1 Na+ Influx Via Orai1 Inhibits Intracellular ATP Induced Mtorc2 Signaling To

Total Page:16

File Type:pdf, Size:1020Kb

1 Na+ Influx Via Orai1 Inhibits Intracellular ATP Induced Mtorc2 Signaling To 1 Na+ Influx via Orai1 Inhibits Intracellular ATP Induced mTORC2 Signaling to 2 Disrupt CD4 T Cell Gene Expression and Differentiation 3 4 Yong Miao, Jaya Bhushan, Adish Dani and Monika Vig* 5 Department of Pathology and Immunology 6 Washington University School of Medicine, St. Louis MO 63110, USA 7 *Correspondence to: [email protected] 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 1 23 Abstract: T cell effector functions require sustained calcium influx. However, the 24 signaling and phenotypic consequences of non-specific sodium permeation via calcium 25 channels remain unknown. α-SNAP is a crucial component of Orai1 channels, and its 26 depletion disrupts the functional assembly of Orai1 multimers. Here we show that α- 27 SNAP hypomorph, hydrocephalos with hopping gait, Napahyh/hyh mice harbor significant 28 defects in CD4 T cell gene expression and Foxp3 regulatory T cell (Treg) differentiation. 29 Mechanistically, TCR stimulation induced rapid sodium influx in Napahyh/hyh CD4 T cells, 30 which reduced intracellular ATP, [ATP]i. Depletion of [ATP]i inhibited mTORC2 31 dependent NFκB activation in Napahyh/hyh cells but ablation of Orai1 restored it. 32 Remarkably, TCR stimulation in the presence of monensin phenocopied the defects in 33 Napahyh/hyh signaling and Treg differentiation, but not IL-2 expression. Thus, non-specific 34 sodium influx via bonafide calcium channels disrupts unexpected signaling nodes and 35 may provide mechanistic insights into some divergent phenotypes associated with Orai1 36 function. 37 38 39 40 41 42 43 44 45 2 46 Introduction: 47 A sustained rise in cytosolic calcium levels is necessary for nuclear translocation 48 of calcium dependent transcription factors such as nuclear factor of activated T cell 49 (NFAT) (Crabtree, 1999, Parekh and Putney, 2005, Winslow et al., 2003, Macian, 2005, 50 Vig and Kinet, 2009, Vig and Kinet, 2007, Crabtree, 2001). NFAT proteins are essential 51 for the development of several tissues but have been found to be dispensable for thymic 52 development and function of Foxp3 regulatory T cells (Tregs) (Crabtree and Olson, 53 2002, Crabtree, 2001, Crabtree, 1999, Timmerman et al., 1997, Vaeth et al., 2012). The 54 role of proteins directly involved in sustained calcium influx, however, remains less well 55 established. Specifically, genetic ablation of ORAI1, the pore forming subunit of calcium 56 release activated calcium (CRAC) channels (Vig et al., 2006b, Vig et al., 2006a, Peinelt 57 et al., 2006), partially inhibits T cell effector cytokines in mice and does not affect Foxp3 58 Treg development (Vig et al., 2008, Vig and Kinet, 2009, Gwack et al., 2008, McCarl et 59 al., 2010). The role of ORAI2 as well as ORAI3, the two closely related homologs of 60 ORAI1 that are highly expressed in mouse T cells remains unestablished in mice and 61 humans although all ORAIs are capable of reconstituting CRAC currents in vitro (Mercer 62 et al., 2006, Lis et al., 2007, DeHaven et al., 2007). 63 STIM1 and STIM2, the ER resident calcium sensor proteins, are required for ER 64 calcium release and Orai1 activation and T lymphocyte effector functions (Oh-Hora et 65 al., 2008). However, ablation of STIMs, but not ORAIs, affects thymic development of 66 Tregs (Oh-Hora et al., 2013, McCarl et al., 2010) and ablation of STIMs, but not ORAI1, 67 resulting in multi-organ autoimmunity in mice and humans (Oh-Hora et al., 2008, McCarl 68 et al., 2010, Picard et al., 2009). Because STIMs perform several additional functions 3 69 such as regulation of calcium selectivity of ORAI1 channels (McNally et al., 2012) as 70 well as inhibition of voltage gated calcium channel Cav1.2 (Wang et al., 2010, Park et 71 al., 2010), role of sustained calcium influx or store-operated calcium entry (SOCE) in the 72 development of Tregs and autoimmunity remains correlative (Oh-Hora et al., 2008, Oh- 73 Hora et al., 2013). Likewise, the phenotypes of human patients harboring different Stim 74 and Orai mutations range from immunodeficiency to autoimmunity and cancer. Despite 75 this diversity, all phenotypes are currently correlated with reduced SOCE (Picard et al., 76 2009). 77 We have previously shown that α-soluble NSF-attachment protein (α-SNAP), a 78 cytosolic protein traditionally studied in the context of soluble NSF attachment protein 79 receptor (SNARE) complex disassembly and membrane trafficking (Clary et al., 1990), 80 directly binds Stim1 and Orai1 and is necessary for the functional assembly and ion 81 specificity of multimeric Orai1 channels (Miao et al., 2013, Li et al., 2016). In addition, α- 82 SNAP has been implicated in AMP kinase (AMPK) inhibition and zippering of SNAREs 83 in vitro (Park et al., 2014, Baur et al., 2007, Wang and Brautigan, 2013). SNAREs play a 84 direct role in exocytosis and are therefore required for cytotoxic T, natural killer and 85 mast cell degranulation (Baram et al., 2001, Puri et al., 2003, Hepp et al., 2005, Suzuki 86 and Verma, 2008). However, the role of α-SNAP is less clear in vivo, and remains 87 unexplored in the immune system. α-SNAP deletion is embryonic lethal in mice and a 88 hypomorphic missense mutation in α-SNAP, hydrocephalous with hopping gait, 89 (Napahyh/hyh) has been previously reported to cause neuro-developmental defects 90 (Bronson and Lane, 1990, Chae et al., 2004, Hong et al., 2004). 91 Here we show that reduced expression of α-SNAP causes unexpected defects in 4 92 CD4 T cell signaling, gene expression and Foxp3 Treg differentiation. Using RNAi 93 mediated ablation of Orai1 in Napahyh/hyh CD4 T cells and monensin treatment of 94 wildtype CD4 T cells, we demonstrate that Orai1 mediated sodium influx, but not hyh/hyh 95 reduced SOCE, depletes [ATP]i in T cell receptor (TCR) stimulated Napa CD4 T 96 cells. Furthermore, we find that depletion of [ATP]i levels disrupts mTORC2 activation 97 which, in turn, inhibits NFκB activation and in vivo as well as in vitro differentiation of 98 Foxp3 Tregs in Napahyh/hyh mice. Therefore, analysis of α-SNAP deficient mice reveals 99 that non-specific sodium permeation via Orai1 disrupts a novel signaling node and could 100 provide alternate mechanistic insights into the diversity of phenotypes observed in Stim 101 and Orai mutant human patients. 102 103 104 105 106 107 108 109 110 5 111 Results: 112 Napahyh/hyh mice harbor severe defects in the production of CD4 T cell 113 effector cytokines 114 Mice bearing Napahyh/hyh mutation on a mixed background have been 115 characterized previously in the context of neurodevelopmental disorders (Bronson and 116 Lane, 1990, Chae et al., 2004, Hong et al., 2004). We backcrossed Napahyh/hyh mice on 117 to C57BL/6 background and found that homozygous mutant Napahyh/hyh mice were 118 significantly smaller in size and died perinatally, within 2-3 weeks. To overcome the 119 issue of perinatal lethality, we generated fetal liver chimeras using irradiated CD45.1+ 120 congenic recipients reconstituted with CD45.2+ wildtype or Napahyh/hyh E15.5 embryos. 121 We analyzed fetal liver chimeras at 8-12 week post-transfer and found that the 122 reconstitution efficiency and total thymic (Figure 1A) and spleen cell numbers (Figure 123 1B) were comparable in wildtype (WT) and Napahyh/hyh chimeras. Relative abundance of 124 CD4 and CD8 T cells in the thymus (Figure 1C) and spleen (Figure 1D) was also normal 125 in Napahyh/hyh fetal liver chimeras. Therefore, we performed all the subsequent analysis 126 of wildtype and Napahyh/hyh CD4 T cells and Foxp3 Tregs using fetal liver chimeras, 127 unless otherwise specified. 128 α-SNAP null mice are embryonic lethal and, in accordance with previous reports, 129 Napahyh/hyh CD4 T cells showed ~40% depletion of α-SNAP levels (Figure 1E). Given 130 the role of α-SNAP in SNARE recycling (Clary et al., 1990), we first compared the levels 131 of cell surface receptors. Surprisingly, surface expression TCR and co-receptors was 132 found to be normal in Napahyh/hyh peripheral CD4 T cells (Figure 1F). Resting Napahyh/hyh 6 133 T lymphocytes showed largely normal surface expression of CD25, CD44 and CD69 134 and their up-regulation following receptor mediated stimulation was comparable to WT 135 (Figure 1G). 136 CRAC channel components, Orai1 and Stim1 are necessary for optimal 137 production and secretion of several T cell effector cytokines (Vig et al., 2008, Vig and 138 Kinet, 2009, Gwack et al., 2008, Oh-Hora et al., 2008). However, given a partial 139 depletion of α-SNAP in Napahyh/hyh mice, we first sought to determine whether 140 Napahyh/hyh CD4 T cells showed defects in the production of effector cytokines. 141 Surprisingly, we found significant defects in IL-2 (Figure 1H, 1J) and TNF-α production 142 by TCR stimulated Napahyh/hyh CD4 T cells (Figure 1I, 1J). Napahyh/hyh CD4 T cells 143 cultured under T helper 1 (Th1) polarizing conditions showed a minor defect in IFN-γ 144 production (Figure 1K & 1M), however, we observed a robust defect in IL-4 expression 145 in Th2 polarized Napahyh/hyh CD4 T cells (Figure 1L & 1M). Intracellular levels of T-bet or 146 Gata-3 did not appear to be significantly altered in Napahyh/hyh mice (Figure 1, figure 147 supplement 1). Furthermore, Napahyh/hyh CD4 T cells (Figure 1N-1O) and splenocytes 148 (Figure 1P) showed a partial defect in anti-CD3 induced proliferation. Taken together, 149 these data demonstrate that Napahyh/hyh CD4 T lymphocytes harbor a significant defect 150 in the production of several key effector cytokines, while exhibiting normal levels of cell 151 surface receptors.
Recommended publications
  • ORAI1 and ORAI2 Modulate Murine Neutrophil Calcium Signaling, Cellular Activation, and Host Defense
    ORAI1 and ORAI2 modulate murine neutrophil calcium signaling, cellular activation, and host defense Derayvia Grimesa,1, Ryan Johnsona,1, Madeline Pashosa, Celeste Cummingsa, Chen Kangb, Georgia R. Sampedroa, Eric Tycksenc, Helen J. McBrided, Rajan Sahb, Clifford A. Lowelle, and Regina A. Clemensa,2 aDepartment of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110; bDepartment of Internal Medicine, Washington University School of Medicine, St. Louis, MO 63110; cMcDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO 63110; dInflammation Research, Amgen, Thousand Oaks, CA 91320; and eDepartment of Laboratory Medicine, University of California, San Francisco, CA 94143 Edited by Michael D. Cahalan, University of California, Irvine, CA, and approved August 11, 2020 (received for review May 5, 2020) Calcium signals are initiated in immune cells by the process of in isoform features, such as activation or inactivation kinetics and store-operated calcium entry (SOCE), where receptor activation sensitivity to modulatory factors, also influence CRAC-channel triggers transient calcium release from the endoplasmic reticulum, function (2–10). ORAI1 and ORAI2 are broadly expressed in im- followed by opening of plasma-membrane calcium-release acti- mune cells, and humans with ORAI1 mutations develop a severe vated calcium (CRAC) channels. ORAI1, ORAI2, and ORAI3 are known combined immunodeficiency-like immunodeficiency, highlighting to comprise the CRAC channel; however, the contributions of indi- the importance of this isoform in immune function (11, 12). In mice, vidual isoforms to neutrophil function are not well understood. ORAI1 also appears to be the dominant functional isoform in im- Here, we show that loss of ORAI1 partially decreases calcium influx, mune cells, with substantial deficits in ORAI1-deficient T cells, while loss of both ORAI1 and ORAI2 completely abolishes SOCE.
    [Show full text]
  • Association of the IP3R to STIM1 Provides a Reduced Intraluminal
    www.nature.com/scientificreports OPEN Association of the IP3R to STIM1 provides a reduced intraluminal calcium microenvironment, Received: 15 March 2018 Accepted: 15 May 2018 resulting in enhanced store- Published: xx xx xxxx operated calcium entry Alicia Sampieri1, Karla Santoyo1, Alexander Asanov2 & Luis Vaca 1 The involvement of inositol trisphosphate receptor (IP3R) in modulating store-operated calcium entry (SOCE) was established many years ago. Nevertheless, the molecular mechanism responsible for this observation has not been elucidated to this date. In the present study we show that IP3R associates to STIM1 upon depletion of the endoplasmic reticulum (ER) by activation of the inositol trisphosphate signaling cascade via G-protein coupled receptors. IP3R-STIM1 association results in enhanced STIM1 puncta formation and larger Orai-mediated whole-cell currents as well as increased calcium infux. Depleting the ER with a calcium ATPase inhibitor (thapsigargin, TG) does not induce IP3R-STIM1 association, indicating that this association requires an active IP3R. The IP3R-STIM1 association is only observed after IP3R activation, as evidenced by FRET experiments and co-immunoprecipitation assays. ER intraluminal calcium measurements using Mag-Fluo-4 showed enhanced calcium depletion when IP3R is overexpressed. A STIM1-GCaMP fusion protein indicates that STIM1 detects lower calcium concentrations near its EF-hand domain when IP3R is overexpressed when compared with the fuorescence reported by a GCaMP homogenously distributed in the ER lumen (ER-GCaMP). All these data together strongly suggest that activation of inositol trisphosphate signaling cascade induces the formation of the IP3R-STIM1 complex. The activated IP3R provides a reduced intraluminal calcium microenvironment near STIM1, resulting in enhanced activation of Orai currents and SOCE.
    [Show full text]
  • Na+ Influx Via Orai1 Inhibits Intracellular ATP-Induced Mtorc2 Signaling to Disrupt CD4 T Cell Gene Expression and Differentiation." Elife.6
    Washington University School of Medicine Digital Commons@Becker Open Access Publications 2017 Na+ influx via Orai1 inhibits intracellular ATP- induced mTORC2 signaling to disrupt CD4 T cell gene expression and differentiation Yong Miao Washington University School of Medicine in St. Louis Jaya Bhushan Washington University School of Medicine in St. Louis Adish Dani Washington University School of Medicine in St. Louis Monika Vig Washington University School of Medicine in St. Louis Follow this and additional works at: https://digitalcommons.wustl.edu/open_access_pubs Recommended Citation Miao, Yong; Bhushan, Jaya; Dani, Adish; and Vig, Monika, ,"Na+ influx via Orai1 inhibits intracellular ATP-induced mTORC2 signaling to disrupt CD4 T cell gene expression and differentiation." Elife.6,. e25155. (2017). https://digitalcommons.wustl.edu/open_access_pubs/6064 This Open Access Publication is brought to you for free and open access by Digital Commons@Becker. It has been accepted for inclusion in Open Access Publications by an authorized administrator of Digital Commons@Becker. For more information, please contact [email protected]. RESEARCH ARTICLE Na+ influx via Orai1 inhibits intracellular ATP-induced mTORC2 signaling to disrupt CD4 T cell gene expression and differentiation Yong Miao1, Jaya Bhushan1, Adish Dani1,2, Monika Vig1* 1Department of Pathology and Immunology, Washington University School of Medicine, St Louis, United States; 2Hope Center for Neurological Disorders, Washington University School of Medicine, St Louis, United States Abstract T cell effector functions require sustained calcium influx. However, the signaling and phenotypic consequences of non-specific sodium permeation via calcium channels remain unknown. a-SNAP is a crucial component of Orai1 channels, and its depletion disrupts the functional assembly of Orai1 multimers.
    [Show full text]
  • KRAP Tethers IP3 Receptors to Actin and Licenses Them to Evoke Cytosolic Ca2+ Signals ✉ ✉ Nagendra Babu Thillaiappan 1,2 , Holly A
    ARTICLE https://doi.org/10.1038/s41467-021-24739-9 OPEN KRAP tethers IP3 receptors to actin and licenses them to evoke cytosolic Ca2+ signals ✉ ✉ Nagendra Babu Thillaiappan 1,2 , Holly A. Smith1, Peace Atakpa-Adaji1 & Colin W. Taylor 1 2+ 2+ Regulation of IP3 receptors (IP3Rs) by IP3 and Ca allows regenerative Ca signals, the 2+ smallest being Ca puffs, which arise from coordinated openings of a few clustered IP3Rs. 2+ Cells express thousands of mostly mobile IP3Rs, yet Ca puffs occur at a few immobile IP3R 1234567890():,; clusters. By imaging cells with endogenous IP3Rs tagged with EGFP, we show that KRas- induced actin-interacting protein (KRAP) tethers IP3Rs to actin beneath the plasma mem- brane. Loss of KRAP abolishes Ca2+ puffs and the global increases in cytosolic Ca2+ con- centration evoked by more intense stimulation. Over-expressing KRAP immobilizes additional 2+ 2+ IP3R clusters and results in more Ca puffs and larger global Ca signals. Endogenous KRAP determines which IP3Rs will respond: it tethers IP3R clusters to actin alongside sites 2+ 2+ where store-operated Ca entry occurs, licenses IP3Rs to evoke Ca puffs and global 2+ cytosolic Ca signals, implicates the actin cytoskeleton in IP3R regulation and may allow local activation of Ca2+ entry. 1 Department of Pharmacology, Tennis Court Road, Cambridge, UK. 2 Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar ✉ University, Doha, Qatar. email: [email protected]; [email protected] NATURE COMMUNICATIONS | (2021) 12:4514 | https://doi.org/10.1038/s41467-021-24739-9 | www.nature.com/naturecommunications 1 ARTICLE NATURE COMMUNICATIONS | https://doi.org/10.1038/s41467-021-24739-9 2+ ytosolic Ca signals regulate diverse activities in all from EGFP-IP3R1 HeLa cells in the same ratio as their overall 1 fi eukaryotic cells .
    [Show full text]
  • A Sulfur-Aromatic Gate Latch Is Essential for Opening of the Orai1 Channel Pore
    RESEARCH ARTICLE A sulfur-aromatic gate latch is essential for opening of the Orai1 channel pore Priscilla S-W Yeung1†, Christopher E Ing2,3†, Megumi Yamashita1, Re´ gis Pome` s2,3, Murali Prakriya1* 1Department of Pharmacology, Northwestern University, Feinberg School of Medicine, Chicago, United States; 2Molecular Medicine, Hospital for Sick Children, Toronto, Canada; 3Department of Biochemistry, University of Toronto, Toronto, Canada Abstract Sulfur-aromatic interactions occur in the majority of protein structures, yet little is known about their functional roles in ion channels. Here, we describe a novel molecular motif, the M101 gate latch, which is essential for gating of human Orai1 channels via its sulfur-aromatic interactions with the F99 hydrophobic gate. Molecular dynamics simulations of different Orai variants reveal that the gate latch is mostly engaged in open but not closed channels. In experimental studies, we use metal-ion bridges to show that promoting an M101-F99 bond directly activates Orai1, whereas disrupting this interaction triggers channel closure. Mutational analysis demonstrates that the methionine residue at this position has a unique combination of length, flexibility, and chemistry to act as an effective latch for the phenylalanine gate. Because sulfur- aromatic interactions provide additional stabilization compared to purely hydrophobic interactions, we infer that the six M101-F99 pairs in the hexameric channel provide a substantial energetic contribution to Orai1 activation. *For correspondence: [email protected] Introduction †These authors contributed The opening and closing of ion channels constitute an important means by which extracellular signals equally to this work are translated into the activation of specific intracellular signaling cascades.
    [Show full text]
  • Supplementary Table S4. FGA Co-Expressed Gene List in LUAD
    Supplementary Table S4. FGA co-expressed gene list in LUAD tumors Symbol R Locus Description FGG 0.919 4q28 fibrinogen gamma chain FGL1 0.635 8p22 fibrinogen-like 1 SLC7A2 0.536 8p22 solute carrier family 7 (cationic amino acid transporter, y+ system), member 2 DUSP4 0.521 8p12-p11 dual specificity phosphatase 4 HAL 0.51 12q22-q24.1histidine ammonia-lyase PDE4D 0.499 5q12 phosphodiesterase 4D, cAMP-specific FURIN 0.497 15q26.1 furin (paired basic amino acid cleaving enzyme) CPS1 0.49 2q35 carbamoyl-phosphate synthase 1, mitochondrial TESC 0.478 12q24.22 tescalcin INHA 0.465 2q35 inhibin, alpha S100P 0.461 4p16 S100 calcium binding protein P VPS37A 0.447 8p22 vacuolar protein sorting 37 homolog A (S. cerevisiae) SLC16A14 0.447 2q36.3 solute carrier family 16, member 14 PPARGC1A 0.443 4p15.1 peroxisome proliferator-activated receptor gamma, coactivator 1 alpha SIK1 0.435 21q22.3 salt-inducible kinase 1 IRS2 0.434 13q34 insulin receptor substrate 2 RND1 0.433 12q12 Rho family GTPase 1 HGD 0.433 3q13.33 homogentisate 1,2-dioxygenase PTP4A1 0.432 6q12 protein tyrosine phosphatase type IVA, member 1 C8orf4 0.428 8p11.2 chromosome 8 open reading frame 4 DDC 0.427 7p12.2 dopa decarboxylase (aromatic L-amino acid decarboxylase) TACC2 0.427 10q26 transforming, acidic coiled-coil containing protein 2 MUC13 0.422 3q21.2 mucin 13, cell surface associated C5 0.412 9q33-q34 complement component 5 NR4A2 0.412 2q22-q23 nuclear receptor subfamily 4, group A, member 2 EYS 0.411 6q12 eyes shut homolog (Drosophila) GPX2 0.406 14q24.1 glutathione peroxidase
    [Show full text]
  • Alternative Splicing Switches STIM1 Targeting to Specialized Membrane Contact Sites and Modifies SOCE
    bioRxiv preprint doi: https://doi.org/10.1101/2020.03.25.005199; this version posted March 25, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. Alternative splicing switches STIM1 targeting to specialized membrane contact sites and modifies SOCE Mona L. Knapp1, Kathrin Förderer1, Dalia Alansary1, Martin Jung3, Yvonne Schwarz4, Annette Lis2, and Barbara A. Niemeyer1, 1Molecular Biophysics, 2Biophysics and 4Molecular Neurophysiology, Center of Integrative Physiology and Molecular Medicine (CIPMM), Bld. 48, 3Medical Biochemistry and Molecular Biology, Bld. 44, Saarland University, 66421 Homburg, Germany Alternative splicing is a potent modifier of protein function. within the first weeks 10. The early lethality indicates es- Stromal interaction molecule 1 (Stim1) is the essential activa- sential roles for Stim1 that are independent of their immune 2+ tor molecule of store-operated Ca entry (SOCE) and a sort- cell function as also indicated by the fact that also gain- ing regulator of certain ER proteins such as Stimulator of in- of-function (GOF) mutations result in multisystemic pheno- terferon genes (STING). Here, we characterize a conserved new types (see above). A mouse model expressing the STIM1 variant, Stim1A, where splice-insertion translates into an ad- gain-of-function mutation R304W, which causes Stormorken ditional C-terminal domain. We find prominent expression of syndrome in humans, also shows very few surviving off- exonA mRNA in testes, astrocytes, kidney and heart and con- firm Stim1A protein in Western blot of testes. In situ, endoge- spring with small size, abnormal bone architecture, abnor- nous Stim1 with domain A, but not Stim1 without domain A mal epithelial cell fate as well as defects in skeletal mus- 11,12 localizes to unique adhesion junctions and to specialized mem- cle, spleen and eye .
    [Show full text]
  • Blockage of Store-Operated Ca2+ Influx by Synta66 Is Mediated by Direct Inhibition of the Ca2+ Selective Orai1 Pore
    cancers Article Blockage of Store-Operated Ca2+ Influx by Synta66 is Mediated by Direct Inhibition of the Ca2+ Selective Orai1 Pore Linda Waldherr 1 , Adela Tiffner 2 , Deepti Mishra 3, Matthias Sallinger 2, Romana Schober 1,2, Irene Frischauf 2 , Tony Schmidt 1 , Verena Handl 4, Peter Sagmeister 5, Manuel Köckinger 5 , Isabella Derler 2, Muammer Üçal 4 , Daniel Bonhenry 3,*, Silke Patz 4,* and Rainer Schindl 1,2,* 1 Gottfried Schatz Research Centre, Medical University of Graz, A-8010 Graz, Austria; [email protected] (L.W.); [email protected] (R.S.); [email protected] (T.S.) 2 Institute of Biophysics, JKU Life Science Centre, Johannes Kepler University Linz, A-4020 Linz, Austria; adela.tiff[email protected] (A.T.); [email protected] (M.S.); [email protected] (I.F.); [email protected] (I.D.) 3 Centre for Nanobiology and Structural Biology, Academy of Sciences of the Czech Republic, 373 33 Nové Hrady, Czech Republic; [email protected] 4 Department of Neurosurgery, Medical University of Graz, A-8010 Graz, Austria; [email protected] (V.H.); [email protected] (M.Ü.) 5 Institute of Chemistry, University of Graz, Heinrichstraße 28, A-8010 Graz, Austria; [email protected] (P.S.); [email protected] (M.K.) * Correspondence: [email protected] (D.B.); [email protected] (S.P.); [email protected] (R.S.) Received: 3 August 2020; Accepted: 30 September 2020; Published: 6 October 2020 Simple Summary: Store-operated calcium channels constituted from the proteins Orai and STIM are important targets for development of new drugs, especially for the treatment of auto-immune diseases.
    [Show full text]
  • Human Induced Pluripotent Stem Cell–Derived Podocytes Mature Into Vascularized Glomeruli Upon Experimental Transplantation
    BASIC RESEARCH www.jasn.org Human Induced Pluripotent Stem Cell–Derived Podocytes Mature into Vascularized Glomeruli upon Experimental Transplantation † Sazia Sharmin,* Atsuhiro Taguchi,* Yusuke Kaku,* Yasuhiro Yoshimura,* Tomoko Ohmori,* ‡ † ‡ Tetsushi Sakuma, Masashi Mukoyama, Takashi Yamamoto, Hidetake Kurihara,§ and | Ryuichi Nishinakamura* *Department of Kidney Development, Institute of Molecular Embryology and Genetics, and †Department of Nephrology, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan; ‡Department of Mathematical and Life Sciences, Graduate School of Science, Hiroshima University, Hiroshima, Japan; §Division of Anatomy, Juntendo University School of Medicine, Tokyo, Japan; and |Japan Science and Technology Agency, CREST, Kumamoto, Japan ABSTRACT Glomerular podocytes express proteins, such as nephrin, that constitute the slit diaphragm, thereby contributing to the filtration process in the kidney. Glomerular development has been analyzed mainly in mice, whereas analysis of human kidney development has been minimal because of limited access to embryonic kidneys. We previously reported the induction of three-dimensional primordial glomeruli from human induced pluripotent stem (iPS) cells. Here, using transcription activator–like effector nuclease-mediated homologous recombination, we generated human iPS cell lines that express green fluorescent protein (GFP) in the NPHS1 locus, which encodes nephrin, and we show that GFP expression facilitated accurate visualization of nephrin-positive podocyte formation in
    [Show full text]
  • STIM1 Phosphorylation at Y361 Recruits Orai1 to STIM1 Puncta And
    www.nature.com/scientificreports OPEN STIM1 Phosphorylation at Y361 Recruits Orai1 to STIM1 Puncta and Induces Ca2+ Entry Received: 05 December 2016 Pascal Yazbeck1, Mohammad Tauseef1,2, Kevin Kruse1, Md-Ruhul Amin1, Rayees Sheikh1, Accepted: 12 January 2017 Stefan Feske3, Yulia Komarova1 & Dolly Mehta1 Published: 20 February 2017 Store-operated Ca2+ entry (SOCE) mediates the increase in intracellular calcium (Ca2+) in endothelial cells (ECs) that regulates several EC functions including tissue-fluid homeostasis. Stromal-interaction molecule 1 (STIM1), upon sensing the depletion of (Ca2+) from the endoplasmic reticulum (ER) store, organizes as puncta that trigger store-operated Ca2+ entry (SOCE) via plasmalemmal Ca2+- selective Orai1 channels. While the STIM1 and Orai1 binding interfaces have been mapped, signaling mechanisms activating STIM1 recruitment of Orai1 and STIM1-Orai1 interaction remains enigmatic. Here, we show that ER Ca2+-store depletion rapidly induces STIM1 phosphorylation at Y361 via proline- rich kinase 2 (Pyk2) in ECs. Surprisingly, the phospho-defective STIM1-Y361F mutant formed puncta but failed to recruit Orai1, thereby preventing. SOCE Furthermore, studies in mouse lungs, expression of phosphodefective STIM1-Y361F mutant in ECs prevented the increase in vascular permeability induced by the thrombin receptor, protease activated receptor 1 (PAR1). Hence, Pyk2-dependent phosphorylation of STIM1 at Y361 is a critical phospho-switch enabling recruitment of Orai1 into STIM1 puncta leading to SOCE. Therefore, Y361 in STIM1 represents a novel target for limiting SOCE- associated vascular leak. Endothelial barrier function is vital in the regulation of tissue-fluid homeostasis, angiogenesis, and inflammation1,2. Loss of endothelial barrier function following burn, trauma, or sepsis leads to acute lung injury (ALI), a life threatening condition due to respiratory failure3,4.
    [Show full text]
  • A Trafficome-Wide Rnai Screen Reveals Deployment of Early and Late Secretory Host Proteins and the Entire Late Endo-/Lysosomal V
    bioRxiv preprint doi: https://doi.org/10.1101/848549; this version posted November 19, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license. 1 A trafficome-wide RNAi screen reveals deployment of early and late 2 secretory host proteins and the entire late endo-/lysosomal vesicle fusion 3 machinery by intracellular Salmonella 4 5 Alexander Kehl1,4, Vera Göser1, Tatjana Reuter1, Viktoria Liss1, Maximilian Franke1, 6 Christopher John1, Christian P. Richter2, Jörg Deiwick1 and Michael Hensel1, 7 8 1Division of Microbiology, University of Osnabrück, Osnabrück, Germany; 2Division of Biophysics, University 9 of Osnabrück, Osnabrück, Germany, 3CellNanOs – Center for Cellular Nanoanalytics, Fachbereich 10 Biologie/Chemie, Universität Osnabrück, Osnabrück, Germany; 4current address: Institute for Hygiene, 11 University of Münster, Münster, Germany 12 13 Running title: Host factors for SIF formation 14 Keywords: siRNA knockdown, live cell imaging, Salmonella-containing vacuole, Salmonella- 15 induced filaments 16 17 Address for correspondence: 18 Alexander Kehl 19 Institute for Hygiene 20 University of Münster 21 Robert-Koch-Str. 4148149 Münster, Germany 22 Tel.: +49(0)251/83-55233 23 E-mail: [email protected] 24 25 or bioRxiv preprint doi: https://doi.org/10.1101/848549; this version posted November 19, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.
    [Show full text]
  • Cardiomyocyte-Specific Deletion of Orai1 Reveals Its Protective Role in Angiotensin-II-Induced Pathological Cardiac Remodeling
    cells Article Cardiomyocyte-Specific Deletion of Orai1 Reveals Its Protective Role in Angiotensin-II-Induced Pathological Cardiac Remodeling Sebastian Segin 1,2, Michael Berlin 1,2, Christin Richter 1, Rebekka Medert 1,2, Veit Flockerzi 3, Paul Worley 4, Marc Freichel 1,2 and Juan E. Camacho Londoño 1,2,* 1 Pharmakologisches Institut, Ruprecht-Karls-Universität Heidelberg, INF 366, 69120 Heidelberg, Germany; [email protected] (S.S.); [email protected] (M.B.); [email protected] (C.R.); [email protected] (R.M.); [email protected] (M.F.) 2 DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, 69120 Heidelberg, Germany 3 Experimentelle und Klinische Pharmakologie und Toxikologie, Universität des Saarlandes, 66421 Homburg, Germany; [email protected] 4 The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, School of Medicine, Baltimore, MD 21205, USA; [email protected] * Correspondence: [email protected]; Tel.: +49-6221-54-86863; Fax: +49-6221-54-8644 Received: 26 March 2020; Accepted: 24 April 2020; Published: 28 April 2020 Abstract: Pathological cardiac remodeling correlates with chronic neurohumoral stimulation and abnormal Ca2+ signaling in cardiomyocytes. Store-operated calcium entry (SOCE) has been described in adult and neonatal murine cardiomyocytes, and Orai1 proteins act as crucial ion-conducting constituents of this calcium entry pathway that can be engaged not only by passive Ca2+ store depletion but also by neurohumoral stimuli such as angiotensin-II. In this study, we, therefore, analyzed the consequences of Orai1 deletion for cardiomyocyte hypertrophy in neonatal and adult cardiomyocytes as well as for other features of pathological cardiac remodeling including cardiac contractile function in vivo.
    [Show full text]