1
|
Goriounova AS, Flori Sassano M, Wrennall JA, Tarran R. ELD607 specifically traffics Orai1 to the lysosome leading to inhibition of store operated calcium entry. Cell Calcium 2024; 123:102945. [PMID: 39191091 DOI: 10.1016/j.ceca.2024.102945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 08/11/2024] [Accepted: 08/12/2024] [Indexed: 08/29/2024]
Abstract
Orai1 is a plasma membrane Ca2+ channel involved in store operated calcium entry (SOCE). SOCE can regulate cell growth, exocytosis, gene expression and inflammation. We previously found that short palate lung and nasal epithelial clone 1's (SPLUNC1) sixth α-helix (α6) bound Orai1 to inhibit SOCE. SPLUNC1 was not proteolytically stable, so we developed ELD607, an 11 amino acid peptide based on SPLUNC1's α6 region which was more stable and more potent than SPLUNC1/α6. Here, we studied ELD607's mechanism of action. We overexpressed either Orai1-HA or Orai1-YFP in HEK293T cells to probe ELD607-Orai1 interactions by confocal microscopy. We also measured changes in Fluo-4 fluorescence in a multiplate reader as a marker of cytoplasmic Ca2+ levels. ELD607 internalized Orai1 independently of STIM1. Both 15 min and 3 h exposure to ELD607 similarly depleted Orai1 in the plasma membrane. However, 3 h exposure to ELD607 yielded greater inhibition of SOCE. ELD607 continued to colocalize with Orai1 after internalization and this process was dependent on the presence of the ubiquitin ligase NEDD4.2. Similarly, ELD607 increased the colocalization between Orai1 and ubiquitin. ELD607 also increased the colocalization between Orai1 and Rab5 and 7, but not Rab11, suggesting that Orai1 trafficked through early and late but not recycling endosomes. Finally, ELD607 caused Orai1, but not Orai2, Orai3, or STIM1 to traffic to lysosomes. We conclude that ELD607 rapidly binds to Orai1 and works in an identical fashion as full length SPLUNC1 by internalizing Orai1 and sending it to lysosomes, leading to a decrease in SOCE.
Collapse
Affiliation(s)
- Alexandra S Goriounova
- Department of Pharmacology, The University of North Carolina at Chapel Hill, NC 27599, USA
| | - M Flori Sassano
- Division of Genetic, Environmental and Inhalational Disease, Department of Internal Medicine, Kansas University Medical Center, Kansas City, KS 66103, USA
| | - Joe A Wrennall
- Department of Cell Biology & Physiology, The University of North Carolina at Chapel Hill, NC 27599, USA
| | - Robert Tarran
- Division of Genetic, Environmental and Inhalational Disease, Department of Internal Medicine, Kansas University Medical Center, Kansas City, KS 66103, USA.
| |
Collapse
|
2
|
Hu P, Xiao H, Alba MA, Atkins HM, Gou S, Hu Y, Gomez JC, Jania CM, Martin JR, Morrison TE, Tilley SL, Heise MT, Doerschuk CM, Falk RJ, Jennette JC. Myeloperoxidase-ANCA IgG induces different forms of small vessel vasculitis based on type of synergistic immune stimuli. Kidney Int 2024; 106:870-886. [PMID: 39216658 DOI: 10.1016/j.kint.2024.08.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 07/29/2024] [Accepted: 08/08/2024] [Indexed: 09/04/2024]
Abstract
Anti-neutrophil cytoplasmic autoantibody (ANCA) vasculitis has diverse patterns of injury including microscopic polyangiitis (MPA), granulomatosis with polyangiitis (GPA), and eosinophilic granulomatosis with polyangiitis (EGPA). Necrotizing and crescentic glomerulonephritis (NCGN) occurs in all syndromes and as renal limited vasculitis (RLV). Single-dose intravenous ANCA IgG-specific for mouse myeloperoxidase (MPO) causes RLV in mice. Although multiple mouse models have elucidated ANCA-IgG induced necrotizing and crescentic glomerulonephritis (NCGN), pathogenesis of ANCA-induced granulomatosis and vasculitis outside the kidney has not been clarified. To investigate this, we used intravenous MPO-ANCA IgG in the same strain of mice to induce different patterns of lung disease mirroring patients with granulomatosis with polyangiitis (GPA), microscopic polyangiitis (MPA), and eosinophilic granulomatosis with polyangiitis (EGPA). Repeated intravenous MPO-ANCA IgG induced GPA with NCGN, lung capillaritis, arteritis and granulomatosis. Lung leukocyte phenotypes were evaluated by immunohistochemical image analysis and by flow cytometry. ANCA lung capillaritis and microabscesses began within one day and evolved into granulomas in under seven days. Influenza plus single-dose MPO-ANCA IgG induced MPA with NCGN, lung capillaritis and arteritis, but no granulomatosis. Allergic airway disease caused by house dust mites or ovalbumin plus single-dose intravenous MPO-ANCA IgG induced EGPA with eosinophilic bronchiolitis, NCGN, capillaritis, arteritis, and granulomatosis. Thus, our study shows that the occurrence and pattern of lung lesions are determined by the same ANCA IgG accompanied by different synergistic immune factors.
Collapse
Affiliation(s)
- Peiqi Hu
- Department of Pathology and Laboratory Medicine, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA; University of North Carolina Kidney Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Hong Xiao
- Department of Pathology and Laboratory Medicine, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA; University of North Carolina Kidney Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Marco A Alba
- Department of Pathology and Laboratory Medicine, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Hannah M Atkins
- Department of Pathology and Laboratory Medicine, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Shenju Gou
- Department of Pathology and Laboratory Medicine, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Yanglin Hu
- Department of Pathology and Laboratory Medicine, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - John C Gomez
- Marsico Lung Institute, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Corey M Jania
- Marsico Lung Institute, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Jessica R Martin
- Marsico Lung Institute, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Thomas E Morrison
- Department of Microbiology and Immunology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Stephen L Tilley
- Marsico Lung Institute, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Mark T Heise
- Department of Microbiology and Immunology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Claire M Doerschuk
- Marsico Lung Institute, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA; Department of Medicine, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Ronald J Falk
- Department of Pathology and Laboratory Medicine, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA; University of North Carolina Kidney Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA; Department of Medicine, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - J Charles Jennette
- Department of Pathology and Laboratory Medicine, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA; University of North Carolina Kidney Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA; Department of Medicine, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA.
| |
Collapse
|
3
|
Zhang J, Dong F, Ju G, Pan X, Mao X, Zhang X. Sodium Houttuyfonate Alleviates Monocrotaline-induced Pulmonary Hypertension by Regulating Orai1 and Orai2. Am J Respir Cell Mol Biol 2024; 71:332-342. [PMID: 38709251 DOI: 10.1165/rcmb.2023-0015oc] [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: 01/09/2023] [Accepted: 05/06/2024] [Indexed: 05/07/2024] Open
Abstract
An increased intracellular Ca2+ concentration ([Ca2+]i) is a key trigger for pulmonary arterial smooth muscle cell (PASMC) proliferation and contributes greatly to pulmonary hypertension (PH). Extracellular Ca2+ influx via a store-operated Ca2+ channel, termed store-operated Ca2+ entry (SOCE), is a crucial mechanism for [Ca2+]i increase in PASMCs. Calcium release-activated calcium modulator (Orai) proteins, consisting of three members (Orai1-3), are the main components of the store-operated Ca2+ channel. Sodium houttuyfonate (SH) is a product of the addition reaction of sodium bisulfite and houttuynin and has antibacterial, antiinflammatory, and other properties. In this study, we assessed the contributions of Orai proteins to monocrotaline (MCT)-enhanced SOCE, [Ca2+]i, and cell proliferation in PASMCs and determined the effect of SH on MCT-PH and the underlying mechanism, focusing on Orai proteins, SOCE, and [Ca2+]i in PASMCs. Our results showed that: 1) Orai1 and Orai2 were selectively upregulated in the distal pulmonary arteries and the PASMCs of MCT-PH rats; 2) knockdown of Orai1 or Orai2 reduced SOCE, [Ca2+]i, and cell proliferation without affecting their expression in PASMCs in MCT-PH rats; 3) SH significantly normalized the characteristic parameters in a dose-dependent manner in the MCT-PH rat model; and 4) SH decreased MCT-enhanced SOCE, [Ca2+]i, and PASMC proliferation via Orai1 or Orai2. These results indicate that SH likely exerts its protective role in MCT-PH by inhibiting the Orai1,2-SOCE-[Ca2+]i signaling pathway.
Collapse
MESH Headings
- Animals
- Monocrotaline/toxicity
- Hypertension, Pulmonary/chemically induced
- Hypertension, Pulmonary/metabolism
- Hypertension, Pulmonary/pathology
- Hypertension, Pulmonary/drug therapy
- ORAI1 Protein/metabolism
- ORAI1 Protein/genetics
- Sulfites/pharmacology
- Rats
- Male
- Cell Proliferation/drug effects
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/pathology
- Pulmonary Artery/pathology
- Pulmonary Artery/drug effects
- Pulmonary Artery/metabolism
- ORAI2 Protein/metabolism
- Rats, Sprague-Dawley
- Calcium/metabolism
- Calcium Signaling/drug effects
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/pathology
- Alkanes
Collapse
Affiliation(s)
- Jun Zhang
- School of Medicine, Lishui University, Lishui, China
| | - Fang Dong
- School of Medicine, Lishui University, Lishui, China
| | - Gaojia Ju
- School of Pharmacy, Fujian Medical University, Fuzhou, China; and
| | - Xinli Pan
- School of Medicine, Lishui University, Lishui, China
| | - Xinwu Mao
- Department of Pathology, Lishui Municipal People Hospital, Lishui, China
| | - Xiaowen Zhang
- Department of Pathology, Lishui Municipal People Hospital, Lishui, China
| |
Collapse
|
4
|
Hanif SZ, Au CC, Torregroza I, Jannath SY, Fabiha T, Bhinder B, Washburn M, Devost D, Liu S, Bhardwaj P, Evans T, Anand PK, Tarran R, Palikhe S, Elemento O, Dow L, Blenis J, Hébert TE, Brown KA. The Orphan G Protein-Coupled Receptor GPR52 is a Novel Regulator of Breast Cancer Multicellular Organization. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.22.604482. [PMID: 39091857 PMCID: PMC11291042 DOI: 10.1101/2024.07.22.604482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/04/2024]
Abstract
G protein-coupled receptors (GPCRs) are the largest class of membrane-bound receptors and transmit critical signals from the extracellular to the intracellular spaces. Transcriptomic data of resected breast tumors shows that low mRNA expression of the orphan GPCR GPR52 correlates with reduced overall survival in breast cancer patients, leading to the hypothesis that loss of GPR52 supports breast cancer progression. CRISPR-Cas9 was used to knockout GPR52 in human triple-negative breast cancer (TNBC) cell lines MDA-MB-468 and MDA-MB-231, and in the non-cancerous breast epithelial cell line, MCF10A. Loss of GPR52 was found to be associated with increased cell-cell interaction in 2D cultures, altered 3D spheroid morphology, and increased propensity to organize and invade collectively in Matrigel. Furthermore, GPR52 loss was associated with features of EMT in MDA-MB-468 cells. To determine the in vivo impact of GPR52 loss, MDA-MB-468 cells were injected into zebrafish and loss of GPR52 was associated with a greater total cancer area compared to control cells. RNA-sequencing and proteomic analyses of GPR52-null breast cancer cells reveal an increased cAMP signaling signature. Consistently, we found that treatment of wild-type (WT) cells with forskolin, which stimulates production of cAMP, induces some phenotypic changes associated with GPR52 loss, and inhibition of cAMP production rescued some of the GPR52 KO phenotypes. Overall, our results reveal GPR52 loss as a potential mechanism by which breast cancer progression may occur and support the investigation of GPR52 agonism as a therapeutic option in breast cancer.
Collapse
Affiliation(s)
- Sarah Z Hanif
- Weill Cornell Medicine/Rockefeller University/Memorial Sloan-Kettering Cancer Center Tri-Institutional MD-PhD Program, New York, NY, USA
- Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | | | | | - Syeda Y Jannath
- Department of Medicine, Weill Cornell Medicine, New York, NY, USA
- Hunter College, City University of New York, New York, NY, USA
| | - Tabassum Fabiha
- Columbia University Computational & Systems Biology Program, Sloan-Kettering Institute of Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Bhavneet Bhinder
- Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, New York, USA
| | - Michael Washburn
- Department of Cancer Biology, University of Kansas Medical Center Kansas City KS USA
- University of Kansas Cancer Center, Kansas City, KS, USA
| | - Dominic Devost
- Department of Pharmacology and Therapeutics, McGill University, Montréal, QC, Canada
| | - Shuchen Liu
- Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Priya Bhardwaj
- Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Todd Evans
- Department of Surgery, Weill Cornell Medicine, New York, NY, USA
| | - Pradeep Kumar Anand
- Division of Genetic, Environmental and Inhalational Disease, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | - Robert Tarran
- Division of Genetic, Environmental and Inhalational Disease, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | - Sailesh Palikhe
- Department of Cell Biology and Physiology, University of Kansas Medical Center Kansas City, KS, USA
| | - Olivier Elemento
- Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, New York, USA
| | - Lukas Dow
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
| | - John Blenis
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
- Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA
- Department of Biochemistry, Weill Cornell Medicine, New York, NY, USA
| | - Terence E Hébert
- Department of Pharmacology and Therapeutics, McGill University, Montréal, QC, Canada
| | - Kristy A Brown
- Department of Medicine, Weill Cornell Medicine, New York, NY, USA
- University of Kansas Cancer Center, Kansas City, KS, USA
- Department of Cell Biology and Physiology, University of Kansas Medical Center Kansas City, KS, USA
| |
Collapse
|
5
|
Saint-Martin Willer A, Montani D, Capuano V, Antigny F. Orai1/STIMs modulators in pulmonary vascular diseases. Cell Calcium 2024; 121:102892. [PMID: 38735127 DOI: 10.1016/j.ceca.2024.102892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 03/27/2024] [Accepted: 04/23/2024] [Indexed: 05/14/2024]
Abstract
Calcium (Ca2+) is a secondary messenger that regulates various cellular processes. However, Ca2+ mishandling could lead to pathological conditions. Orai1 is a Ca2+channel contributing to the store-operated calcium entry (SOCE) and plays a critical role in Ca2+ homeostasis in several cell types. Dysregulation of Orai1 contributed to severe combined immune deficiency syndrome, some cancers, pulmonary arterial hypertension (PAH), and other cardiorespiratory diseases. During its activation process, Orai1 is mainly regulated by stromal interacting molecule (STIM) proteins, especially STIM1; however, many other regulatory partners have also been recently described. Increasing knowledge about these regulatory partners provides a better view of the downstream signalling pathways of SOCE and offers an excellent opportunity to decipher Orai1 dysregulation in these diseases. These proteins participate in other cellular functions, making them attractive therapeutic targets. This review mainly focuses on Orai1 regulatory partners in the physiological and pathological conditions of the pulmonary circulation and inflammation.
Collapse
Affiliation(s)
- Anaïs Saint-Martin Willer
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicêtre, France; INSERM UMR_S 999 Hypertension pulmonaire: Physiopathologie et Innovation Thérapeutique, Hôpital Marie Lannelongue, Le Plessis-Robinson, France
| | - David Montani
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicêtre, France; INSERM UMR_S 999 Hypertension pulmonaire: Physiopathologie et Innovation Thérapeutique, Hôpital Marie Lannelongue, Le Plessis-Robinson, France; 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
| | - Véronique Capuano
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicêtre, France; INSERM UMR_S 999 Hypertension pulmonaire: Physiopathologie et Innovation Thérapeutique, Hôpital Marie Lannelongue, Le Plessis-Robinson, France; Hôptal Marie Lannelongue, Groupe Hospitalier Paris Saint-Joseph, Le Plessis-Robinson, France
| | - Fabrice Antigny
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicêtre, France; INSERM UMR_S 999 Hypertension pulmonaire: Physiopathologie et Innovation Thérapeutique, Hôpital Marie Lannelongue, Le Plessis-Robinson, France.
| |
Collapse
|
6
|
Ahmad S, Wrennall JA, Goriounova AS, Sekhri M, Iskarpatyoti JA, Ghosh A, Abdelwahab SH, Voeller A, Rai M, Mahida RY, Krajewski K, Ignar DM, Greenbaum A, Moran TP, Tilley SL, Thickett DR, Sassano MF, Tarran R. Specific Inhibition of Orai1-mediated Calcium Signalling Resolves Inflammation and Clears Bacteria in an Acute Respiratory Distress Syndrome Model. Am J Respir Crit Care Med 2024; 209:703-715. [PMID: 37972349 PMCID: PMC10945054 DOI: 10.1164/rccm.202308-1393oc] [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: 08/10/2023] [Accepted: 11/16/2023] [Indexed: 11/19/2023] Open
Abstract
Rationale: Acute respiratory distress syndrome (ARDS) has an unacceptably high mortality rate (35%) and is without effective therapy. Orai1 is a Ca2+ channel involved in store-operated Ca2+ entry (SOCE), a process that exquisitely regulates inflammation. Orai1 is considered a druggable target, but no Orai1-specific inhibitors exist to date. Objectives: To evaluate whether ELD607, a first-in-class Orai1 antagonist, can treat ARDS caused by bacterial pneumonia in preclinical models. Methods: ELD607 pharmacology was evaluated in HEK293T cells and freshly isolated immune cells from patients with ARDS. A murine acute lung injury model caused by bacterial pneumonia was then used: mice were infected with Pseudomonas aeruginosa, Staphylococcus aureus, methicillin-resistant S. aureus, or multidrug-resistant P. aeruginosa and then treated with ELD607 intranasally. Measurements and Main Results: ELD607 specifically inhibited SOCE in HEK293T cells with a half-maximal inhibitory concentration of 9 nM. ELD607 was stable in ARDS airway secretions and inhibited SOCE in ARDS immune cells. In vivo, inhaled ELD607 significantly reduced neutrophilia and improved survival. Surprisingly, Orai1 inhibition by ELD607 caused a significant reduction in lung bacteria, including methicillin-resistant S. aureus. ELD607 worked as an immunomodulator that reduced cytokine levels, reduced neutrophilia, and promoted macrophage-mediated resolution of inflammation and clearance of bacteria. Indeed, when alveolar macrophages were depleted with inhaled clodronate, ELD607 was no longer able to resolve inflammation or clear bacteria. Conclusions: These data indicate that specific Orai1 inhibition by ELD607 may be a novel approach to reduce multiorgan inflammation and treat antibiotic-resistant bacteria.
Collapse
Affiliation(s)
- Saira Ahmad
- Department of Cell Biology and Physiology
- Eldec Pharmaceuticals, Chapel Hill, North Carolina
| | | | | | | | | | | | | | | | - Mani Rai
- Department of Biomedical Engineering, North Carolina State University, Raleigh, North Carolina; and
| | - Rahul Y. Mahida
- Birmingham Acute Care Research Group, University of Birmingham, Birmingham, United Kingdom
| | | | | | - Alon Greenbaum
- Department of Biomedical Engineering, North Carolina State University, Raleigh, North Carolina; and
| | - Timothy P. Moran
- Department of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Stephen L. Tilley
- Department of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - David R. Thickett
- Birmingham Acute Care Research Group, University of Birmingham, Birmingham, United Kingdom
| | - M. Flori Sassano
- Department of Cell Biology and Physiology
- Eldec Pharmaceuticals, Chapel Hill, North Carolina
| | - Robert Tarran
- Department of Cell Biology and Physiology
- Eldec Pharmaceuticals, Chapel Hill, North Carolina
| |
Collapse
|
7
|
Hough RF, Mthunzi L. Macrophage SPLUNC-ing Clears Bacteria in the Lung. Am J Respir Crit Care Med 2024; 209:623-625. [PMID: 38064242 PMCID: PMC10945050 DOI: 10.1164/rccm.202311-2075ed] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2024] Open
Affiliation(s)
- Rebecca F Hough
- Department of Pediatrics Columbia University New York, New York
| | - Liberty Mthunzi
- Department of Medicine Columbia University New York, New York
| |
Collapse
|
8
|
Ben-Meir E, Perrem L, Shaw M, Ratjen F, Grasemann H. SPLUNC1 as a biomarker of pulmonary exacerbations in children with cystic fibrosis. J Cyst Fibros 2024; 23:288-292. [PMID: 38413298 DOI: 10.1016/j.jcf.2024.02.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 02/14/2024] [Accepted: 02/19/2024] [Indexed: 02/29/2024]
Abstract
BACKGROUND Short palate, lung, and nasal epithelium clone 1 (SPLUNC1) is an innate defence protein that acts as an anti-microbial agent and regulates airway surface liquid volume through inhibition of the epithelial sodium channel (ENaC). SPLUNC1 levels were found to be reduced in airway secretions of adults with cystic fibrosis (CF). The potential of SPLUNC1 as a biomarker in children with CF is unknown. METHODS We quantified SPLUNC1, interleukin-8 (IL-8) and neutrophil elastase (NE) in sputum of CF children treated with either intravenous antibiotics or oral antibiotics for a pulmonary exacerbation (PEx)s, and in participants of a prospective cohort of CF children with preserved lung function on spirometry, followed over a period of two years. RESULTS Sputum SPLUNC1 levels were significantly lower before compared to after intravenous and oral antibiotic therapy for PEx. In the longitudinal cohort, SPLUNC1 levels were found to be decreased at PEx visits compared to both previous and subsequent stable visits. Higher SPLUNC1 levels at stable visits were associated with longer PEx-free time (hazard ratio 0.85, p = 0.04). SPLUNC1 at PEx visits did not correlate with IL-8 or NE levels in sputum or forced expiratory volume in one second (FEV1) but did correlate with the lung clearance index (LCI) (r=-0.53, p < 0.001). CONCLUSION SPLUNC1 demonstrates promising clinometric properties as a biomarker of PEx in children with CF.
Collapse
Affiliation(s)
- E Ben-Meir
- Division of Respiratory Medicine, Department of Paediatrics, The Hospital for Sick Children, Toronto, Canada; Translational Medicine, Research Institute, The Hospital for Sick Children, Toronto, Ontario, Canada; University of Toronto, Toronto, Ontario, Canada
| | - L Perrem
- Division of Respiratory Medicine, Department of Paediatrics, The Hospital for Sick Children, Toronto, Canada; Translational Medicine, Research Institute, The Hospital for Sick Children, Toronto, Ontario, Canada; University of Toronto, Toronto, Ontario, Canada
| | - M Shaw
- Division of Respiratory Medicine, Department of Paediatrics, The Hospital for Sick Children, Toronto, Canada; Translational Medicine, Research Institute, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - F Ratjen
- Division of Respiratory Medicine, Department of Paediatrics, The Hospital for Sick Children, Toronto, Canada; Translational Medicine, Research Institute, The Hospital for Sick Children, Toronto, Ontario, Canada; University of Toronto, Toronto, Ontario, Canada
| | - H Grasemann
- Division of Respiratory Medicine, Department of Paediatrics, The Hospital for Sick Children, Toronto, Canada; Translational Medicine, Research Institute, The Hospital for Sick Children, Toronto, Ontario, Canada; University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
9
|
Parekh AB. House dust mite allergens, store-operated Ca 2+ channels and asthma. J Physiol 2023. [PMID: 38054814 DOI: 10.1113/jp284931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 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
|
10
|
Iannuzo N, Welfley H, Li NC, Johnson MDL, Rojas-Quintero J, Polverino F, Guerra S, Li X, Cusanovich DA, Langlais PR, Ledford JG. CC16 drives VLA-2-dependent SPLUNC1 expression. Front Immunol 2023; 14:1277582. [PMID: 38053993 PMCID: PMC10694244 DOI: 10.3389/fimmu.2023.1277582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 10/30/2023] [Indexed: 12/07/2023] Open
Abstract
Rationale CC16 (Club Cell Secretory Protein) is a protein produced by club cells and other non-ciliated epithelial cells within the lungs. CC16 has been shown to protect against the development of obstructive lung diseases and attenuate pulmonary pathogen burden. Despite recent advances in understanding CC16 effects in circulation, the biological mechanisms of CC16 in pulmonary epithelial responses have not been elucidated. Objectives We sought to determine if CC16 deficiency impairs epithelial-driven host responses and identify novel receptors expressed within the pulmonary epithelium through which CC16 imparts activity. Methods We utilized mass spectrometry and quantitative proteomics to investigate how CC16 deficiency impacts apically secreted pulmonary epithelial proteins. Mouse tracheal epithelial cells (MTECS), human nasal epithelial cells (HNECs) and mice were studied in naïve conditions and after Mp challenge. Measurements and main results We identified 8 antimicrobial proteins significantly decreased by CC16-/- MTECS, 6 of which were validated by mRNA expression in Severe Asthma Research Program (SARP) cohorts. Short Palate Lung and Nasal Epithelial Clone 1 (SPLUNC1) was the most differentially expressed protein (66-fold) and was the focus of this study. Using a combination of MTECs and HNECs, we found that CC16 enhances pulmonary epithelial-driven SPLUNC1 expression via signaling through the receptor complex Very Late Antigen-2 (VLA-2) and that rCC16 given to mice enhances pulmonary SPLUNC1 production and decreases Mycoplasma pneumoniae (Mp) burden. Likewise, rSPLUNC1 results in decreased Mp burden in mice lacking CC16 mice. The VLA-2 integrin binding site within rCC16 is necessary for induction of SPLUNC1 and the reduction in Mp burden. Conclusion Our findings demonstrate a novel role for CC16 in epithelial-driven host defense by up-regulating antimicrobials and define a novel epithelial receptor for CC16, VLA-2, through which signaling is necessary for enhanced SPLUNC1 production.
Collapse
Affiliation(s)
- Natalie Iannuzo
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, United States
| | - Holly Welfley
- Asthma and Airway Disease Research Center, Tucson, AZ, United States
| | | | | | | | | | - Stefano Guerra
- Asthma and Airway Disease Research Center, Tucson, AZ, United States
- Department of Medicine, Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, University of Arizona, Tucson, AZ, United States
| | - Xingnan Li
- Department of Medicine, Division of Genetics, Genomics, and Precision Medicine, University of Arizona, Tucson, AZ, United States
| | - Darren A. Cusanovich
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, United States
- Asthma and Airway Disease Research Center, Tucson, AZ, United States
| | - Paul R. Langlais
- Department of Medicine, Division of Endocrinology, University of Arizona, Tucson, AZ, United States
| | - Julie G. Ledford
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, United States
- Asthma and Airway Disease Research Center, Tucson, AZ, United States
| |
Collapse
|
11
|
Hodeify R, Machaca K. Methods to Quantify the Dynamic Recycling of Plasma Membrane Channels. Bio Protoc 2023; 13:e4800. [PMID: 37719078 PMCID: PMC10501913 DOI: 10.21769/bioprotoc.4800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 06/19/2023] [Accepted: 06/19/2023] [Indexed: 09/19/2023] Open
Abstract
Store-operated Ca2+ entry (SOCE) is a ubiquitous Ca2+ signaling modality mediated by Orai Ca2+ channels at the plasma membrane (PM) and the endoplasmic reticulum (ER) Ca2+ sensors STIM1/2. At steady state, Orai1 constitutively cycles between an intracellular compartment and the PM. Orai1 PM residency is modulated by its endocytosis and exocytosis rates. Therefore, Orai1 trafficking represents an important regulatory mechanism to define the levels of Ca2+ influx. Here, we present a protocol using the dually tagged YFP-HA-Orai1 with a cytosolic YFP and extracellular hemagglutinin (HA) tag to quantify Orai1 cycling rates. For measuring Orai1 endocytosis, cells expressing YFP-HA-Orai1 are incubated with mouse anti-HA antibody for various periods of time before being fixed and stained for surface Orai1 with Cy5-labeled anti-mouse IgG. The cells are fixed again, permeabilized, and stained with Cy3-labeled anti-mouse IgG to reveal anti-HA that has been internalized. To quantify Orai1 exocytosis rate, cells are incubated with anti-HA antibody for various incubation periods before being fixed, permeabilized, and then stained with Cy5-labeled anti-mouse IgG. The Cy5/YFP ratio is plotted over time and fitted with a mono-exponential growth curve to determine exocytosis rate. Although the described assays were developed to measure Orai1 trafficking, they are readily adaptable to other PM channels. Key features Detailed protocols to quantify endocytosis and exocytosis rates of Orai1 at the plasma membrane that can be used in various cell lines. The endocytosis and exocytosis assays are readily adaptable to study the trafficking of other plasma membrane channels.
Collapse
Affiliation(s)
- Rawad Hodeify
- Biotechnology Department, School of Arts and
Sciences, American University of Ras Al Khaimah, Ras Al Khaimah, United Arab
Emirates
| | - Khaled Machaca
- Ca
- Department of Physiology and Biophysics, Weill
Cornell Medicine, New York, USA
| |
Collapse
|
12
|
Yao Y, Borkar NA, Zheng M, Wang S, Pabelick CM, Vogel ER, Prakash YS. Interactions between calcium regulatory pathways and mechanosensitive channels in airways. Expert Rev Respir Med 2023; 17:903-917. [PMID: 37905552 PMCID: PMC10872943 DOI: 10.1080/17476348.2023.2276732] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 10/25/2023] [Indexed: 11/02/2023]
Abstract
INTRODUCTION Asthma is a chronic lung disease influenced by environmental and inflammatory triggers and involving complex signaling pathways across resident airway cells such as epithelium, airway smooth muscle, fibroblasts, and immune cells. While our understanding of asthma pathophysiology is continually progressing, there is a growing realization that cellular microdomains play critical roles in mediating signaling relevant to asthma in the context of contractility and remodeling. Mechanosensitive pathways are increasingly recognized as important to microdomain signaling, with Piezo and transient receptor protein (TRP) channels at the plasma membrane considered important for converting mechanical stimuli into cellular behavior. Given their ion channel properties, particularly Ca2+ conduction, a question becomes whether and how mechanosensitive channels contribute to Ca2+ microdomains in airway cells relevant to asthma. AREAS COVERED Mechanosensitive TRP and Piezo channels regulate key Ca2+ regulatory proteins such as store operated calcium entry (SOCE) involving STIM and Orai channels, and sarcoendoplasmic (SR) mechanisms such as IP3 receptor channels (IP3Rs), and SR Ca2+ ATPase (SERCA) that are important in asthma pathophysiology including airway hyperreactivity and remodeling. EXPERT OPINION Physical and/or functional interactions between Ca2+ regulatory proteins and mechanosensitive channels such as TRP and Piezo can toward understanding asthma pathophysiology and identifying novel therapeutic approaches.
Collapse
Affiliation(s)
- Yang Yao
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi’an Medical University, Xi’an, Shaanxi, China
- Department of Anesthesiology, Mayo Clinic, Rochester, MN, USA
| | - Niyati A Borkar
- Department of Anesthesiology, Mayo Clinic, Rochester, MN, USA
| | - Mengning Zheng
- Department of Anesthesiology, Mayo Clinic, Rochester, MN, USA
- Department of Respiratory and Critical Care Medicine, Guizhou Province People’s Hospital, Guiyang, Guizhou, China
| | - Shengyu Wang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi’an Medical University, Xi’an, Shaanxi, China
| | - Christina M Pabelick
- Department of Anesthesiology, Mayo Clinic, Rochester, MN, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Elizabeth R Vogel
- Department of Anesthesiology, Mayo Clinic, Rochester, MN, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - YS Prakash
- Department of Anesthesiology, Mayo Clinic, Rochester, MN, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
13
|
Protasi F, Girolami B, Roccabianca S, Rossi D. Store-operated calcium entry: From physiology to tubular aggregate myopathy. Curr Opin Pharmacol 2023; 68:102347. [PMID: 36608411 DOI: 10.1016/j.coph.2022.102347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 11/30/2022] [Accepted: 12/04/2022] [Indexed: 01/06/2023]
Abstract
Store-Operated Ca2+ entry (SOCE) is recognized as a key mechanism in muscle physiology necessary to refill intracellular Ca2+ stores during sustained muscle activity. For many years the cell structures expected to mediate SOCE in skeletal muscle fibres remained unknown. Recently, the identification of Ca2+ Entry Units (CEUs) in exercised muscle fibres opened new insights into the role of extracellular Ca2+ in muscle contraction and, more generally, in intracellular Ca2+ homeostasis. Accordingly, intracellular Ca2+ unbalance due to alterations in SOCE strictly correlates with muscle disfunction and disease. Mutations in proteins involved in SOCE (STIM1, ORAI1, and CASQ1) have been linked to tubular aggregate myopathy (TAM), a disease that causes muscle weakness and myalgia and is characterized by a typical accumulation of highly ordered and packed membrane tubules originated from the sarcoplasmic reticulum (SR). Achieving a full understanding of the molecular pathways activated by alterations in Ca2+ entry mechanisms is a necessary step to design effective therapies for human SOCE-related disorders.
Collapse
Affiliation(s)
- Feliciano Protasi
- CAST, Center for Advanced Studies and Technology; University G. d'Annunzio of Chieti-Pescara, I-66100, Italy; DMSI, Department of Medicine and Aging Sciences; University G. d'Annunzio of Chieti-Pescara, I-66100, Italy
| | - Barbara Girolami
- CAST, Center for Advanced Studies and Technology; University G. d'Annunzio of Chieti-Pescara, I-66100, Italy; DMSI, Department of Medicine and Aging Sciences; University G. d'Annunzio of Chieti-Pescara, I-66100, Italy
| | - Sara Roccabianca
- DMMS, Department of Molecular and Developmental Medicine; University of Siena, I-53100, Siena Italy
| | - Daniela Rossi
- DMMS, Department of Molecular and Developmental Medicine; University of Siena, I-53100, Siena Italy.
| |
Collapse
|
14
|
Clifton C, Niemeyer BF, Novak R, Can UI, Hainline K, Benam KH. BPIFA1 is a secreted biomarker of differentiating human airway epithelium. Front Cell Infect Microbiol 2022; 12:1035566. [PMID: 36519134 PMCID: PMC9744250 DOI: 10.3389/fcimb.2022.1035566] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 11/07/2022] [Indexed: 11/29/2022] Open
Abstract
In vitro culture and differentiation of human-derived airway basal cells under air-liquid interface (ALI) into a pseudostratified mucociliated mucosal barrier has proven to be a powerful preclinical tool to study pathophysiology of respiratory epithelium. As such, identifying differentiation stage-specific biomarkers can help investigators better characterize, standardize, and validate populations of regenerating epithelial cells prior to experimentation. Here, we applied longitudinal transcriptomic analysis and observed that the pattern and the magnitude of OMG, KRT14, STC1, BPIFA1, PLA2G7, TXNIP, S100A7 expression create a unique biosignature that robustly indicates the stage of epithelial cell differentiation. We then validated our findings by quantitative hemi-nested real-time PCR from in vitro cultures sourced from multiple donors. In addition, we demonstrated that at protein-level secretion of BPIFA1 accurately reflects the gene expression profile, with very low quantities present at the time of ALI induction but escalating levels were detectable as the epithelial cells terminally differentiated. Moreover, we observed that increase in BPIFA1 secretion closely correlates with emergence of secretory cells and an anti-inflammatory phenotype as airway epithelial cells undergo mucociliary differentiation under air-liquid interface in vitro.
Collapse
Affiliation(s)
- Clarissa Clifton
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Brian F. Niemeyer
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Richard Novak
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, United States
| | - Uryan Isik Can
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Kelly Hainline
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, United States
| | - Kambez H. Benam
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, United States,Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States,Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, United States,*Correspondence: Kambez H. Benam,
| |
Collapse
|
15
|
SPLUNC1 regulates LPS-induced progression of nasopharyngeal carcinoma and proliferation of myeloid-derived suppressor cells. MEDICAL ONCOLOGY (NORTHWOOD, LONDON, ENGLAND) 2022; 39:214. [PMID: 36175598 DOI: 10.1007/s12032-022-01816-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 07/29/2022] [Indexed: 10/14/2022]
Abstract
Nasopharyngeal carcinoma (NPC) is one of the aggressive malignant tumors with high mortality, and the proliferation of myeloid-derived suppressor cells (MDSCs) could promote the metastasis of NPC through inhibiting the function of T cells. Meanwhile, SPLUNC1 was known to inhibit the malignant behavior of NPC cells, while the detailed function of SPLUNC1 in LPS-modified immune microenvironment of NPC remains unclear. To assess the impact of SPLUNC1 in immune microenvironment during the progression of NPC, NPC cells were exposed to LPS and then co-cultured with MDSCs for 48 h. RT-qPCR and western blot were performed to evaluate the mRNA and protein level of SPLUNC1, CXCL-2 and CXCR-2, respectively. The level of IL-1β, IL-6, TNF-α, PD-L1, Arg-1 and iNOS were tested by ELISA. Meanwhile, the expression of CD33+ was tested by flow cytometry. The expression of CXCL-2 and CXCR-2 in NPC cells was higher, compared to that in NP69 cells. In contrast, SPLUNC1 level in NPC cells was much lower than that in NP69 cells. SPLUNC1 level was negatively correlated with CXCL-2 and CXCR-2. Overexpression of SPLUNC1 reversed LPS-induced inflammatory responses and proliferation in NPC cells. In addition, SPLUNC1 upregulation could reverse LPS-induced proliferation of MDSCs in tumor microenvironment. Meanwhile, SPLUNC1 overexpression could regulate CXCL-2/CXCR-2 axis through decreasing CXCL-2 and CXCR-2 protein and mRNA expression. SPLUNC1 regulates LPS-induced progression of nasopharyngeal carcinoma and proliferation of MDSCs. Thus, our study might provide a theoretical basis for discovering new strategies against NPC.
Collapse
|
16
|
Wu T, Goriounova AS, Worthington EN, Wrennall JA, Ghosh A, Ahmad S, Flori Sassano M, Tarran R. SPLUNC1 is a negative regulator of the Orai1 Ca 2+ channel. Physiol Rep 2022; 10:e15306. [PMID: 35581745 PMCID: PMC9114653 DOI: 10.14814/phy2.15306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 04/22/2022] [Accepted: 04/27/2022] [Indexed: 06/15/2023] Open
Abstract
Orai1 is a ubiquitously-expressed plasma membrane Ca2+ channel that is involved in store-operated Ca2+ entry (SOCE): a fundamental biological process that regulates gene expression, the onset of inflammation, secretion, and the contraction of airway smooth muscle (ASM). During SOCE, Ca2+ leaves the endoplasmic reticulum, which then stimulates a second, amplifying wave of Ca2+ influx through Orai1 into the cytoplasm. Short Palate LUng and Nasal epithelial Clone 1 (SPLUNC1; gene name BPIFA1) is a multi-functional, innate defense protein that is highly abundant in the lung. We have previously reported that SPLUNC1 was secreted from epithelia, where it bound to and inhibited Orai1, leading to reduced SOCE and ASM relaxation. However, the underlying mechanism of action is unknown. Here, we probed the SPLUNC1-Orai1 interactions in ASM and HEK293T cells using biochemical and imaging techniques. We observed that SPLUNC1 caused a conformational change in Orai1, as measured using Forster resonance energy transfer (FRET). SPLUNC1 binding also led to Nedd4-2 dependent ubiquitination of Orai1. Moreover, SPLUNC1 internalized Orai1 to lysosomes, leading to Orai1 degradation. Thus, we conclude that SPLUNC1 is an allosteric regulator of Orai1. Our data indicate that SPLUNC1-mediated Orai1 inhibition could be utilized as a therapeutic strategy to reduce SOCE.
Collapse
Affiliation(s)
- Tongde Wu
- Department of Cell Biology & PhysiologyThe University of North Carolina at Chapel HillNorth Carolina27599USA
| | - Alexandra S. Goriounova
- Department of PharmacologyThe University of North Carolina at Chapel HillNorth Carolina27599USA
| | - Erin N. Worthington
- Divison of PulmonologyDepartment of PediatricsThe University of North Carolina at Chapel HillNorth Carolina27599USA
- Division of Pulmonology, Department of PediatricsCarilion Clinic and Virginia Tech Carilion School of MedicineRoanokeVirginia24016USA
| | - Joe A. Wrennall
- Department of Cell Biology & PhysiologyThe University of North Carolina at Chapel HillNorth Carolina27599USA
| | - Arunava Ghosh
- Department of Cell Biology & PhysiologyThe University of North Carolina at Chapel HillNorth Carolina27599USA
| | - Saira Ahmad
- Department of Cell Biology & PhysiologyThe University of North Carolina at Chapel HillNorth Carolina27599USA
| | - M. Flori Sassano
- Department of Cell Biology & PhysiologyThe University of North Carolina at Chapel HillNorth Carolina27599USA
| | - Robert Tarran
- Department of Cell Biology & PhysiologyThe University of North Carolina at Chapel HillNorth Carolina27599USA
| |
Collapse
|
17
|
Affiliation(s)
- Ross Vlahos
- RMIT University, School of Health and Biomedical Sciences, Bundoora, Victoria, Australia;
| |
Collapse
|