathom, Thailand
| |
Collapse
|
3
|
Macias-Díaz A, Nieto-Felipe J, Jardín I, Camello PJ, Martinez-Quintana EM, Salido GM, Smani T, Lopez JJ, Rosado JA. Filamin A C-terminal fragment modulates Orai1 expression by inhibition of protein degradation. Am J Physiol Cell Physiol 2025; 328:C657-C669. [PMID: 39764579 DOI: 10.1152/ajpcell.00745.2024] [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: 10/08/2024] [Revised: 12/16/2024] [Accepted: 12/30/2024] [Indexed: 02/05/2025]
Abstract
Filamin A (FLNA) is an actin-binding protein that has been reported to interact with STIM1 modulating the activation of Orai1 channels. Cleaving of FLNA by calpain leads to a C-terminal fragment that is involved in a variety of functional and pathological events, including pro-oncogenic activity in different types of cancer. Here, we show that full-length FLNA is downregulated in samples from patients with colon cancer as well as in the adenocarcinoma cell line HT-29. This is consistent with an increased calpain-dependent FLNA cleaving with enhanced expression of the C-terminal FLNA fragment accompanied by enhanced expression of Orai1 and STIM1, as well as store-operated Ca2+ entry (SOCE). To further explore the mechanism underlying the enhancement of SOCE by the C-terminal FLNA fragment, we expressed in HEK-293 cells the C-terminal FLNA region encompassing repeats 16-24 (FLNA16-24 fragment), which enhanced both Orai1 and STIM1 as well as SOCE. Transfection of the FLNA16-24 fragment attenuates Orai1 and STIM1 protein degradation, and, specifically, abrogates Orai1α lysosomal degradation and retains this channel in the plasma membrane. However, the C-terminal FLNA fragment did not induce a detectable modification in Orai1β degradation. Due to the relevance of SOCE in cell physiology, our results provide evidence of a novel mechanism for the regulation of Ca2+ influx with relevant pathophysiological implications.NOTE & NOTEWORTHY FLNA cleaving by calpain has been observed in a variety of tumoral, including prostate and colorectal cancer cells, as well as in nontumoral cells, leading to a C-terminal fragment encompassing repeats 16-24. Expression of the FLNA16-24 fragment in HEK-293 cells enhances Orai1 and STIM1 expression, as well as SOCE, a mechanism mediated by attenuation of Orai1α and STIM1 degradation, providing evidence for a novel mechanism for the regulation of SOCE in normal and malignant cells.
Collapse
Affiliation(s)
- Alvaro Macias-Díaz
- Department of Physiology (Cellular Physiology Research Group), Institute of Molecular Pathology Biomarkers (IMPB), University of Extremadura, Caceres, Spain
| | - Joel Nieto-Felipe
- Department of Physiology (Cellular Physiology Research Group), Institute of Molecular Pathology Biomarkers (IMPB), University of Extremadura, Caceres, Spain
| | - Isaac Jardín
- Department of Physiology (Cellular Physiology Research Group), Institute of Molecular Pathology Biomarkers (IMPB), University of Extremadura, Caceres, Spain
| | - Pedro J Camello
- Department of Physiology (Cellular Physiology Research Group), Institute of Molecular Pathology Biomarkers (IMPB), University of Extremadura, Caceres, Spain
| | | | - Gines M Salido
- Department of Physiology (Cellular Physiology Research Group), Institute of Molecular Pathology Biomarkers (IMPB), University of Extremadura, Caceres, Spain
| | - Tarik Smani
- Group of Cardiovascular Pathophysiology, Institute of Biomedicine of Seville, University Hospital of Virgen del Rocío/University of Seville/CSIC, Seville, Spain
- Department of Medical Physiology and Biophysics, Faculty of Medicine, University of Seville, Seville, Spain
| | - Jose J Lopez
- Department of Physiology (Cellular Physiology Research Group), Institute of Molecular Pathology Biomarkers (IMPB), University of Extremadura, Caceres, Spain
| | - Juan A Rosado
- Department of Physiology (Cellular Physiology Research Group), Institute of Molecular Pathology Biomarkers (IMPB), University of Extremadura, Caceres, Spain
| |
Collapse
|
4
|
Courjaret RJ, Wagner II LE, Ammouri RR, Yule DI, Machaca K. Ca2+ tunneling architecture and function are important for secretion. J Cell Biol 2025; 224:e202402107. [PMID: 39499286 PMCID: PMC11540855 DOI: 10.1083/jcb.202402107] [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: 02/18/2024] [Revised: 06/29/2024] [Accepted: 10/10/2024] [Indexed: 11/07/2024] Open
Abstract
Ca2+ tunneling requires both store-operated Ca2+ entry (SOCE) and Ca2+ release from the endoplasmic reticulum (ER). Tunneling expands the SOCE microdomain through Ca2+ uptake by SERCA into the ER lumen where it diffuses and is released via IP3 receptors. In this study, using high-resolution imaging, we outline the spatial remodeling of the tunneling machinery (IP3R1; SERCA; PMCA; and Ano1 as an effector) relative to STIM1 in response to store depletion. We show that these modulators redistribute to distinct subdomains laterally at the plasma membrane (PM) and axially within the cortical ER. To functionally define the role of Ca2+ tunneling, we engineered a Ca2+ tunneling attenuator (CaTAr) that blocks tunneling without affecting Ca2+ release or SOCE. CaTAr inhibits Cl- secretion in sweat gland cells and reduces sweating in vivo in mice, showing that Ca2+ tunneling is important physiologically. Collectively our findings argue that Ca2+ tunneling is a fundamental Ca2+ signaling modality.
Collapse
Affiliation(s)
- Raphael J. Courjaret
- Research Department, Ca Signaling Group, Weill Cornell Medicine Qatar, Qatar Foundation, Education City, Qatar
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
| | - Larry E. Wagner II
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY, USA
| | - Rahaf R. Ammouri
- Research Department, Ca Signaling Group, Weill Cornell Medicine Qatar, Qatar Foundation, Education City, Qatar
| | - David I. Yule
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY, USA
| | - Khaled Machaca
- Research Department, Ca Signaling Group, Weill Cornell Medicine Qatar, Qatar Foundation, Education City, Qatar
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
| |
Collapse
|
5
|
Nieto‐Felipe J, Macias‐Díaz A, Jimenez‐Velarde V, Lopez JJ, Salido GM, Smani T, Jardin I, Rosado JA. Feedback modulation of Orai1α and Orai1β protein content mediated by STIM proteins. J Cell Physiol 2025; 240:e31450. [PMID: 39359018 PMCID: PMC11730744 DOI: 10.1002/jcp.31450] [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: 05/18/2024] [Revised: 09/03/2024] [Accepted: 09/20/2024] [Indexed: 10/04/2024]
Abstract
Store-operated Ca2+ entry is a mechanism controlled by the filling state of the intracellular Ca2+ stores, predominantly the endoplasmic reticulum (ER), where ER-resident proteins STIM1 and STIM2 orchestrate the activation of Orai channels in the plasma membrane, and Orai1 playing a predominant role. Two forms of Orai1, Orai1α and Orai1β, have been identified, which arises the question whether they are equally regulated by STIM proteins. We demonstrate that STIM1 preferentially activates Orai1α over STIM2, yet both STIM proteins similarly activate Orai1β. Under resting conditions, there is a pronounced association between STIM2 and Orai1α. STIM1 and STIM2 are also shown to influence the protein levels of the Orai1 variants, independently of Ca2+ influx, via lysosomal degradation. Interestingly, Orai1α and Orai1β appear to selectively regulate the protein level of STIM1, but not STIM2. These observations offer crucial insights into the regulatory dynamics between STIM proteins and Orai1 variants, enhancing our understanding of the intricate processes that fine-tune intracellular Ca2+ signaling.
Collapse
Affiliation(s)
- Joel Nieto‐Felipe
- Department of Physiology (Cellular Physiology Research Group)Institute of Molecular Pathology Biomarkers (IMPB), University of ExtremaduraCaceresSpain
| | - Alvaro Macias‐Díaz
- Department of Physiology (Cellular Physiology Research Group)Institute of Molecular Pathology Biomarkers (IMPB), University of ExtremaduraCaceresSpain
| | - Vanesa Jimenez‐Velarde
- Department of Physiology (Cellular Physiology Research Group)Institute of Molecular Pathology Biomarkers (IMPB), University of ExtremaduraCaceresSpain
| | - Jose J. Lopez
- Department of Physiology (Cellular Physiology Research Group)Institute of Molecular Pathology Biomarkers (IMPB), University of ExtremaduraCaceresSpain
| | - Gines M. Salido
- Department of Physiology (Cellular Physiology Research Group)Institute of Molecular Pathology Biomarkers (IMPB), University of ExtremaduraCaceresSpain
| | - Tarik Smani
- Group of Cardiovascular PathophysiologyInstitute of Biomedicine of Seville, University Hospital of Virgen del Rocío/University of Seville/CSICSevilleSpain
- Department of Medical Physiology and BiophysicsFaculty of Medicine, University of SevilleSevilleSpain
| | - Isaac Jardin
- Department of Physiology (Cellular Physiology Research Group)Institute of Molecular Pathology Biomarkers (IMPB), University of ExtremaduraCaceresSpain
| | - Juan A. Rosado
- Department of Physiology (Cellular Physiology Research Group)Institute of Molecular Pathology Biomarkers (IMPB), University of ExtremaduraCaceresSpain
| |
Collapse
|
6
|
Möckl F, Kovacevic D, Werner R, Diercks BP, Weiß M. Visualization and Quantification of NFAT1 Translocation in T Cells. Methods Mol Biol 2025; 2904:11-19. [PMID: 40220223 DOI: 10.1007/978-1-0716-4414-0_2] [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] [Indexed: 04/14/2025]
Abstract
NFAT1 translocation to the nucleus and consequent IL-2 production in T cells can be used as a downstream read-out for initial T cell activation via the T cell receptor complex. Here, we describe a protocol for the detection of NFAT1 translocation using anti-CD3/anti-CD28 coated beads as a stimulus of primary murine CD4+ T cells by immunohistochemistry. Further, an open-source Python code is described to determine NFAT1 translocation.
Collapse
Affiliation(s)
- Franziska Möckl
- The Calcium Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Dejan Kovacevic
- The Calcium Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Institute for Applied Medical Informatics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Institute of Computational Neuroscience, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Center for Biomedical Artificial Intelligence (bAIome), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - René Werner
- Institute for Applied Medical Informatics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Institute of Computational Neuroscience, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Center for Biomedical Artificial Intelligence (bAIome), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Björn-Philip Diercks
- The Calcium Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| | - Mariella Weiß
- The Calcium Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
7
|
Parekh AB. House dust mite allergens, store-operated Ca 2+ channels and asthma. J Physiol 2024; 602:6021-6038. [PMID: 38054814 DOI: 10.1113/jp284931] [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: 08/11/2023] [Accepted: 10/26/2023] [Indexed: 12/07/2023] Open
Abstract
The house dust mite is the principal source of aero-allergen worldwide. Exposure to mite-derived allergens is associated with the development of asthma in susceptible individuals, and the majority of asthmatics are allergic to the mite. Mite-derived allergens are functionally diverse and activate multiple cell types within the lung that result in chronic inflammation. Allergens activate store-operated Ca2+ release-activated Ca2+ (CRAC) channels, which are widely expressed in multiple cell types within the lung that are associated with the pathogenesis of asthma. Opening of CRAC channels stimulates Ca2+-dependent transcription factors, including nuclear factor of activated T cells and nuclear factor-κB, which drive expression of a plethora of pro-inflammatory cytokines and chemokines that help to sustain chronic inflammation. Here, I describe drivers of asthma, properties of mite-derived allergens, how the allergens are recognized by cells, the signalling pathways used by the receptors and how these are transduced into functional effects, with a focus on CRAC channels. In vivo experiments that demonstrate the effectiveness of targeting CRAC channels as a potential new therapy for treating mite-induced asthma are also discussed, in tandem with other possible approaches.
Collapse
Affiliation(s)
- Anant B Parekh
- Laboratory of Signal Transduction, National Institute of Environmental Health Sciences, US National Institutes of Health, Department of Health and Human Services, Research Triangle Park, Durham, NC, USA
| |
Collapse
|
8
|
Gross S, Womer L, Kappes DJ, Soboloff J. Multifaceted control of T cell differentiation by STIM1. Trends Biochem Sci 2023; 48:1083-1097. [PMID: 37696713 PMCID: PMC10787584 DOI: 10.1016/j.tibs.2023.08.006] [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: 03/15/2023] [Revised: 08/08/2023] [Accepted: 08/18/2023] [Indexed: 09/13/2023]
Abstract
In T cells, stromal interaction molecule (STIM) and Orai are dispensable for conventional T cell development, but critical for activation and differentiation. This review focuses on novel STIM-dependent mechanisms for control of Ca2+ signals during T cell activation and its impact on mitochondrial function and transcriptional activation for control of T cell differentiation and function. We highlight areas that require further work including the roles of plasma membrane Ca2+ ATPase (PMCA) and partner of STIM1 (POST) in controlling Orai function. A major knowledge gap also exists regarding the independence of T cell development from STIM and Orai, despite compelling evidence that it requires Ca2+ signals. Resolving these and other outstanding questions ensures that the field will remain active for many years to come.
Collapse
Affiliation(s)
- Scott Gross
- Fels Cancer Institute for Personalized Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, 19140, USA
| | - Lauren Womer
- Fels Cancer Institute for Personalized Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, 19140, USA
| | | | - Jonathan Soboloff
- Fels Cancer Institute for Personalized Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, 19140, USA; Department of Cancer and Cellular Biology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, 19140, USA.
| |
Collapse
|
9
|
Weiß M, Hernandez LC, Gil Montoya DC, Löhndorf A, Krüger A, Kopdag M, Uebler L, Landwehr M, Nawrocki M, Huber S, Woelk LM, Werner R, Failla AV, Flügel A, Dupont G, Guse AH, Diercks BP. Adhesion to laminin-1 and collagen IV induces the formation of Ca 2+ microdomains that sensitize mouse T cells for activation. Sci Signal 2023; 16:eabn9405. [PMID: 37339181 DOI: 10.1126/scisignal.abn9405] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2022] [Accepted: 05/31/2023] [Indexed: 06/22/2023]
Abstract
During an immune response, T cells migrate from blood vessel walls into inflamed tissues by migrating across the endothelium and through extracellular matrix (ECM). Integrins facilitate T cell binding to endothelial cells and ECM proteins. Here, we report that Ca2+ microdomains observed in the absence of T cell receptor (TCR)/CD3 stimulation are initial signaling events triggered by adhesion to ECM proteins that increase the sensitivity of primary murine T cells to activation. Adhesion to the ECM proteins collagen IV and laminin-1 increased the number of Ca2+ microdomains in a manner dependent on the kinase FAK, phospholipase C (PLC), and all three inositol 1,4,5-trisphosphate receptor (IP3R) subtypes and promoted the nuclear translocation of the transcription factor NFAT-1. Mathematical modeling predicted that the formation of adhesion-dependent Ca2+ microdomains required the concerted activity of two to six IP3Rs and ORAI1 channels to achieve the increase in the Ca2+ concentration in the ER-plasma membrane junction that was observed experimentally and that required SOCE. Further, adhesion-dependent Ca2+ microdomains were important for the magnitude of the TCR-induced activation of T cells on collagen IV as assessed by the global Ca2+ response and NFAT-1 nuclear translocation. Thus, adhesion to collagen IV and laminin-1 sensitizes T cells through a mechanism involving the formation of Ca2+ microdomains, and blocking this low-level sensitization decreases T cell activation upon TCR engagement.
Collapse
Affiliation(s)
- Mariella Weiß
- Calcium Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Lola C Hernandez
- Calcium Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Diana C Gil Montoya
- Calcium Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Anke Löhndorf
- Calcium Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Aileen Krüger
- Calcium Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Miriam Kopdag
- Calcium Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Liana Uebler
- Calcium Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Marie Landwehr
- Calcium Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Mikolaj Nawrocki
- Section of Molecular Immunology und Gastroenterology, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Samuel Huber
- Section of Molecular Immunology und Gastroenterology, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Lena-Marie Woelk
- Department of Computational Neuroscience, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - René Werner
- Department of Computational Neuroscience, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Antonio V Failla
- Microscopy Imaging Facility (UMIF), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Alexander Flügel
- Institute for Neuroimmunology and Multiple Sclerosis Research, University Medical Centre Göttingen, 37075 Göttingen, Germany
| | - Geneviève Dupont
- Unité de Chronobiologie Théorique, Faculté des Sciences, CP231, Université Libre de Bruxelles (ULB), B-1050 Brussels, Belgium
| | - Andreas H Guse
- Calcium Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Björn-Philipp Diercks
- Calcium Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| |
Collapse
|
10
|
Nieto-Felipe J, Macias-Diaz A, Sanchez-Collado J, Berna-Erro A, Jardin I, Salido GM, Lopez JJ, Rosado JA. Role of Orai-family channels in the activation and regulation of transcriptional activity. J Cell Physiol 2023; 238:714-726. [PMID: 36952615 DOI: 10.1002/jcp.30971] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 01/12/2023] [Accepted: 01/27/2023] [Indexed: 03/25/2023]
Abstract
Store operated Ca2+ entry (SOCE) is a cornerstone for the maintenance of intracellular Ca2+ homeostasis and the regulation of a variety of cellular functions. SOCE is mediated by STIM and Orai proteins following the activation of inositol 1,4,5-trisphosphate receptors. Then, a reduction of the endoplasmic reticulum intraluminal Ca2+ concentration is sensed by STIM proteins, which undergo a conformational change and activate plasma membrane Ca2+ channels comprised by Orai proteins. STIM1/Orai-mediated Ca2+ signals are finely regulated and modulate the activity of different transcription factors, including certain isoforms of the nuclear factor of activated T-cells, the cAMP-response element binding protein, the nuclear factor κ-light chain-enhancer of activated B cells, c-fos, and c-myc. These transcription factors associate SOCE with a plethora of signaling events and cellular functions. Here we provide an overview of the current knowledge about the role of Orai channels in the regulation of transcription factors through Ca2+ -dependent signaling pathways.
Collapse
Affiliation(s)
- Joel Nieto-Felipe
- Departamento de Fisiología, Instituto Universitario de Biomarcadores de Patologías Moleculares, Universidad de Extremadura, Caceres, Spain
| | - Alvaro Macias-Diaz
- Departamento de Fisiología, Instituto Universitario de Biomarcadores de Patologías Moleculares, Universidad de Extremadura, Caceres, Spain
| | - Jose Sanchez-Collado
- Departamento de Fisiología, Instituto Universitario de Biomarcadores de Patologías Moleculares, Universidad de Extremadura, Caceres, Spain
| | - Alejandro Berna-Erro
- Departamento de Fisiología, Instituto Universitario de Biomarcadores de Patologías Moleculares, Universidad de Extremadura, Caceres, Spain
| | - Isaac Jardin
- Departamento de Fisiología, Instituto Universitario de Biomarcadores de Patologías Moleculares, Universidad de Extremadura, Caceres, Spain
| | - Gines M Salido
- Departamento de Fisiología, Instituto Universitario de Biomarcadores de Patologías Moleculares, Universidad de Extremadura, Caceres, Spain
| | - Jose J Lopez
- Departamento de Fisiología, Instituto Universitario de Biomarcadores de Patologías Moleculares, Universidad de Extremadura, Caceres, Spain
| | - Juan A Rosado
- Departamento de Fisiología, Instituto Universitario de Biomarcadores de Patologías Moleculares, Universidad de Extremadura, Caceres, Spain
| |
Collapse
|
11
|
Shin P, Pian Q, Ishikawa H, Hamanaka G, Mandeville ET, Shuzhen G, Buyin F, Alfadhel M, Allu SR, Şencan-Eğilmez I, Li B, Ran C, Vinogradov SA, Ayata C, Lo EH, Arai K, Devor A, Sakadžić S. Aerobic exercise reverses aging-induced depth-dependent decline in cerebral microcirculation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.12.528244. [PMID: 36824939 PMCID: PMC9949059 DOI: 10.1101/2023.02.12.528244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/19/2023]
Abstract
Aging is a major risk factor for cognitive impairment. Aerobic exercise benefits brain function and may promote cognitive health in older adults. However, underlying biological mechanisms across cerebral gray and white matter are poorly understood. Selective vulnerability of the white matter to small vessel disease and a link between white matter health and cognitive function suggests a potential role for responses in deep cerebral microcirculation. Here, we tested whether aerobic exercise modulates cerebral microcirculatory changes induced by aging. To this end, we carried out a comprehensive quantitative examination of changes in cerebral microvascular physiology in cortical gray and subcortical white matter in mice (3-6 vs. 19-21 months old), and asked whether and how exercise may rescue age-induced deficits. In the sedentary group, aging caused a more severe decline in cerebral microvascular perfusion and oxygenation in deep (infragranular) cortical layers and subcortical white matter compared with superficial (supragranular) cortical layers. Five months of voluntary aerobic exercise partly renormalized microvascular perfusion and oxygenation in aged mice in a depth-dependent manner, and brought these spatial distributions closer to those of young adult sedentary mice. These microcirculatory effects were accompanied by an improvement in cognitive function. Our work demonstrates the selective vulnerability of the deep cortex and subcortical white matter to aging-induced decline in microcirculation, as well as the responsiveness of these regions to aerobic exercise.
Collapse
Affiliation(s)
- Paul Shin
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
- Corresponding author:
| | - Qi Pian
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Hidehiro Ishikawa
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Gen Hamanaka
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Emiri T Mandeville
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Guo Shuzhen
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Fu Buyin
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Mohammed Alfadhel
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
- Department of Bioengineering, Northeastern University, Boston, MA, USA
| | - Srinivasa Rao Allu
- Department of Biochemistry and Biophysics, University of Pennsylvania, Philadelphia, PA, USA
- Department of Chemistry, University of Pennsylvania, Philadelphia, PA, USA
| | - Ikbal Şencan-Eğilmez
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
- Biophotonics Research Center, Mallinckrodt Institute of Radiology, Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Baoqiang Li
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
- Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, China
| | - Chongzhao Ran
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Sergei A Vinogradov
- Department of Biochemistry and Biophysics, University of Pennsylvania, Philadelphia, PA, USA
- Department of Chemistry, University of Pennsylvania, Philadelphia, PA, USA
| | - Cenk Ayata
- Neurovascular Research Laboratory, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
- Stroke Service, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Eng H Lo
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Ken Arai
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Anna Devor
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Sava Sakadžić
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| |
Collapse
|
12
|
Yuan X, Tang B, Chen Y, Zhou L, Deng J, Han L, Zhai Y, Zhou Y, Gill DL, Lu C, Wang Y. Celastrol inhibits store operated calcium entry and suppresses psoriasis. Front Pharmacol 2023; 14:1111798. [PMID: 36817139 PMCID: PMC9928759 DOI: 10.3389/fphar.2023.1111798] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 01/20/2023] [Indexed: 02/05/2023] Open
Abstract
Introduction: Psoriasis is an inflammatory autoimmune skin disease that is hard to cure and prone to relapse. Currently available global immunosuppressive agents for psoriasis may cause severe side effects, thus it is crucial to identify new therapeutic reagents and druggable signaling pathways for psoriasis. Methods: To check the effects of SOCE inhibitors on psoriasis, we used animal models, biochemical approaches, together with various imaging techniques, including calcium, confocal and FRET imaging. Results and discussion: Store operated calcium (Ca2+) entry (SOCE), mediated by STIM1 and Orai1, is crucial for the function of keratinocytes and immune cells, the two major players in psoriasis. Here we showed that a natural compound celastrol is a novel SOCE inhibitor, and it ameliorated the skin lesion and reduced PASI scores in imiquimod-induced psoriasis-like mice. Celastrol dose- and time-dependently inhibited SOCE in HEK cells and HaCaT cells, a keratinocyte cell line. Mechanistically, celastrol inhibited SOCE via its actions both on STIM1 and Orai1. It inhibited Ca2+ entry through constitutively-active Orai1 mutants independent of STIM1. Rather than blocking the conformational switch and oligomerization of STIM1 during SOCE activation, celastrol diminished the transition from oligomerized STIM1 into aggregates, thus locking STIM1 in a partially active state. As a result, it abolished the functional coupling between STIM1 and Orai1, diminishing SOCE signals. Overall, our findings identified a new SOCE inhibitor celastrol that suppresses psoriasis, suggesting that SOCE pathway may serve as a new druggable target for treating psoriasis.
Collapse
Affiliation(s)
- Xiaoman Yuan
- Beijing Key Laboratory of Gene Resource and Molecular Development, College of Life Sciences, Beijing Normal University, Beijing, China
| | - Bin Tang
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China,Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yilan Chen
- Beijing Key Laboratory of Gene Resource and Molecular Development, College of Life Sciences, Beijing Normal University, Beijing, China
| | - Lijuan Zhou
- Beijing Key Laboratory of Gene Resource and Molecular Development, College of Life Sciences, Beijing Normal University, Beijing, China
| | - Jingwen Deng
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China,Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Lin Han
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China,Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yonggong Zhai
- Beijing Key Laboratory of Gene Resource and Molecular Development, College of Life Sciences, Beijing Normal University, Beijing, China
| | - Yandong Zhou
- Department of Cellular and Molecular Physiology, The Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - Donald L. Gill
- Department of Cellular and Molecular Physiology, The Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - Chuanjian Lu
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China,Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou University of Chinese Medicine, Guangzhou, China,*Correspondence: Youjun Wang, ; Chuanjian Lu,
| | - Youjun Wang
- Beijing Key Laboratory of Gene Resource and Molecular Development, College of Life Sciences, Beijing Normal University, Beijing, China,Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, College of Life Sciences, Beijing Normal University, Beijing, China,*Correspondence: Youjun Wang, ; Chuanjian Lu,
| |
Collapse
|
13
|
The store-operated Ca 2+ channel Orai1α is required for agonist-evoked NF-κB activation by a mechanism dependent on PKCβ2. J Biol Chem 2023; 299:102882. [PMID: 36623731 PMCID: PMC9922819 DOI: 10.1016/j.jbc.2023.102882] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 12/30/2022] [Accepted: 12/31/2022] [Indexed: 01/09/2023] Open
Abstract
Store-operated Ca2+ entry is a ubiquitous mechanism for Ca2+ influx in mammalian cells that regulates a variety of physiological processes. The identification of two forms of Orai1, the predominant store-operated channel, Orai1α and Orai1β, raises the question whether they differentially regulate cell function. Orai1α is the full-length Orai1, containing 301 amino acids, whereas Orai1β lacks the N-terminal 63 amino acids. Here, using a combination of biochemistry and imaging combined with the use of human embryonic kidney 293 KO cells, missing the native Orai1, transfected with plasmids encoding for either Orai1α or Orai1β, we show that Orai1α plays a relevant role in agonist-induced NF-κB transcriptional activity. In contrast, functional Orai1β is not required for the activation of these transcription factors. The role of Orai1α in the activation of NF-κB is entirely dependent on Ca2+ influx and involves PKCβ activation. Our results indicate that Orai1α interacts with PKCβ2 by a mechanism involving the Orai1α exclusive AKAP79 association region, which strongly suggests a role for AKAP79 in this process. These findings provide evidence of the role of Orai1α in agonist-induced NF-κB transcriptional activity and reveal functional differences between Orai1 variants.
Collapse
|
14
|
Ulengin-Talkish I, Cyert MS. A cellular atlas of calcineurin signaling. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2023; 1870:119366. [PMID: 36191737 PMCID: PMC9948804 DOI: 10.1016/j.bbamcr.2022.119366] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Revised: 09/25/2022] [Accepted: 09/27/2022] [Indexed: 11/06/2022]
Abstract
Intracellular Ca2+ signals are temporally controlled and spatially restricted. Signaling occurs adjacent to sites of Ca2+ entry and/or release, where Ca2+-dependent effectors and their substrates co-localize to form signaling microdomains. Here we review signaling by calcineurin, the Ca2+/calmodulin regulated protein phosphatase and target of immunosuppressant drugs, Cyclosporin A and FK506. Although well known for its activation of the adaptive immune response via NFAT dephosphorylation, systematic mapping of human calcineurin substrates and regulators reveals unexpected roles for this versatile phosphatase throughout the cell. We discuss calcineurin function, with an emphasis on where signaling occurs and mechanisms that target calcineurin and its substrates to signaling microdomains, especially binding of cognate short linear peptide motifs (SLiMs). Calcineurin is ubiquitously expressed and regulates events at the plasma membrane, other intracellular membranes, mitochondria, the nuclear pore complex and centrosomes/cilia. Based on our expanding knowledge of localized CN actions, we describe a cellular atlas of Ca2+/calcineurin signaling.
Collapse
Affiliation(s)
| | - Martha S Cyert
- Department of Biology, Stanford University, Stanford, CA 94035, United States.
| |
Collapse
|
15
|
Jardin I, Berna-Erro A, Nieto-Felipe J, Macias A, Sanchez-Collado J, Lopez JJ, Salido GM, Rosado JA. Similarities and Differences between the Orai1 Variants: Orai1α and Orai1β. Int J Mol Sci 2022; 23:ijms232314568. [PMID: 36498894 PMCID: PMC9735889 DOI: 10.3390/ijms232314568] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 11/16/2022] [Accepted: 11/21/2022] [Indexed: 11/24/2022] Open
Abstract
Orai1, the first identified member of the Orai protein family, is ubiquitously expressed in the animal kingdom. Orai1 was initially characterized as the channel responsible for the store-operated calcium entry (SOCE), a major mechanism that allows cytosolic calcium concentration increments upon receptor-mediated IP3 generation, which results in intracellular Ca2+ store depletion. Furthermore, current evidence supports that abnormal Orai1 expression or function underlies several disorders. Orai1 is, together with STIM1, the key element of SOCE, conducting the Ca2+ release-activated Ca2+ (CRAC) current and, in association with TRPC1, the store-operated Ca2+ (SOC) current. Additionally, Orai1 is involved in non-capacitative pathways, as the arachidonate-regulated or LTC4-regulated Ca2+ channel (ARC/LRC), store-independent Ca2+ influx activated by the secretory pathway Ca2+-ATPase (SPCA2) and the small conductance Ca2+-activated K+ channel 3 (SK3). Furthermore, Orai1 possesses two variants, Orai1α and Orai1β, the latter lacking 63 amino acids in the N-terminus as compared to the full-length Orai1α form, which confers distinct features to each variant. Here, we review the current knowledge about the differences between Orai1α and Orai1β, the implications of the Ca2+ signals triggered by each variant, and their downstream modulatory effect within the cell.
Collapse
|
16
|
Nuanced Interactions between AKAP79 and STIM1 with Orai1 Ca 2+ Channels at Endoplasmic Reticulum-Plasma Membrane Junctions Sustain NFAT Activation. Mol Cell Biol 2022; 42:e0017522. [PMID: 36317924 PMCID: PMC9670898 DOI: 10.1128/mcb.00175-22] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
A-kinase anchoring protein 79 (AKAP79) is a human scaffolding protein that organizes Ca2+/calmodulin-dependent protein phosphatase calcineurin, calmodulin, cAMP-dependent protein kinase, protein kinase C, and the transcription factor nuclear factor of activated T cells (NFAT1) into a signalosome at the plasma membrane. Upon Ca2+ store depletion, AKAP79 interacts with the N-terminus of STIM1-gated Orai1 Ca2+ channels, enabling Ca2+ nanodomains to stimulate calcineurin. Calcineurin then dephosphorylates and activates NFAT1, which then translocates to the nucleus. A fundamental question is how signalosomes maintain long-term signaling when key effectors are released and therefore removed beyond the reach of the activating signal. Here, we show that the AKAP79-Orai1 interaction is considerably more transient than that of STIM1-Orai1. Free AKAP79, with calcineurin and NFAT1 in tow, is able to replace rapidly AKAP79 devoid of NFAT1 on Orai1, in the presence of continuous Ca2+ entry. We also show that Ca2+ nanodomains near Orai1 channels activate almost the entire cytosolic pool of NFAT1. Recycling of inactive NFAT1 from the cytoplasm to AKAP79 in the plasma membrane, coupled with the relatively weak interaction between AKAP79 and Orai1, maintain excitation-transcription coupling. By measuring rates for AKAP79-NFAT interaction, we formulate a mathematical model that simulates NFAT dynamics at the plasma membrane.
Collapse
|
17
|
Masson B, Le Ribeuz H, Sabourin J, Laubry L, Woodhouse E, Foster R, Ruchon Y, Dutheil M, Boët A, Ghigna MR, De Montpreville VT, Mercier O, Beech DJ, Benitah JP, Bailey MA, Humbert M, Montani D, Capuano V, Antigny F. Orai1 Inhibitors as Potential Treatments for Pulmonary Arterial Hypertension. Circ Res 2022; 131:e102-e119. [PMID: 36164973 DOI: 10.1161/circresaha.122.321041] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
BACKGROUND Pulmonary arterial hypertension (PAH) is characterized by progressive distal pulmonary artery (PA) obstruction, leading to right ventricular hypertrophy and failure. Exacerbated intracellular calcium (Ca2+) signaling contributes to abnormalities in PA smooth muscle cells (PASMCs), including aberrant proliferation, apoptosis resistance, exacerbated migration, and arterial contractility. Store-operated Ca2+ entry is involved in Ca2+ homeostasis in PASMCs, but its properties in PAH are unclear. METHODS Using a combination of Ca2+ imaging, molecular biology, in vitro, ex vivo, and in vivo approaches, we investigated the roles of the Orai1 SOC channel in PA remodeling in PAH and determined the consequences of pharmacological Orai1 inhibition in vivo using experimental models of pulmonary hypertension (PH). RESULTS Store-operated Ca2+ entry and Orai1 mRNA and protein were increased in human PASMCs (hPASMCs) from patients with PAH (PAH-hPASMCs). We found that MEK1/2 (mitogen-activated protein kinase kinase 1/2), NFAT (nuclear factor of activated T cells), and NFκB (nuclear factor-kappa B) contribute to the upregulation of Orai1 expression in PAH-hPASMCs. Using small interfering RNA (siRNA) and Orai1 inhibitors, we found that Orai1 inhibition reduced store-operated Ca2+ entry, mitochondrial Ca2+ uptake, aberrant proliferation, apoptosis resistance, migration, and excessive calcineurin activity in PAH-hPASMCs. Orai1 inhibitors reduced agonist-evoked constriction in human PAs. In experimental rat models of PH evoked by chronic hypoxia, monocrotaline, or Sugen/hypoxia, administration of Orai1 inhibitors (N-{4-[3,5-bis(Trifluoromethyl)-1H-pyrazol-1-yl]phenyl}-4-methyl-1,2,3-thiadiazole-5-carboxamide [BTP2], 4-(2,5-dimethoxyphenyl)-N-[(pyridin-4-yl)methyl]aniline [JPIII], or 5J4) protected against PH. CONCLUSIONS In human PAH and experimental PH, Orai1 expression and activity are increased. Orai1 inhibition normalizes the PAH-hPASMCs phenotype and attenuates PH in rat models. These results suggest that Orai1 should be considered as a relevant therapeutic target for PAH.
Collapse
Affiliation(s)
- Bastien Masson
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicêtre, France (B.M., H.L.R., L.L.., Y.R, M.D, A.B., M.-R.G., M.H., D.M., V.C., F.A.).,INSERM UMR_S 999 « Hypertension pulmonaire: Physiopathologie et Innovation Thérapeutique », Hôpital Marie Lannelongue, Le Plessis-Robinson, France. B.M., H.L.R., L.L.., Y.R, M.D, A.B., M.-R.G., M.H., D.M., V.C., F.A.)
| | - Hélène Le Ribeuz
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicêtre, France (B.M., H.L.R., L.L.., Y.R, M.D, A.B., M.-R.G., M.H., D.M., V.C., F.A.).,INSERM UMR_S 999 « Hypertension pulmonaire: Physiopathologie et Innovation Thérapeutique », Hôpital Marie Lannelongue, Le Plessis-Robinson, France. B.M., H.L.R., L.L.., Y.R, M.D, A.B., M.-R.G., M.H., D.M., V.C., F.A.)
| | - Jessica Sabourin
- Inserm, UMR-S 1180, Signalisation et Physiopathologie Cardiovasculaire, Université Paris-Saclay, Châtenay-Malabry, France (J.S., J.-P.B.)
| | - Loann Laubry
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicêtre, France (B.M., H.L.R., L.L.., Y.R, M.D, A.B., M.-R.G., M.H., D.M., V.C., F.A.).,INSERM UMR_S 999 « Hypertension pulmonaire: Physiopathologie et Innovation Thérapeutique », Hôpital Marie Lannelongue, Le Plessis-Robinson, France. B.M., H.L.R., L.L.., Y.R, M.D, A.B., M.-R.G., M.H., D.M., V.C., F.A.).,Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, United Kingdom (E.W., R.F., L.C., D.J.B., M.A.B.)
| | - Emily Woodhouse
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, United Kingdom (E.W., R.F., L.C., D.J.B., M.A.B.)
| | - Richard Foster
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, United Kingdom (E.W., R.F., L.C., D.J.B., M.A.B.)
| | - Yann Ruchon
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicêtre, France (B.M., H.L.R., L.L.., Y.R, M.D, A.B., M.-R.G., M.H., D.M., V.C., F.A.).,INSERM UMR_S 999 « Hypertension pulmonaire: Physiopathologie et Innovation Thérapeutique », Hôpital Marie Lannelongue, Le Plessis-Robinson, France. B.M., H.L.R., L.L.., Y.R, M.D, A.B., M.-R.G., M.H., D.M., V.C., F.A.).,Hôptal Marie Lannelongue, Groupe Hospitalier Paris Saint-Joseph, Le Plessis Robinson, France (Y.R., M.D., A.B., V.C.)
| | - Mary Dutheil
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicêtre, France (B.M., H.L.R., L.L.., Y.R, M.D, A.B., M.-R.G., M.H., D.M., V.C., F.A.).,INSERM UMR_S 999 « Hypertension pulmonaire: Physiopathologie et Innovation Thérapeutique », Hôpital Marie Lannelongue, Le Plessis-Robinson, France. B.M., H.L.R., L.L.., Y.R, M.D, A.B., M.-R.G., M.H., D.M., V.C., F.A.).,Hôptal Marie Lannelongue, Groupe Hospitalier Paris Saint-Joseph, Le Plessis Robinson, France (Y.R., M.D., A.B., V.C.)
| | - Angèle Boët
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicêtre, France (B.M., H.L.R., L.L.., Y.R, M.D, A.B., M.-R.G., M.H., D.M., V.C., F.A.).,INSERM UMR_S 999 « Hypertension pulmonaire: Physiopathologie et Innovation Thérapeutique », Hôpital Marie Lannelongue, Le Plessis-Robinson, France. B.M., H.L.R., L.L.., Y.R, M.D, A.B., M.-R.G., M.H., D.M., V.C., F.A.).,Hôptal Marie Lannelongue, Groupe Hospitalier Paris Saint-Joseph, Le Plessis Robinson, France (Y.R., M.D., A.B., V.C.)
| | - Maria-Rosa Ghigna
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicêtre, France (B.M., H.L.R., L.L.., Y.R, M.D, A.B., M.-R.G., M.H., D.M., V.C., F.A.).,INSERM UMR_S 999 « Hypertension pulmonaire: Physiopathologie et Innovation Thérapeutique », Hôpital Marie Lannelongue, Le Plessis-Robinson, France. B.M., H.L.R., L.L.., Y.R, M.D, A.B., M.-R.G., M.H., D.M., V.C., F.A.)
| | | | - Olaf Mercier
- Service de Chirurgie Thoracique, Vasculaire et Transplantation Cardio-Pulmonaire, Hôpital Marie Lannelongue, Groupe Hospitalier Paris Saint Joseph, Le Plessis Robinson, France (O.M.)
| | - David J Beech
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, United Kingdom (E.W., R.F., L.C., D.J.B., M.A.B.)
| | - Jean-Pierre Benitah
- Inserm, UMR-S 1180, Signalisation et Physiopathologie Cardiovasculaire, Université Paris-Saclay, Châtenay-Malabry, France (J.S., J.-P.B.)
| | - Marc A Bailey
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, United Kingdom (E.W., R.F., L.C., D.J.B., M.A.B.)
| | - Marc Humbert
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicêtre, France (B.M., H.L.R., L.L.., Y.R, M.D, A.B., M.-R.G., M.H., D.M., V.C., F.A.).,INSERM UMR_S 999 « Hypertension pulmonaire: Physiopathologie et Innovation Thérapeutique », Hôpital Marie Lannelongue, Le Plessis-Robinson, France. B.M., H.L.R., L.L.., Y.R, M.D, A.B., M.-R.G., M.H., D.M., V.C., F.A.).,Assistance Publique - Hôpitaux de Paris (AP-HP), Service de Pneumologie et Soins Intensifs Respiratoires, Centre de Référence de l'Hypertension Pulmonaire, Hôpital Bicêtre, Le Kremlin-Bicêtre, France (M.H., D.M.)
| | - David Montani
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicêtre, France (B.M., H.L.R., L.L.., Y.R, M.D, A.B., M.-R.G., M.H., D.M., V.C., F.A.).,INSERM UMR_S 999 « Hypertension pulmonaire: Physiopathologie et Innovation Thérapeutique », Hôpital Marie Lannelongue, Le Plessis-Robinson, France. B.M., H.L.R., L.L.., Y.R, M.D, A.B., M.-R.G., M.H., D.M., V.C., F.A.).,Assistance Publique - Hôpitaux de Paris (AP-HP), Service de Pneumologie et Soins Intensifs Respiratoires, Centre de Référence de l'Hypertension Pulmonaire, Hôpital Bicêtre, Le Kremlin-Bicêtre, France (M.H., D.M.)
| | - Véronique Capuano
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicêtre, France (B.M., H.L.R., L.L.., Y.R, M.D, A.B., M.-R.G., M.H., D.M., V.C., F.A.).,INSERM UMR_S 999 « Hypertension pulmonaire: Physiopathologie et Innovation Thérapeutique », Hôpital Marie Lannelongue, Le Plessis-Robinson, France. B.M., H.L.R., L.L.., Y.R, M.D, A.B., M.-R.G., M.H., D.M., V.C., F.A.).,Hôptal Marie Lannelongue, Groupe Hospitalier Paris Saint-Joseph, Le Plessis Robinson, France (Y.R., M.D., A.B., V.C.)
| | - Fabrice Antigny
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicêtre, France (B.M., H.L.R., L.L.., Y.R, M.D, A.B., M.-R.G., M.H., D.M., V.C., F.A.).,INSERM UMR_S 999 « Hypertension pulmonaire: Physiopathologie et Innovation Thérapeutique », Hôpital Marie Lannelongue, Le Plessis-Robinson, France. B.M., H.L.R., L.L.., Y.R, M.D, A.B., M.-R.G., M.H., D.M., V.C., F.A.)
| |
Collapse
|
18
|
Barak P, Kaur S, Scappini E, Tucker CJ, Parekh AB. Plasma Membrane Ca 2+ ATPase Activity Enables Sustained Store-operated Ca 2+ Entry in the Absence of a Bulk Cytosolic Ca 2+ Rise. FUNCTION 2022; 3:zqac040. [PMID: 38989036 PMCID: PMC11234650 DOI: 10.1093/function/zqac040] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 08/11/2022] [Accepted: 08/12/2022] [Indexed: 07/12/2024] Open
Abstract
In many cell types, the rise in cytosolic Ca2+ due to opening of Ca2+ release-activated Ca2+ (CRAC) channels drives a plethora of responses, including secretion, motility, energy production, and gene expression. The amplitude and time course of the cytosolic Ca2+ rise is shaped by the rates of Ca2+ entry into and removal from the cytosol. However, an extended bulk Ca2+ rise is toxic to cells. Here, we show that the plasma membrane Ca2+ ATPase (PMCA) pump plays a major role in preventing a prolonged cytosolic Ca2+ signal following CRAC channel activation. Ca2+ entry through CRAC channels leads to a sustained sub-plasmalemmal Ca2+ rise but bulk Ca2+ is kept low by the activity of PMCA4b. Despite the low cytosolic Ca2+, membrane permeability to Ca2+ is still elevated and Ca2+ continues to enter through CRAC channels. Ca2+-dependent NFAT activation, driven by Ca2+ nanodomains near the open channels, is maintained despite the return of bulk Ca2+ to near pre-stimulation levels. Our data reveal a central role for PMCA4b in determining the pattern of a functional Ca2+ signal and in sharpening local Ca2+ gradients near CRAC channels, whilst protecting cells from a toxic Ca2+ overload.
Collapse
Affiliation(s)
- Pradeep Barak
- Department of Physiology, Anatomy and Genetics, Oxford University, Oxford OX1 3PT, UK
- Oxford Nanoimaging, Linacre House, Jordan Hill Business Park Banbury Road, Oxford OX2 8TA, UK
| | - Suneet Kaur
- Laboratory of Signal Transduction, National Institute of Environmental Health Sciences, NIH, Research Triangle Park NC 27709, USA
| | - Erica Scappini
- Laboratory of Signal Transduction, National Institute of Environmental Health Sciences, NIH, Research Triangle Park NC 27709, USA
| | - Charles J Tucker
- Laboratory of Signal Transduction, National Institute of Environmental Health Sciences, NIH, Research Triangle Park NC 27709, USA
| | - Anant B Parekh
- Department of Physiology, Anatomy and Genetics, Oxford University, Oxford OX1 3PT, UK
- Laboratory of Signal Transduction, National Institute of Environmental Health Sciences, NIH, Research Triangle Park NC 27709, USA
| |
Collapse
|
19
|
STIM Proteins and Regulation of SOCE in ER-PM Junctions. Biomolecules 2022; 12:biom12081152. [PMID: 36009047 PMCID: PMC9405863 DOI: 10.3390/biom12081152] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 08/12/2022] [Accepted: 08/15/2022] [Indexed: 11/30/2022] Open
Abstract
ER-PM junctions are membrane contact sites formed by the endoplasmic reticulum (ER) and plasma membrane (PM) in close apposition together. The formation and stability of these junctions are dependent on constitutive and dynamic enrichment of proteins, which either contribute to junctional stability or modulate the lipid levels of both ER and plasma membranes. The ER-PM junctions have come under much scrutiny recently as they serve as hubs for assembling the Ca2+ signaling complexes. This review summarizes: (1) key findings that underlie the abilities of STIM proteins to accumulate in ER-PM junctions; (2) the modulation of Orai/STIM complexes by other components found within the same junction; and (3) how Orai1 channel activation is coordinated and coupled with downstream signaling pathways.
Collapse
|
20
|
Augustynek B, Gyimesi G, Dernič J, Sallinger M, Albano G, Klesse GJ, Kandasamy P, Grabmayr H, Frischauf I, Fuster DG, Peinelt C, Hediger MA, Bhardwaj R. Discovery of novel gating checkpoints in the Orai1 calcium channel by systematic analysis of constitutively active mutants of its paralogs and orthologs. Cell Calcium 2022; 105:102616. [PMID: 35792401 DOI: 10.1016/j.ceca.2022.102616] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 06/17/2022] [Accepted: 06/20/2022] [Indexed: 11/24/2022]
Abstract
In humans, there are three paralogs of the Orai Ca2+ channel that form the core of the store-operated calcium entry (SOCE) machinery. While the STIM-mediated gating mechanism of Orai channels is still under active investigation, several artificial and natural variants are known to cause constitutive activity of the human Orai1 channel. Surprisingly, little is known about the conservation of the gating checkpoints among the different human Orai paralogs and orthologs in other species. In our work, we show that the mutation corresponding to the activating mutation H134A in transmembrane helix 2 (TM2) of human Orai1 also activates Orai2 and Orai3, likely via a similar mechanism. However, this cross-paralog conservation does not apply to the "ANSGA" nexus mutations in TM4 of human Orai1, which is reported to mimic the STIM1-activated state of the channel. In investigating the mechanistic background of these differences, we identified two positions, H171 and F246 in human Orai1, that are not conserved among paralogs and that seem to be crucial for the channel activation triggered by the "ANSGA" mutations in Orai1. However, mutations of the same residues still allow gating of Orai1 by STIM1, suggesting that the ANSGA mutant of Orai1 may not be a surrogate for the STIM1-activated state of the Orai1 channel. Our results shed new light on these important gating checkpoints and show that the gating mechanism of Orai channels is affected by multiple factors that are not necessarily conserved among orai homologs, such as the TM4-TM3 coupling.
Collapse
Affiliation(s)
- Bartłomiej Augustynek
- Membrane Transport Discovery Lab, Department of Nephrology and Hypertension and Department of Biomedical Research, Inselspital, University of Bern, Freiburgstrasse 15, CH-3010 Bern, Switzerland
| | - Gergely Gyimesi
- Membrane Transport Discovery Lab, Department of Nephrology and Hypertension and Department of Biomedical Research, Inselspital, University of Bern, Freiburgstrasse 15, CH-3010 Bern, Switzerland
| | - Jan Dernič
- Membrane Transport Discovery Lab, Department of Nephrology and Hypertension and Department of Biomedical Research, Inselspital, University of Bern, Freiburgstrasse 15, CH-3010 Bern, Switzerland
| | - Matthias Sallinger
- Institute of Biophysics, JKU Life Science Center, Johannes Kepler University Linz, A-4020 Linz, Austria
| | - Giuseppe Albano
- Membrane Transport Discovery Lab, Department of Nephrology and Hypertension and Department of Biomedical Research, Inselspital, University of Bern, Freiburgstrasse 15, CH-3010 Bern, Switzerland
| | - Gabriel J Klesse
- Membrane Transport Discovery Lab, Department of Nephrology and Hypertension and Department of Biomedical Research, Inselspital, University of Bern, Freiburgstrasse 15, CH-3010 Bern, Switzerland
| | - Palanivel Kandasamy
- Membrane Transport Discovery Lab, Department of Nephrology and Hypertension and Department of Biomedical Research, Inselspital, University of Bern, Freiburgstrasse 15, CH-3010 Bern, Switzerland
| | - Herwig Grabmayr
- Institute of Biophysics, JKU Life Science Center, Johannes Kepler University Linz, A-4020 Linz, Austria
| | - Irene Frischauf
- Institute of Biophysics, JKU Life Science Center, Johannes Kepler University Linz, A-4020 Linz, Austria
| | - Daniel G Fuster
- Membrane Transport Discovery Lab, Department of Nephrology and Hypertension and Department of Biomedical Research, Inselspital, University of Bern, Freiburgstrasse 15, CH-3010 Bern, Switzerland
| | - Christine Peinelt
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bühlstrasse 28, 3012 Bern, Switzerland
| | - Matthias A Hediger
- Membrane Transport Discovery Lab, Department of Nephrology and Hypertension and Department of Biomedical Research, Inselspital, University of Bern, Freiburgstrasse 15, CH-3010 Bern, Switzerland.
| | - Rajesh Bhardwaj
- Membrane Transport Discovery Lab, Department of Nephrology and Hypertension and Department of Biomedical Research, Inselspital, University of Bern, Freiburgstrasse 15, CH-3010 Bern, Switzerland; Current address: Signal Transduction Laboratory, National Institute of Environmental Health Sciences, NIH, 111 TW Alexander Drive, NC 27709, USA.
| |
Collapse
|
21
|
Maltan L, Andova AM, Derler I. The Role of Lipids in CRAC Channel Function. Biomolecules 2022; 12:biom12030352. [PMID: 35327543 PMCID: PMC8944985 DOI: 10.3390/biom12030352] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 02/12/2022] [Accepted: 02/20/2022] [Indexed: 11/28/2022] Open
Abstract
The composition and dynamics of the lipid membrane define the physical properties of the bilayer and consequently affect the function of the incorporated membrane transporters, which also applies for the prominent Ca2+ release-activated Ca2+ ion channel (CRAC). This channel is activated by receptor-induced Ca2+ store depletion of the endoplasmic reticulum (ER) and consists of two transmembrane proteins, STIM1 and Orai1. STIM1 is anchored in the ER membrane and senses changes in the ER luminal Ca2+ concentration. Orai1 is the Ca2+-selective, pore-forming CRAC channel component located in the plasma membrane (PM). Ca2+ store-depletion of the ER triggers activation of STIM1 proteins, which subsequently leads to a conformational change and oligomerization of STIM1 and its coupling to as well as activation of Orai1 channels at the ER-PM contact sites. Although STIM1 and Orai1 are sufficient for CRAC channel activation, their efficient activation and deactivation is fine-tuned by a variety of lipids and lipid- and/or ER-PM junction-dependent accessory proteins. The underlying mechanisms for lipid-mediated CRAC channel modulation as well as the still open questions, are presented in this review.
Collapse
|
22
|
Bortolin A, Neto E, Lamghari M. Calcium Signalling in Breast Cancer Associated Bone Pain. Int J Mol Sci 2022; 23:ijms23031902. [PMID: 35163823 PMCID: PMC8836937 DOI: 10.3390/ijms23031902] [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: 02/01/2022] [Accepted: 02/03/2022] [Indexed: 02/05/2023] Open
Abstract
Calcium (Ca2+) is involved as a signalling mediator in a broad variety of physiological processes. Some of the fastest responses in human body like neuronal action potential firing, to the slowest gene transcriptional regulation processes are controlled by pathways involving calcium signalling. Under pathological conditions these mechanisms are also involved in tumoral cells reprogramming, resulting in the altered expression of genes associated with cell proliferation, metastatisation and homing to the secondary metastatic site. On the other hand, calcium exerts a central function in nociception, from cues sensing in distal neurons, to signal modulation and interpretation in the central nervous system leading, in pathological conditions, to hyperalgesia, allodynia and pain chronicization. It is well known the relationship between cancer and pain when tumoral metastatic cells settle in the bones, especially in late breast cancer stage, where they alter the bone micro-environment leading to bone lesions and resulting in pain refractory to the conventional analgesic therapies. The purpose of this review is to address the Ca2+ signalling mechanisms involved in cancer cell metastatisation as well as the function of the same signalling tools in pain regulation and transmission. Finally, the possible interactions between these two cells types cohabiting the same Ca2+ rich environment will be further explored attempting to highlight new possible therapeutical targets.
Collapse
Affiliation(s)
- Andrea Bortolin
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 280, 4200-135 Porto, Portugal; (A.B.); (E.N.)
- INEB—Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 280, 4200-135 Porto, Portugal
- FEUP—Faculdade de Engenharia da Universidade do Porto, Rua Dr. Roberto Frias s/n, 4200-465 Porto, Portugal
| | - Estrela Neto
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 280, 4200-135 Porto, Portugal; (A.B.); (E.N.)
- INEB—Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 280, 4200-135 Porto, Portugal
| | - Meriem Lamghari
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 280, 4200-135 Porto, Portugal; (A.B.); (E.N.)
- INEB—Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 280, 4200-135 Porto, Portugal
- Correspondence:
| |
Collapse
|
23
|
Liu W, Ren D, Xiong W, Jin X, Zhu L. A novel FBW7/NFAT1 axis regulates cancer immunity in sunitinib-resistant renal cancer by inducing PD-L1 expression. J Exp Clin Cancer Res 2022; 41:38. [PMID: 35081978 PMCID: PMC8790872 DOI: 10.1186/s13046-022-02253-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 01/12/2022] [Indexed: 01/11/2023] Open
Abstract
Background Tyrosine kinase inhibitors (TKIs) alone and in combination with immune checkpoint inhibitors (ICIs) have been shown to be beneficial for the survival of metastatic renal cell carcinoma (mRCC) patients, but resistance to targeted therapy and ICIs is common in the clinic. Understanding the underlying mechanism is critical for further prolonging the survival of renal cancer patients. Nuclear factor of activated T cell 1 (NFAT1) is expressed in immune and nonimmune cells, and the dysregulation of NFAT1 contributes to the progression of various type of malignant tumors. However, the specific role of NFAT1 in RCC is elusive. As a regulator of the immune response, we would like to systemically study the role of NFAT1 in RCC. Methods TCGA-KIRC dataset analysis, Western blot analysis and RT-qPCR analysis was used to determine the clinic-pathological characteristic of NFAT1 in RCC. CCK-8 assays, colony formation assays and xenograft assays were performed to examine the biological role of NFAT1 in renal cancer cells. RNA-seq analysis was used to examine the pathways changed after NFAT1 silencing. ChIP-qPCR, coimmunoprecipitation analysis, Western blot analysis and RT-qPCR analysis were applied to explore the mechanism by NAFT1 was regulated in the renal cancer cells. Results In our study, we found that NFAT1 was abnormally overexpressed in RCC and that NFAT1 overexpression was associated with an unfavorable prognosis. Then, we showed that NFAT1 enhanced tumor growth and regulated the immune response by increasing PD-L1 expression in RCC. In addition, we demonstrated that NFAT1 was stabilized in sunitinib-resistant RCC via hyperactivation of the PI3K/AKT/GSK-3β signaling pathway. Furthermore, our study indicated that downregulation of the expression of FBW7, which promotes NFAT1 degradation, was induced by FOXA1 and SETD2 in sunitinib-resistant RCC. Finally, FBW7 was found to contribute to modulating the immune response in RCC. Conclusions Our data reveal a novel role for the FBW7/NFAT1 axis in the RCC response to TKIs and ICIs. NFAT1 and its associated signaling pathway might be therapeutic targets for RCC treatment, especially when combined with ICIs and/or TKIs. Supplementary Information The online version contains supplementary material available at 10.1186/s13046-022-02253-0.
Collapse
|
24
|
Humer C, Romanin C, Höglinger C. Highlighting the Multifaceted Role of Orai1 N-Terminal- and Loop Regions for Proper CRAC Channel Functions. Cells 2022; 11:371. [PMID: 35159181 PMCID: PMC8834118 DOI: 10.3390/cells11030371] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/14/2022] [Accepted: 01/15/2022] [Indexed: 11/16/2022] Open
Abstract
Orai1, the Ca2+-selective pore in the plasma membrane, is one of the key components of the Ca2+release-activated Ca2+ (CRAC) channel complex. Activated by the Ca2+ sensor in the endoplasmic reticulum (ER) membrane, stromal interaction molecule 1 (STIM1), via direct interaction when ER luminal Ca2+ levels recede, Orai1 helps to maintain Ca2+ homeostasis within a cell. It has already been proven that the C-terminus of Orai1 is indispensable for channel activation. However, there is strong evidence that for CRAC channels to function properly and maintain all typical hallmarks, such as selectivity and reversal potential, additional parts of Orai1 are needed. In this review, we focus on these sites apart from the C-terminus; namely, the second loop and N-terminus of Orai1 and on their multifaceted role in the functioning of CRAC channels.
Collapse
Affiliation(s)
| | | | - Carmen Höglinger
- Institute of Biophysics, Johannes Kepler University Linz, Gruberstrasse 40, 4020 Linz, Austria; (C.H.); (C.R.)
| |
Collapse
|
25
|
Sanchez-Collado J, Lopez JJ, Jardin I, Berna-Erro A, Camello PJ, Cantonero C, Smani T, Salido GM, Rosado JA. Orai1α, but not Orai1β, co-localizes with TRPC1 and is required for its plasma membrane location and activation in HeLa cells. Cell Mol Life Sci 2022; 79:33. [PMID: 34988680 PMCID: PMC8732813 DOI: 10.1007/s00018-021-04098-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 12/02/2021] [Accepted: 12/15/2021] [Indexed: 12/15/2022]
Abstract
The identification of two variants of the canonical pore-forming subunit of the Ca2+ release-activated Ca2+ (CRAC) channel Orai1, Orai1α and Orai1β, in mammalian cells arises the question whether they exhibit different functional characteristics. Orai1α and Orai1β differ in the N-terminal 63 amino acids, exclusive of Orai1α, and show different sensitivities to Ca2+-dependent inactivation, as well as distinct ability to form arachidonate-regulated channels. We have evaluated the role of both Orai1 variants in the activation of TRPC1 in HeLa cells. We found that Orai1α and Orai1β are required for the maintenance of regenerative Ca2+ oscillations, while TRPC1 plays a role in agonist-induced Ca2+ influx but is not essential for Ca2+ oscillations. Using APEX2 proximity labeling, co-immunoprecipitation and the fluorescence of G-GECO1.2 fused to Orai1α our results indicate that agonist stimulation and Ca2+ store depletion enhance Orai1α–TRPC1 interaction. Orai1α is essential for TRPC1 plasma membrane location and activation. Thus, TRPC1 function in HeLa cells depends on Ca2+ influx through Orai1α exclusively.
Collapse
Affiliation(s)
- Jose Sanchez-Collado
- Department of Physiology (Cellular Physiology Research Group), Institute of Molecular Pathology Biomarkers (IMPB), University of Extremadura, 10003, Caceres, Spain
| | - Jose J Lopez
- Department of Physiology (Cellular Physiology Research Group), Institute of Molecular Pathology Biomarkers (IMPB), University of Extremadura, 10003, Caceres, Spain.
| | - Isaac Jardin
- Department of Physiology (Cellular Physiology Research Group), Institute of Molecular Pathology Biomarkers (IMPB), University of Extremadura, 10003, Caceres, Spain
| | - Alejandro Berna-Erro
- Department of Physiology (Cellular Physiology Research Group), Institute of Molecular Pathology Biomarkers (IMPB), University of Extremadura, 10003, Caceres, Spain
| | - Pedro J Camello
- Department of Physiology, (Smooth Muscle Physiology Research Group), Institute of Molecular Pathology Biomarkers, University of Extremadura, 10003, Caceres, Spain
| | - Carlos Cantonero
- Department of Physiology (Cellular Physiology Research Group), Institute of Molecular Pathology Biomarkers (IMPB), University of Extremadura, 10003, Caceres, Spain
| | - Tarik Smani
- Department of Medical Physiology and Biophysics, University of Seville, Seville, Spain.,Group of Cardiovascular Pathophysiology, Institute of Biomedicine of Seville, University Hospital of Virgen del Rocio/University of Seville/CSIC, Seville, Spain
| | - Gines M Salido
- Department of Physiology (Cellular Physiology Research Group), Institute of Molecular Pathology Biomarkers (IMPB), University of Extremadura, 10003, Caceres, Spain
| | - Juan A Rosado
- Department of Physiology (Cellular Physiology Research Group), Institute of Molecular Pathology Biomarkers (IMPB), University of Extremadura, 10003, Caceres, Spain.
| |
Collapse
|
26
|
Orai2 Modulates Store-Operated Ca 2+ Entry and Cell Cycle Progression in Breast Cancer Cells. Cancers (Basel) 2021; 14:cancers14010114. [PMID: 35008277 PMCID: PMC8749845 DOI: 10.3390/cancers14010114] [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: 11/25/2021] [Revised: 12/07/2021] [Accepted: 12/23/2021] [Indexed: 12/15/2022] Open
Abstract
Breast cancer is a heterogeneous disease from the histological and molecular expression point of view, and this heterogeneity determines cancer aggressiveness. Store-operated Ca2+ entry (SOCE), a major mechanism for Ca2+ entry in non-excitable cells, is significantly remodeled in cancer cells and plays an important role in the development and support of different cancer hallmarks. The store-operated CRAC (Ca2+ release-activated Ca2+) channels are predominantly comprised of Orai1 but the participation of Orai2 and Orai3 subunits has been reported to modulate the magnitude of Ca2+ responses. Here we provide evidence for a heterogeneous expression of Orai2 among different breast cancer cell lines. In the HER2 and triple negative breast cancer cell lines SKBR3 and BT20, respectively, where the expression of Orai2 was greater, Orai2 modulates the magnitude of SOCE and sustain Ca2+ oscillations in response to carbachol. Interestingly, in these cells Orai2 modulates the activation of NFAT1 and NFAT4 in response to high and low agonist concentrations. Finally, we have found that, in cells with high Orai2 expression, Orai2 knockdown leads to cell cycle arrest at the G0-G1 phase and decreases apoptosis resistance upon cisplatin treatment. Altogether, these findings indicate that, in breast cancer cells with a high Orai2 expression, Orai2 plays a relevant functional role in agonist-evoked Ca2+ signals, cell proliferation and apoptosis resistance.
Collapse
|
27
|
Knapp ML, Alansary D, Poth V, Förderer K, Sommer F, Zimmer D, Schwarz Y, Künzel N, Kless A, Machaca K, Helms V, Mühlhaus T, Schroda M, Lis A, Niemeyer BA. A longer isoform of Stim1 is a negative SOCE regulator but increases cAMP-modulated NFAT signaling. EMBO Rep 2021; 23:e53135. [PMID: 34942054 PMCID: PMC8892257 DOI: 10.15252/embr.202153135] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 11/23/2021] [Accepted: 12/07/2021] [Indexed: 11/23/2022] Open
Abstract
Alternative splicing is a potent modifier of protein function. Stromal interaction molecule 1 (Stim1) is the essential activator of store‐operated Ca2+ entry (SOCE) triggering activation of transcription factors. Here, we characterize Stim1A, a splice variant with an additional 31 amino acid domain inserted in frame within its cytosolic domain. Prominent expression of exon A is found in astrocytes, heart, kidney, and testes. Full‐length Stim1A functions as a dominant‐negative regulator of SOCE and ICRAC, facilitating sequence‐specific fast calcium‐dependent inactivation and destabilizing gating of Orai channels. Downregulation or absence of native Stim1A results in increased SOCE. Despite reducing SOCE, Stim1A leads to increased NFAT translocation. Differential proteomics revealed an interference of Stim1A with the cAMP‐SOCE crosstalk by altered modulation of phosphodiesterase 8 (PDE8), resulting in reduced cAMP degradation and increased PIP5K activity, facilitating NFAT activation. Our study uncovers a hitherto unknown mechanism regulating NFAT activation and indicates that cell‐type‐specific splicing of Stim1 is a potent means to regulate the NFAT signalosome and cAMP‐SOCE crosstalk.
Collapse
Affiliation(s)
- Mona L Knapp
- Molecular Biophysics, Saarland University, Homburg, Germany
| | - Dalia Alansary
- Molecular Biophysics, Saarland University, Homburg, Germany
| | - Vanessa Poth
- Molecular Biophysics, Saarland University, Homburg, Germany
| | | | - Frederik Sommer
- Molecular Biotechnology and Systems Biology, TU Kaiserslautern, Kaiserslautern, Germany
| | - David Zimmer
- Computational Systems Biology, TU Kaiserslautern, Kaiserslautern, Germany
| | - Yvonne Schwarz
- Molecular Neurophysiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), Saarland University, Homburg, Germany
| | - Nicolas Künzel
- Center for Bioinformatics, Saarland University, Saarbruecken, Germany
| | - Achim Kless
- Grünenthal Innovation, Drug Discovery Technologies, Grünenthal GmbH, Aachen, Germany
| | | | - Volkhard Helms
- Center for Bioinformatics, Saarland University, Saarbruecken, Germany
| | - Timo Mühlhaus
- Computational Systems Biology, TU Kaiserslautern, Kaiserslautern, Germany
| | - Michael Schroda
- Molecular Biotechnology and Systems Biology, TU Kaiserslautern, Kaiserslautern, Germany
| | - Annette Lis
- Biophysics, Saarland University, Homburg, Germany
| | | |
Collapse
|
28
|
Lu F, Li Y, Lin S, Cheng H, Yang S. Spatiotemporal regulation of store-operated calcium entry in cancer metastasis. Biochem Soc Trans 2021; 49:2581-2589. [PMID: 34854917 PMCID: PMC9436031 DOI: 10.1042/bst20210307] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 11/09/2021] [Accepted: 11/12/2021] [Indexed: 01/08/2023]
Abstract
The store-operated calcium (Ca2+) entry (SOCE) is the Ca2+ entry mechanism used by cells to replenish depleted Ca2+ store. The dysregulation of SOCE has been reported in metastatic cancer. It is believed that SOCE promotes migration and invasion by remodeling the actin cytoskeleton and cell adhesion dynamics. There is recent evidence supporting that SOCE is critical for the spatial and the temporal coding of Ca2+ signals in the cell. In this review, we critically examined the spatiotemporal control of SOCE signaling and its implication in the specificity and robustness of signaling events downstream of SOCE, with a focus on the spatiotemporal SOCE signaling during cancer cell migration, invasion and metastasis. We further discuss the limitation of our current understanding of SOCE in cancer metastasis and potential approaches to overcome such limitation.
Collapse
Affiliation(s)
- Fujian Lu
- State Key Laboratory of Membrane Biology, Institute of Molecular Medicine, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100871, China
- Department of Cardiology, Boston Children’s Hospital, 300 Longwood Ave, Boston, MA, 02115, USA
| | - Yunzhan Li
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, PA, 17033, United States
| | - Shengchen Lin
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, PA, 17033, United States
| | - Heping Cheng
- State Key Laboratory of Membrane Biology, Institute of Molecular Medicine, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100871, China
| | - Shengyu Yang
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, PA, 17033, United States
| |
Collapse
|
29
|
Tiffner A, Hopl V, Schober R, Sallinger M, Grabmayr H, Höglinger C, Fahrner M, Lunz V, Maltan L, Frischauf I, Krivic D, Bhardwaj R, Schindl R, Hediger MA, Derler I. Orai1 Boosts SK3 Channel Activation. Cancers (Basel) 2021; 13:6357. [PMID: 34944977 PMCID: PMC8699475 DOI: 10.3390/cancers13246357] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 12/13/2021] [Indexed: 12/20/2022] Open
Abstract
The interplay of SK3, a Ca2+ sensitive K+ ion channel, with Orai1, a Ca2+ ion channel, has been reported to increase cytosolic Ca2+ levels, thereby triggering proliferation of breast and colon cancer cells, although a molecular mechanism has remained elusive to date. We show in the current study, via heterologous protein expression, that Orai1 can enhance SK3 K+ currents, in addition to constitutively bound calmodulin (CaM). At low cytosolic Ca2+ levels that decrease SK3 K+ permeation, co-expressed Orai1 potentiates SK3 currents. This positive feedback mechanism of SK3 and Orai1 is enabled by their close co-localization. Remarkably, we discovered that loss of SK3 channel activity due to overexpressed CaM mutants could be restored by Orai1, likely via its interplay with the SK3-CaM binding site. Mapping for interaction sites within Orai1, we identified that the cytosolic strands and pore residues are critical for a functional communication with SK3. Moreover, STIM1 has a bimodal role in SK3-Orai1 regulation. Under physiological ionic conditions, STIM1 is able to impede SK3-Orai1 interplay by significantly decreasing their co-localization. Forced STIM1-Orai1 activity and associated Ca2+ influx promote SK3 K+ currents. The dynamic regulation of Orai1 to boost endogenous SK3 channels was also determined in the human prostate cancer cell line LNCaP.
Collapse
Affiliation(s)
- Adéla Tiffner
- JKU Life Science Center, Institute of Biophysics, Johannes Kepler University Linz, A-4020 Linz, Austria; (A.T.); (V.H.); (R.S.); (M.S.); (H.G.); (C.H.); (M.F.); (V.L.); (L.M.); (I.F.)
| | - Valentina Hopl
- JKU Life Science Center, Institute of Biophysics, Johannes Kepler University Linz, A-4020 Linz, Austria; (A.T.); (V.H.); (R.S.); (M.S.); (H.G.); (C.H.); (M.F.); (V.L.); (L.M.); (I.F.)
| | - Romana Schober
- JKU Life Science Center, Institute of Biophysics, Johannes Kepler University Linz, A-4020 Linz, Austria; (A.T.); (V.H.); (R.S.); (M.S.); (H.G.); (C.H.); (M.F.); (V.L.); (L.M.); (I.F.)
- Gottfried Schatz Research Centre, Medical University of Graz, A-8010 Graz, Austria; (D.K.); (R.S.)
| | - Matthias Sallinger
- JKU Life Science Center, Institute of Biophysics, Johannes Kepler University Linz, A-4020 Linz, Austria; (A.T.); (V.H.); (R.S.); (M.S.); (H.G.); (C.H.); (M.F.); (V.L.); (L.M.); (I.F.)
| | - Herwig Grabmayr
- JKU Life Science Center, Institute of Biophysics, Johannes Kepler University Linz, A-4020 Linz, Austria; (A.T.); (V.H.); (R.S.); (M.S.); (H.G.); (C.H.); (M.F.); (V.L.); (L.M.); (I.F.)
| | - Carmen Höglinger
- JKU Life Science Center, Institute of Biophysics, Johannes Kepler University Linz, A-4020 Linz, Austria; (A.T.); (V.H.); (R.S.); (M.S.); (H.G.); (C.H.); (M.F.); (V.L.); (L.M.); (I.F.)
| | - Marc Fahrner
- JKU Life Science Center, Institute of Biophysics, Johannes Kepler University Linz, A-4020 Linz, Austria; (A.T.); (V.H.); (R.S.); (M.S.); (H.G.); (C.H.); (M.F.); (V.L.); (L.M.); (I.F.)
| | - Victoria Lunz
- JKU Life Science Center, Institute of Biophysics, Johannes Kepler University Linz, A-4020 Linz, Austria; (A.T.); (V.H.); (R.S.); (M.S.); (H.G.); (C.H.); (M.F.); (V.L.); (L.M.); (I.F.)
| | - Lena Maltan
- JKU Life Science Center, Institute of Biophysics, Johannes Kepler University Linz, A-4020 Linz, Austria; (A.T.); (V.H.); (R.S.); (M.S.); (H.G.); (C.H.); (M.F.); (V.L.); (L.M.); (I.F.)
| | - Irene Frischauf
- JKU Life Science Center, Institute of Biophysics, Johannes Kepler University Linz, A-4020 Linz, Austria; (A.T.); (V.H.); (R.S.); (M.S.); (H.G.); (C.H.); (M.F.); (V.L.); (L.M.); (I.F.)
| | - Denis Krivic
- Gottfried Schatz Research Centre, Medical University of Graz, A-8010 Graz, Austria; (D.K.); (R.S.)
| | - Rajesh Bhardwaj
- Department of Nephrology and Hypertension, University of Bern, Inselspital, Freiburgstrasse 15, CH-3010 Bern, Switzerland; (R.B.); (M.A.H.)
- Department of Biomedical Research, University of Bern, Inselspital, Freiburgstrasse 15, CH-3010 Bern, Switzerland
| | - Rainer Schindl
- Gottfried Schatz Research Centre, Medical University of Graz, A-8010 Graz, Austria; (D.K.); (R.S.)
| | - Matthias A. Hediger
- Department of Nephrology and Hypertension, University of Bern, Inselspital, Freiburgstrasse 15, CH-3010 Bern, Switzerland; (R.B.); (M.A.H.)
- Department of Biomedical Research, University of Bern, Inselspital, Freiburgstrasse 15, CH-3010 Bern, Switzerland
| | - Isabella Derler
- JKU Life Science Center, Institute of Biophysics, Johannes Kepler University Linz, A-4020 Linz, Austria; (A.T.); (V.H.); (R.S.); (M.S.); (H.G.); (C.H.); (M.F.); (V.L.); (L.M.); (I.F.)
| |
Collapse
|
30
|
Bokhobza A, Ziental-Gelus N, Allart L, Iamshanova O, Vanden Abeele F. Impact of SOCE Abolition by ORAI1 Knockout on the Proliferation, Adhesion, and Migration of HEK-293 Cells. Cells 2021; 10:3016. [PMID: 34831241 PMCID: PMC8616168 DOI: 10.3390/cells10113016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 11/01/2021] [Accepted: 11/02/2021] [Indexed: 11/16/2022] Open
Abstract
Store-operated calcium entry (SOCE) provided through channels formed by ORAI proteins is a major regulator of several cellular processes. In immune cells, it controls fundamental processes such as proliferation, cell adhesion, and migration, while in cancer, SOCE and ORAI1 gene expression are dysregulated and lead to abnormal migration and/or cell proliferation. In the present study, we used the CRISPR/Cas9 technique to delete the ORAI1 gene and to identify its role in proliferative and migrative properties of the model cell line HEK-293. We showed that ORAI1 deletion greatly reduced SOCE. Thereby, we found that this decrease and the absence of ORAI1 protein did not affect HEK-293 proliferation. In addition, we determined that ORAI1 suppression did not affect adhesive properties but had a limited impact on HEK-293 migration. Overall, we showed that ORAI1 and SOCE are largely dispensable for cellular proliferation, migration, and cellular adhesion of HEK-293 cells. Thus, despite its importance in providing Ca2+ entry in non-excitable cells, our results indicate that the lack of SOCE does not deeply impact HEK-293 cells. This finding suggests the existence of compensatory mechanism enabling the maintenance of their physiological function.
Collapse
Affiliation(s)
- Alexandre Bokhobza
- Inserm U1003, Laboratory of Cell Physiology, Université de Lille, 59650 Villeneuve d’Ascq, France; (N.Z.-G.); (L.A.); (O.I.)
| | | | | | | | - Fabien Vanden Abeele
- Inserm U1003, Laboratory of Cell Physiology, Université de Lille, 59650 Villeneuve d’Ascq, France; (N.Z.-G.); (L.A.); (O.I.)
| |
Collapse
|
31
|
Voros O, Panyi G, Hajdu P. Immune Synapse Residency of Orai1 Alters Ca 2+ Response of T Cells. Int J Mol Sci 2021; 22:ijms222111514. [PMID: 34768945 PMCID: PMC8583858 DOI: 10.3390/ijms222111514] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 10/11/2021] [Accepted: 10/18/2021] [Indexed: 12/19/2022] Open
Abstract
CRAC, which plays important role in Ca2+-dependent T-lymphocyte activation, is composed of the ER-resident STIM1 and the plasma membrane Orai1 pore-forming subunit. Both accumulate at the immunological synapse (IS) between a T cell and an antigen-presenting cell (APC). We hypothesized that adapter/interacting proteins regulate Orai1 residence in the IS. We could show that mGFP-tagged Orai1-Full channels expressed in Jurkat cells had a biphasic IS-accumulation kinetics peaked at 15 min. To understand the background of Orai1 IS-redistribution we knocked down STIM1 and SAP97 (adaptor protein with a short IS-residency (15 min) and ability to bind Orai1 N-terminus): the mGFP-Orai1-Full channels kept on accumulating in the IS up to the 60th minute in the STIM1- and SAP97-lacking Jurkat cells. Deletion of Orai1 N terminus (mGFP-Orai1-Δ72) resulted in the same time course as described for STIM1/SAP97 knock-down cells. Ca2+-imaging of IS-engaged T-cells revealed that of Orai1 residency modifies the Ca2+-response: cells expressing mGFP-Orai1-Δ72 construct or mGFP-Orai1-Full in SAP-97 knock-down cells showed higher number of Ca2+-oscillation up to the 90th minute after IS formation. Overall, these data suggest that SAP97 may contribute to the short-lived IS-residency of Orai1 and binding of STIM1 to Orai1 N-terminus is necessary for SAP97-Orai1 interaction.
Collapse
Affiliation(s)
- Orsolya Voros
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Debrecen, Egyetem tér 1, 4032 Debrecen, Hungary; (O.V.); (G.P.)
| | - György Panyi
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Debrecen, Egyetem tér 1, 4032 Debrecen, Hungary; (O.V.); (G.P.)
| | - Péter Hajdu
- Department of Biophysics and Cell Biology, Faculty of Dentistry, University of Debrecen, Nagyerdei krt. 98, 4032 Debrecen, Hungary
- Correspondence: ; Tel.: +36-52-258603
| |
Collapse
|
32
|
Hofer AM. The Love Story between Orai Calcium Entry Channels and Adenylyl Cyclases Gets even more Complicated. FUNCTION (OXFORD, ENGLAND) 2021; 2:zqab044. [PMID: 35330952 PMCID: PMC8788800 DOI: 10.1093/function/zqab044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 08/18/2021] [Indexed: 01/07/2023]
|
33
|
Noyer L, Feske S. Straight from the channel's mouth: AKAP79 links Ca 2+ influx through ORAI1 to NFAT activation. Cell Calcium 2021; 99:102459. [PMID: 34481158 DOI: 10.1016/j.ceca.2021.102459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 08/19/2021] [Accepted: 08/23/2021] [Indexed: 11/19/2022]
Affiliation(s)
- Lucile Noyer
- Department of Pathology, New York University School of Medicine, 550 First Avenue, Smilow 510, New York, NY 10016, USA
| | - Stefan Feske
- Department of Pathology, New York University School of Medicine, 550 First Avenue, Smilow 510, New York, NY 10016, USA.
| |
Collapse
|
34
|
Kar P, Barak P, Zerio A, Lin YP, Parekh AJ, Watts VJ, Cooper DMF, Zaccolo M, Kramer H, Parekh AB. AKAP79 Orchestrates a Cyclic AMP Signalosome Adjacent to Orai1 Ca 2+ Channels. FUNCTION (OXFORD, ENGLAND) 2021; 2:zqab036. [PMID: 34458850 PMCID: PMC8394516 DOI: 10.1093/function/zqab036] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 07/16/2021] [Accepted: 07/27/2021] [Indexed: 01/12/2023]
Abstract
To ensure specificity of response, eukaryotic cells often restrict signalling molecules to sub-cellular regions. The Ca2+ nanodomain is a spatially confined signal that arises near open Ca2+ channels. Ca2+ nanodomains near store-operated Orai1 channels stimulate the protein phosphatase calcineurin, which activates the transcription factor NFAT1, and both enzyme and target are initially attached to the plasma membrane through the scaffolding protein AKAP79. Here, we show that a cAMP signalling nexus also forms adjacent to Orai1. Protein kinase A and phosphodiesterase 4, an enzyme that rapidly breaks down cAMP, both associate with AKAP79 and realign close to Orai1 after stimulation. PCR and mass spectrometry failed to show expression of Ca2+-activated adenylyl cyclase 8 in HEK293 cells, whereas the enzyme was observed in neuronal cell lines. FRET and biochemical measurements of bulk cAMP and protein kinase A activity consistently failed to show an increase in adenylyl cyclase activity following even a large rise in cytosolic Ca2+. Furthermore, expression of AKAP79-CUTie, a cAMP FRET sensor tethered to AKAP79, did not report a rise in cAMP after stimulation, despite AKAP79 association with Orai1. Hence, HEK293 cells do not express functional active Ca2+-activated adenylyl cyclases including adenylyl cyclase 8. Our results show that two ancient second messengers are independently generated in nanodomains close to Orai1 Ca2+ channels.
Collapse
Affiliation(s)
- Pulak Kar
- Department of Physiology, Anatomy and Genetics, University of Oxford, Parks Road, Oxford, OX1 3PT, UK
| | - Pradeep Barak
- Department of Physiology, Anatomy and Genetics, University of Oxford, Parks Road, Oxford, OX1 3PT, UK
| | - Anna Zerio
- Department of Physiology, Anatomy and Genetics, University of Oxford, Parks Road, Oxford, OX1 3PT, UK
| | - Yu-Ping Lin
- Department of Physiology, Anatomy and Genetics, University of Oxford, Parks Road, Oxford, OX1 3PT, UK,NIEHS/NIH, 111 TW Alexander Drive, Durham, NC 27709, USA
| | - Amy J Parekh
- Stoke Mandeville Hospital, Mandeville Road, Aylesbury, HP21 8AL, UK
| | - Val J Watts
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue Institute of Drug Discovery, Purdue Institute of Neuroscience, Purdue University, West Lafayette, IN 47907, USA
| | - Dermot M F Cooper
- Department of Pharmacology, Tennis Court Road, Cambridge CB2 1PD, UK
| | - Manuela Zaccolo
- Department of Physiology, Anatomy and Genetics, University of Oxford, Parks Road, Oxford, OX1 3PT, UK
| | - Holger Kramer
- Proteomics and Metabolomics Centre, Medical Research Council, London Institute of Medical Sciences, London, W12 0NN, UK
| | | |
Collapse
|
35
|
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: 16] [Impact Index Per Article: 4.0] [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
|
36
|
Sánchez-Collado J, López JJ, Rosado JA. The Orai1-AC8 Interplay: How Breast Cancer Cells Escape from Orai1 Channel Inactivation. Cells 2021; 10:1308. [PMID: 34070268 PMCID: PMC8225208 DOI: 10.3390/cells10061308] [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: 04/12/2021] [Revised: 05/17/2021] [Accepted: 05/20/2021] [Indexed: 11/16/2022] Open
Abstract
The interplay between the Ca2+-sensitive adenylyl cyclase 8 (AC8) and Orai1 channels plays an important role both in the activation of the cAMP/PKA signaling and the modulation of Orai1-dependent Ca2+ signals. AC8 interacts with a N-terminal region that is exclusive to the Orai1 long variant, Orai1α. The interaction between both proteins allows the Ca2+ that enters the cell through Orai1α to activate the generation of cAMP by AC8. Subsequent PKA activation results in Orai1α inactivation by phosphorylation at serine-34, thus shaping Orai1-mediated cellular functions. In breast cancer cells, AC8 plays a relevant role supporting a variety of cancer hallmarks, including proliferation and migration. Breast cancer cells overexpress AC8, which shifts the AC8-Orai1 stoichiometry in favor of the former and leads to the impairment of PKA-dependent Orai1α inactivation. This mechanism contributes to the enhanced SOCE observed in triple-negative breast cancer cells. This review summarizes the functional interaction between AC8 and Orai1α in normal and breast cancer cells and its relevance for different cancer features.
Collapse
Affiliation(s)
| | - José J. López
- Cellular Physiology Research Group, Department of Physiology, Institute of Molecular Pathology Biomarkers, University of Extremadura, 10003 Caceres, Spain;
| | - Juan A. Rosado
- Cellular Physiology Research Group, Department of Physiology, Institute of Molecular Pathology Biomarkers, University of Extremadura, 10003 Caceres, Spain;
| |
Collapse
|