1
|
Ornelas-Guevara R, Diercks BP, Guse AH, Dupont G. Ca 2+ puffs underlie adhesion-triggered Ca 2+ microdomains in T cells. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119808. [PMID: 39151474 DOI: 10.1016/j.bbamcr.2024.119808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 07/29/2024] [Accepted: 08/05/2024] [Indexed: 08/19/2024]
Abstract
Ca2+ signalling is pivotal in T cell activation, an essential process in adaptive immune responses. Key to this activation are Ca2+ microdomains, which are transient increases in cytosolic Ca2+ concentration occurring within narrow regions between the endoplasmic reticulum (ER) and the plasma membrane (PM), lasting a few tens of milliseconds. Adhesion Dependent Ca2+ Microdomains (ADCM) rely on store-operated Ca2+ entry (SOCE) via the ORAI/STIM system. The nanometric scale at which these microdomains form poses challenges for direct experimental observation. Following the previous work of Gil et al. [1], which introduced a three-dimensional model of the ER-PM junction, this study combines a detailed description of the Ca2+ fluxes at the junction with stochastic dynamics of a cluster of D-myo-inositol 1,4,5 trisphosphate receptors (IP3R) located in the ER surrounding the junction. Because the consideration of Ca2+ release through the IP3R calls for the simulation of a portion of the cytoplasm considerably larger than the junction, our study also investigates the spatial distribution of PMCAs, revealing their likely localization outside the ER-PM junction. Simulations indicate that Ca2+ puffs implying the opening of 2-6 IP3Rs create ADCMs by provoking local depletions of ER Ca2+ stimulating Ca2+ entry through the ORAI1 channels. Such conditions allow the reproduction of the amplitude, duration and spatial extent of the observed ADCMs. By integrating advanced computational techniques with insights from experimental studies, our approach provides valuable information on the mechanisms governing early Ca2+ signalling in T cell activation, paving the way for a deeper understanding of immune responses.
Collapse
Affiliation(s)
- Roberto Ornelas-Guevara
- Unit of Theoretical Chronobiology, Université Libre de Bruxelles (ULB), Boulevard du Triomphe, 1, B1050 Brussels, Belgium
| | - Björn-Philipp Diercks
- The Calcium Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Andreas H Guse
- The Calcium Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Geneviève Dupont
- Unit of Theoretical Chronobiology, Université Libre de Bruxelles (ULB), Boulevard du Triomphe, 1, B1050 Brussels, Belgium.
| |
Collapse
|
2
|
Jin S, Wan S, Xiong R, Li Y, Dong T, Guan C. The role of regulatory T cells in vitiligo and therapeutic advances: a mini-review. Inflamm Res 2024; 73:1311-1332. [PMID: 38839628 DOI: 10.1007/s00011-024-01900-w] [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: 03/01/2024] [Revised: 05/21/2024] [Accepted: 05/29/2024] [Indexed: 06/07/2024] Open
Abstract
BACKGROUND Regulatory T cells (Tregs) play vital roles in controlling immune reactions and maintaining immune tolerance in the body. The targeted destruction of epidermal melanocytes by activated CD8+T cells is a key event in the development of vitiligo. However, Tregs may exert immunosuppressive effects on CD8+T cells, which could be beneficial in treating vitiligo. METHODS A comprehensive search of PubMed and Web of Science was conducted to gather information on Tregs and vitiligo. RESULTS In vitiligo, there is a decrease in Treg numbers and impaired Treg functions, along with potential damage to Treg-related signaling pathways. Increasing Treg numbers and enhancing Treg function could lead to immunosuppressive effects on CD8+T cells. Recent research progress on Tregs in vitiligo has been summarized, highlighting various Treg-related therapies being investigated for clinical use. The current status of Treg-related therapeutic strategies and potential future directions for vitiligo treatment are also discussed. CONCLUSIONS A deeper understanding of Tregs will be crucial for advancing Treg-related drug discovery and treatment development in vitiligo.
Collapse
Affiliation(s)
- Shiyu Jin
- Hangzhou Third Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, 310009, China
| | - Sheng Wan
- Hangzhou Third Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, 310009, China
- Department of Dermatology, Hangzhou Third People's Hospital, Hangzhou, 310009, China
| | - Renxue Xiong
- Hangzhou Third Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, 310009, China
- Department of Dermatology, Hangzhou Third People's Hospital, Hangzhou, 310009, China
| | - Yujie Li
- Hangzhou Third Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, 310009, China
| | - Tingru Dong
- Hangzhou Third Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, 310009, China
| | - Cuiping Guan
- Hangzhou Third Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, 310009, China.
- Department of Dermatology, Hangzhou Third People's Hospital, Hangzhou, 310009, China.
| |
Collapse
|
3
|
Morita S, O'Dair MK, Groves JT. Discrete protein condensation events govern calcium signal dynamics in T cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.31.606035. [PMID: 39211144 PMCID: PMC11360922 DOI: 10.1101/2024.07.31.606035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Calcium level variations, which occur downstream of T cell receptor (TCR) signaling, are an essential aspect of T cell antigen recognition. Although coordinated ion channel activities are known to drive calcium oscillations in other cell types, observations of nonperiodic and heterogeneous calcium patterns in T cells are inconsistent with this mechanism. Here, we track the complete ensemble of individual molecular peptide-major histocompatibility complex (pMHC) binding events to TCR, while simultaneously imaging LAT condensation events and calcium level. Individual LAT condensates induce a rapid and additive calcium response, which quickly attenuates upon condensate dissolution. No evidence of cooperativity between LAT condensates or oscillatory calcium response was detected. These results reveal stochastic LAT protein condensation events as a primary driver of calcium signal dynamics in T cells. One-Sentence Summary Ca 2+ fluctuations in T cells reflect stochastic protein condensation events triggered by single molecular antigen-TCR binding.
Collapse
|
4
|
Vaňková L, Bufka J, Křížková V. Pathophysiological and clinical point of view on Kawasaki disease and MIS-C. Pediatr Neonatol 2023; 64:495-504. [PMID: 37453902 PMCID: PMC10286520 DOI: 10.1016/j.pedneo.2023.05.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 04/24/2023] [Accepted: 05/19/2023] [Indexed: 07/18/2023] Open
Abstract
This article compares two important pathophysiological states, Kawasaki disease, and multisystem inflammatory syndrome, in children associated with COVID-19 (MIS-C). Both occur predominantly in children, have a temporal association with an infectious agent, and are associated with immune-system alteration and systemic inflammation under certain circumstances. The two share common pathophysiology, including enhancement of interleukin-1 neutrophils, activation of the inflammasome, pyroptosis, or NETosis. Moreover, the clinical presentation of the diseases overlaps. However, they are indeed two separate diseases, proven by the differences in the epidemiological and etiological aspects and the pathophysiological processes involved in the development and frequency of some clinical signs. This article highlights potentially exciting areas that have not yet been studied in detail, which could help better understand the development of these diseases.
Collapse
Affiliation(s)
- Lenka Vaňková
- Department of Histology and Embryology, Faculty of Medicine in Pilsen, Charles University, Czech Republic.
| | - Jiří Bufka
- Department of Pediatrics, Teaching Hospital in Pilsen, Czech Republic
| | - Věra Křížková
- Department of Histology and Embryology, Faculty of Medicine in Pilsen, Charles University, Czech Republic
| |
Collapse
|
5
|
Neumann J, Van Nieuwenhove E, Terry LE, Staels F, Knebel TR, Welkenhuyzen K, Ahmadzadeh K, Baker MR, Gerbaux M, Willemsen M, Barber JS, Serysheva II, De Waele L, Vermeulen F, Schlenner S, Meyts I, Yule DI, Bultynck G, Schrijvers R, Humblet-Baron S, Liston A. Disrupted Ca 2+ homeostasis and immunodeficiency in patients with functional IP 3 receptor subtype 3 defects. Cell Mol Immunol 2023; 20:11-25. [PMID: 36302985 PMCID: PMC9794825 DOI: 10.1038/s41423-022-00928-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 09/12/2022] [Indexed: 11/27/2022] Open
Abstract
Calcium signaling is essential for lymphocyte activation, with genetic disruptions of store-operated calcium (Ca2+) entry resulting in severe immunodeficiency. The inositol 1,4,5-trisphosphate receptor (IP3R), a homo- or heterotetramer of the IP3R1-3 isoforms, amplifies lymphocyte signaling by releasing Ca2+ from endoplasmic reticulum stores following antigen stimulation. Although knockout of all IP3R isoforms in mice causes immunodeficiency, the seeming redundancy of the isoforms is thought to explain the absence of variants in human immunodeficiency. In this study, we identified compound heterozygous variants of ITPR3 (a gene encoding IP3R subtype 3) in two unrelated Caucasian patients presenting with immunodeficiency. To determine whether ITPR3 variants act in a nonredundant manner and disrupt human immune responses, we characterized the Ca2+ signaling capacity, the lymphocyte response, and the clinical phenotype of these patients. We observed disrupted Ca2+ signaling in patient-derived fibroblasts and immune cells, with abnormal proliferation and activation responses following T-cell receptor stimulation. Reconstitution of IP3R3 in IP3R knockout cell lines led to the identification of variants as functional hypomorphs that showed reduced ability to discriminate between homeostatic and induced states, validating a genotype-phenotype link. These results demonstrate a functional link between defective endoplasmic reticulum Ca2+ channels and immunodeficiency and identify IP3Rs as diagnostic targets for patients with specific inborn errors of immunity. These results also extend the known cause of Ca2+-associated immunodeficiency from store-operated entry to impaired Ca2+ mobilization from the endoplasmic reticulum, revealing a broad sensitivity of lymphocytes to genetic defects in Ca2+ signaling.
Collapse
Affiliation(s)
- Julika Neumann
- VIB Center for Brain and Disease Research, Leuven, Belgium
- Department of Microbiology and Immunology, KU Leuven, Leuven, Belgium
| | - Erika Van Nieuwenhove
- VIB Center for Brain and Disease Research, Leuven, Belgium
- Department of Microbiology and Immunology, KU Leuven, Leuven, Belgium
- UZ Leuven, Leuven, Belgium
| | - Lara E Terry
- Department of Pharmacology and Physiology, University of Rochester, Rochester, NY, 14526, USA
| | - Frederik Staels
- Department of Microbiology and Immunology, KU Leuven, Leuven, Belgium
- UZ Leuven, Leuven, Belgium
| | - Taylor R Knebel
- Department of Pharmacology and Physiology, University of Rochester, Rochester, NY, 14526, USA
| | - Kirsten Welkenhuyzen
- Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine, Leuven Kankerinstituut, KU Leuven, Leuven, Belgium
| | - Kourosh Ahmadzadeh
- Laboratory of Immunobiology, Department Microbiology and Immunology, Rega Institute, KU Leuven, Leuven, Belgium
| | - Mariah R Baker
- Department of Biochemistry and Molecular Biology, Structural Biology Imaging Center, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Margaux Gerbaux
- Department of Microbiology and Immunology, KU Leuven, Leuven, Belgium
- Pediatric Department, Academic Children Hospital Queen Fabiola, Université Libre de Bruxelles, Brussels, Belgium
| | - Mathijs Willemsen
- VIB Center for Brain and Disease Research, Leuven, Belgium
- Department of Microbiology and Immunology, KU Leuven, Leuven, Belgium
| | - John S Barber
- VIB Center for Brain and Disease Research, Leuven, Belgium
- Department of Microbiology and Immunology, KU Leuven, Leuven, Belgium
| | - Irina I Serysheva
- Department of Biochemistry and Molecular Biology, Structural Biology Imaging Center, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Liesbeth De Waele
- Department of Pediatric Neurology, University Hospitals Leuven, Leuven, Belgium
| | | | - Susan Schlenner
- Department of Microbiology and Immunology, KU Leuven, Leuven, Belgium
| | - Isabelle Meyts
- UZ Leuven, Leuven, Belgium.
- Laboratory for Inborn Errors of Immunity, Department of Immunology and Microbiology, KU Leuven, Leuven, Belgium.
| | - David I Yule
- Department of Pharmacology and Physiology, University of Rochester, Rochester, NY, 14526, USA
| | - Geert Bultynck
- Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine, Leuven Kankerinstituut, KU Leuven, Leuven, Belgium
| | - Rik Schrijvers
- UZ Leuven, Leuven, Belgium.
- Laboratory for Allergy and Clinical Immunology and Immunogenetics Research Group, Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium.
| | | | - Adrian Liston
- VIB Center for Brain and Disease Research, Leuven, Belgium.
- Department of Microbiology and Immunology, KU Leuven, Leuven, Belgium.
- Immunology Programme, The Babraham Institute, Babraham Research Campus, Cambridge, CB22 3AT, UK.
| |
Collapse
|
6
|
Lou J, Sagar R, Best MD. Metabolite-Responsive Liposomes Employing Synthetic Lipid Switches Driven by Molecular Recognition Principles. Acc Chem Res 2022; 55:2882-2891. [PMID: 36174148 DOI: 10.1021/acs.accounts.2c00446] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
The ability to exert control over lipid properties, including structure, charge, function, and self-assembly characteristics is a powerful tool that can be implemented to achieve a wide range of biomedical applications. Examples in this arena include the development of caged lipids for controlled activation of signaling properties, metabolic labeling strategies for tracking lipid biosynthesis, lipid activity probes for identifying cognate binding partners, approaches for in situ membrane assembly, and liposome triggered release strategies. In this Account, we describe recent advancements in the latter area entailing the development of stimuli-responsive liposomes through programmable changes to lipid self-assembly properties, which can be harnessed to drive the release of encapsulated contents toward applications including drug delivery. We will focus on an emerging paradigm involving liposomal platforms that are sensitized toward chemical agents ranging from metal cations to small organic molecules that exhibit dysregulation in disease states. This has been achieved by developing synthetic lipid switches that are designed to undergo programmed conformational changes upon the recognition of specific target analytes. These structural alterations are leveraged to perturb the packing of lipids within the membrane and thereby drive the release of encapsulated contents.We provide an overview of the inspiration, design, and characterization of liposomes that selectively respond to wide-ranging target analytes. This series of studies began with the development of calcium-responsive liposomes utilizing a lipid switch inspired by sensors including indo-1. Following this successful demonstration, we next showed that the selectivity of the lipid switch could be altered among different metal cations by producing a liposomal platform for which release is induced through zinc binding. Our next goal was to develop metabolite-responsive liposomes in which switching is driven by molecular recognition events involving phosphorylated small molecules. In this work, screening of lipid switches designed to interact with phosphorylated metabolites led to the identification of liposomal formulations that selectivity release contents in the presence of adenosine triphosphate (ATP). Finally, we were able to modulate the metabolite selectivity by rationally designing a modified lipid switch structure that is activated through complexation of inositol-(1,4,5)-trisphosphate (IP3). These projects show the progression of our approaches for liposome release triggered by molecular recognition principles, building from ion-responsive lipid switches to structures that are activated by small molecules. These "smart" liposomal platforms provide an important addition to the toolbox for controlled cargo release since they respond to ions or small molecules that are commonly overproduced by diseased cells.
Collapse
Affiliation(s)
- Jinchao Lou
- Department of Chemistry, University of Tennessee, 1420 Circle Drive, Knoxville, Tennessee 37996, United States
| | - Ruhani Sagar
- Department of Chemistry, University of Tennessee, 1420 Circle Drive, Knoxville, Tennessee 37996, United States
| | - Michael D Best
- Department of Chemistry, University of Tennessee, 1420 Circle Drive, Knoxville, Tennessee 37996, United States
| |
Collapse
|
7
|
Decreased GZMB, NRP1, ITPR1, and SERPINB9 Transcripts Lead to Reduced Regulatory T Cells Suppressive Capacity in Generalized Vitiligo Patients. J Immunol Res 2022; 2022:3426717. [PMID: 36157881 PMCID: PMC9500245 DOI: 10.1155/2022/3426717] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 06/24/2022] [Accepted: 08/24/2022] [Indexed: 12/02/2022] Open
Abstract
Generalized vitiligo (GV) is an autoimmune skin disease characterized by bilateral white patches over the entire body. Regulatory T cells (Tregs) maintain peripheral tolerance; however, they are found to be reduced in numbers and function in vitiligo patients. The exact mechanism for reduced Treg suppressive capacity is unknown. Therefore, we aimed to assess transcript levels of Tregs-associated immunosuppressive genes (GZMB, NRP1, PDCD1, FASLG, and TNFRS18), regulatory molecules of Tregs suppressive function (SERPINB9, ITPR1, and UBASH3A), and Treg-associated transcription factors (GATA2, GATA3, RUNX1, STAT3, and STAT5) in 52 GV patients and 48 controls by real-time PCR (qPCR). We found significantly reduced GZMB, NRP1, SERPINB9, and ITPR1 transcripts in GV Tregs compared to controls (p = 0.03, p = 0.023, p = 0.0045, and p < 0.0001, respectively). There were 0.44-, 0.45-, 0.32-, and 0.54-fold decrease in GZMB, NRP1, SERPINB9, and ITPR1 transcripts in GV Tregs. Additionally, disease activity and severity-based analyses revealed significantly decreased GZMB (p = 0.019 and 0.034), SERPINB9 (p = 0.031 and p = 0.035), and ITPR1 (p = 0.0003 and p = 0.034) transcripts in active vitiligo and severe GV patients' Tregs. Interestingly, we found a positive correlation for ITPR1 with GZMB (r = 0.45, p = 0.0009) and SERPINB9 (r = 0.52, p = 0.001) transcripts in GV Tregs. Moreover, we found positive correlation for percentage Treg mediated suppression of CD4+ and CD8+T cells with ITPR1 (r = 0.54; r = 0.49), GZMB (r = 0.61; r = 0.58), NRP1 (r = 0.55; r = 0.52), and SERPINB9 (r = 0.56; r = 0.48) in GV Tregs. Further, calcium treatment of Tregs resulted into significantly increased ITPR1, SERPINB9, and GZMB transcripts in GV Tregs (p = 0.023, p = 0.0345, p = 0.02). Overall, our results for the first time revealed the crucial role of GZMB, NRP1, SERPINB9, and ITPR1 transcripts in decreased Treg suppressive capacity leading to GV pathogenesis, progression, and severity. In addition, our study highlighted that ITPR1 might be linked with decreased GZMB and NRP1 expression in GV Tregs. Moreover, our study for the first time suggest that increased SERPINB9 transcripts may lead to endogenous granzyme B-mediated Tregs apoptosis, and calcium treatment of Tregs may improve the Treg suppressive capacity. These findings may further aid in development of Treg-based therapeutics for GV.
Collapse
|
8
|
Store-Operated Ca2+ Entry Is Up-Regulated in Tumour-Infiltrating Lymphocytes from Metastatic Colorectal Cancer Patients. Cancers (Basel) 2022; 14:cancers14143312. [PMID: 35884372 PMCID: PMC9315763 DOI: 10.3390/cancers14143312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 07/03/2022] [Accepted: 07/04/2022] [Indexed: 11/29/2022] Open
Abstract
Simple Summary Store-operated Ca2+ entry (SOCE) has long been known to regulate the differentiation and effector functions of T cells as well as to be instrumental to the ability of cytotoxic T lymphocytes to target cancer cells. Currently, no information is available regarding the expression and function of SOCE in tumour-infiltrating lymphocytes (TILs) that have been expanded in vitro for adoptive cell therapy (ACT). This study provides the first evidence that SOCE is up-regulated in ex vivo-expanded TILs from metastatic colorectal cancer (mCRC) patients. The up-regulation of SOCE mainly depends on diacylglycerol kinase (DGK), which prevents the protein kinase C-dependent inhibition of Ca2+ entry in normal T cells. Of note, the pharmacological blockade of SOCE with the selective inhibitor, BTP-2, during target cell killing significantly increases cytotoxic activity at low TIL density, i.e., when TILs-mediated cancer cell death is rarer. This study, albeit preliminary, could lay the foundation to propose an alternative strategy to effect ACT. It has been shown that ex vivo-expanded TILs did not improve the disease-free survival rate in mCRC patients. Our results strongly suggest that pre-treating autologous TILs with a SOCE or DGK inhibitor before being infused into the patient could improve their cytotoxic activity against cancer cells. Abstract (1) Background: Store-operated Ca2+ entry (SOCE) drives the cytotoxic activity of cytotoxic T lymphocytes (CTLs) against cancer cells. However, SOCE can be enhanced in cancer cells due to an increase in the expression and/or function of its underlying molecular components, i.e., STIM1 and Orai1. Herein, we evaluated the SOCE expression and function in tumour-infiltrating lymphocytes (TILs) from metastatic colorectal cancer (mCRC) patients. (2) Methods: Functional studies were conducted in TILs expanded ex vivo from CRC liver metastases. Peripheral blood T cells from healthy donors (hPBTs) and mCRC patients (cPBTs) were used as controls. (3) Results: SOCE amplitude is enhanced in TILs compared to hPBTs and cPBTs, but the STIM1 protein is only up-regulated in TILs. Pharmacological manipulation showed that the increase in SOCE mainly depends on tonic modulation by diacylglycerol kinase, which prevents the protein kinase C-dependent inhibition of SOCE activity. The larger SOCE caused a stronger Ca2+ response to T-cell receptor stimulation by autologous mCRC cells. Reducing Ca2+ influx with BTP-2 during target cell killing significantly increases cytotoxic activity at low target:effector ratios. (4) Conclusions: SOCE is enhanced in ex vivo-expanded TILs deriving from mCRC patients but decreasing Ca2+ influx with BTP-2 increases cytotoxic activity at a low TIL density.
Collapse
|
9
|
Dridi H, Forrester F, Umanskaya A, Xie W, Reiken S, Lacampagne A, Marks A. Role of oxidation of excitation-contraction coupling machinery in age-dependent loss of muscle function in C. elegans. eLife 2022; 11:75529. [PMID: 35506650 PMCID: PMC9113742 DOI: 10.7554/elife.75529] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 04/27/2022] [Indexed: 11/13/2022] Open
Abstract
Age-dependent loss of body wall muscle function and impaired locomotion occur within 2 weeks in C. elegans; however, the underlying mechanism has not been fully elucidated. In humans, age-dependent loss of muscle function occurs at about 80 years of age and has been linked to dysfunction of ryanodine receptor (RyR)/intracellular calcium (Ca2+) release channels on the sarcoplasmic reticulum (SR). Mammalian skeletal muscle RyR1 channels undergo age-related remodeling due to oxidative overload, leading to loss of the stabilizing subunit calstabin1 (FKBP12) from the channel macromolecular complex. This destabilizes the closed state of the channel resulting in intracellular Ca2+ leak, reduced muscle function, and impaired exercise capacity. We now show that the C. elegans RyR homolog, UNC-68, exhibits a remarkable degree of evolutionary conservation with mammalian RyR channels and similar age-dependent dysfunction. Like RyR1 in mammals UNC-68 encodes a protein that comprises a macromolecular complex which includes the calstabin1 homolog FKB-2 and is immunoreactive with antibodies raised against the RyR1 complex. Further, as in aged mammals, UNC-68 is oxidized and depleted of FKB-2 in an age-dependent manner, resulting in 'leaky' channels, depleted SR Ca2+ stores, reduced body wall muscle Ca2+ transients, and age-dependent muscle weakness. FKB-2 (ok3007)-deficient worms exhibit reduced exercise capacity. Pharmacologically induced oxidization of UNC-68 and depletion of FKB-2 from the channel independently caused reduced body wall muscle Ca2+ transients. Preventing FKB-2 depletion from the UNC-68 macromolecular complex using the Rycal drug S107 improved muscle Ca2+ transients and function. Taken together, these data suggest that UNC-68 oxidation plays a role in age-dependent loss of muscle function. Remarkably, this age-dependent loss of muscle function induced by oxidative overload, which takes ~2 years in mice and ~80 years in humans, occurs in less than 2-3 weeks in C. elegans, suggesting that reduced antioxidant capacity may contribute to the differences in life span amongst species.
Collapse
Affiliation(s)
- Haikel Dridi
- Department of Physiology and Cellular Biophysics, Columbia University Medical Center, New York, United States
| | - Frances Forrester
- Department of Physiology and Cellular Biophysics, Columbia University Medical Center, New York, United States
| | - Alisa Umanskaya
- Department of Physiology and Cellular Biophysics, Columbia University Medical Center, New York, United States
| | - Wenjun Xie
- Department of Physiology and Cellular Biophysics, Columbia University Medical Center, New York, United States
| | - Steven Reiken
- Department of Physiology and Cellular Biophysics, Columbia University Medical Center, New York, United States
| | - Alain Lacampagne
- U1046, Montpellier University, INSERM, CNRS, Montpellier, France
| | - Andrew Marks
- Department of Physiology and Cellular Biophysics, Columbia University Medical Center, New York, United States
| |
Collapse
|
10
|
Bottcher SE, Lou J, Best MD. Liposome triggered content release through molecular recognition of inositol trisphosphate. Chem Commun (Camb) 2022; 58:4520-4523. [PMID: 35302139 DOI: 10.1039/d2cc00951j] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
A stimuli-responsive liposomal platform that is selectively activated by inositol 1,4,5-trisphosphate (IP3) over eleven other phosphorylated metabolites is reported. Dye release assays validated dose-dependent release of both hydrophilic and hydrophobic cargo driven by IP3, showcasing the potential of this platform for triggered release and sensing applications.
Collapse
Affiliation(s)
- Sydney E Bottcher
- Department of Chemistry, University of Tennessee, Knoxville, TN, 37996, USA.
| | - Jinchao Lou
- Department of Chemistry, University of Tennessee, Knoxville, TN, 37996, USA.
| | - Michael D Best
- Department of Chemistry, University of Tennessee, Knoxville, TN, 37996, USA.
| |
Collapse
|
11
|
Lou J, Schuster JA, Barrera FN, Best MD. ATP-Responsive Liposomes via Screening of Lipid Switches Designed to Undergo Conformational Changes upon Binding Phosphorylated Metabolites. J Am Chem Soc 2022; 144:3746-3756. [PMID: 35171601 DOI: 10.1021/jacs.2c00191] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Liposomal delivery vehicles can dramatically enhance drug transport. However, their clinical application requires enhanced control over content release at diseased sites. For this reason, triggered release strategies have been explored, although a limited toolbox of stimuli has thus far been developed. Here, we report a novel strategy for stimuli-responsive liposomes that release encapsulated contents in the presence of phosphorylated small molecules. Our formulation efforts culminated in selective cargo release driven by ATP, a universal energy source that is upregulated in diseases such as cancer. Specifically, we developed lipid switches 1a-b bearing two ZnDPA units designed to undergo substantial conformational changes upon ATP binding, thereby disrupting membrane packing and triggering the release of encapsulated contents. Dye leakage assays using the hydrophobic dye Nile red validated that ATP-driven release was selective over 11 similar phosphorylated metabolites, and release of the hydrophilic dye calcein was also achieved. Multiple alternative lipid switch structures were synthesized and studied (1c-d and 2), which provided insights into the structural features that render 1a-b selective toward ATP-driven release. Importantly, analysis of cellular delivery using fluorescence microscopy in conjunction with pharmacological ATP manipulation showed that liposome delivery was specific, as it increased upon intracellular ATP accumulation, and was inhibited by ATP downregulation. Our new approach shows strong prospects for enhancing the selectivity of release and payload delivery to diseased cells driven by metabolites such as ATP, providing an exciting new paradigm for controlled release.
Collapse
Affiliation(s)
- Jinchao Lou
- Department of Chemistry, University of Tennessee, Knoxville, Tennessee 37996, United States
| | - Jennifer A Schuster
- Department of Biochemistry & Cellular and Molecular Biology, University of Tennessee, Knoxville, Tennessee 37996, United States
| | - Francisco N Barrera
- Department of Biochemistry & Cellular and Molecular Biology, University of Tennessee, Knoxville, Tennessee 37996, United States
| | - Michael D Best
- Department of Chemistry, University of Tennessee, Knoxville, Tennessee 37996, United States
| |
Collapse
|
12
|
Davis LC, Morgan AJ, Galione A. Acidic Ca 2+ stores and immune-cell function. Cell Calcium 2021; 101:102516. [PMID: 34922066 DOI: 10.1016/j.ceca.2021.102516] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 12/03/2021] [Accepted: 12/04/2021] [Indexed: 12/11/2022]
Abstract
Acidic organelles act as intracellular Ca2+ stores; they actively sequester Ca2+ in their lumina and release it to the cytosol upon activation of endo-lysosomal Ca2+ channels. Recent data suggest important roles of endo-lysosomal Ca2+ channels, the Two-Pore Channels (TPCs) and the TRPML channels (mucolipins), in different aspects of immune-cell function, particularly impacting membrane trafficking, vesicle fusion/fission and secretion. Remarkably, different channels on the same acidic vesicles can couple to different downstream physiology. Endo-lysosomal Ca2+ stores can act under different modalities, be they acting alone (via local Ca2+ nanodomains around TPCs/TRPMLs) or in conjunction with the ER Ca2+ store (to either promote or suppress global ER Ca2+ release). These different modalities impinge upon functions as broad as phagocytosis, cell-killing, anaphylaxis, immune memory, thrombostasis, and chemotaxis.
Collapse
Affiliation(s)
- Lianne C Davis
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford, OX1 3QT, UK.
| | - Anthony J Morgan
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford, OX1 3QT, UK
| | - Antony Galione
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford, OX1 3QT, UK.
| |
Collapse
|
13
|
Chiurillo MA, Lander N, Vercesi AE, Docampo R. IP3 receptor-mediated Ca2+ release from acidocalcisomes regulates mitochondrial bioenergetics and prevents autophagy in Trypanosoma cruzi. Cell Calcium 2020; 92:102284. [DOI: 10.1016/j.ceca.2020.102284] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Revised: 08/18/2020] [Accepted: 08/28/2020] [Indexed: 02/07/2023]
|
14
|
Sermersheim M, Kenney AD, Lin PH, McMichael TM, Cai C, Gumpper K, Adesanya TMA, Li H, Zhou X, Park KH, Yount JS, Ma J. MG53 suppresses interferon-β and inflammation via regulation of ryanodine receptor-mediated intracellular calcium signaling. Nat Commun 2020; 11:3624. [PMID: 32681036 PMCID: PMC7368064 DOI: 10.1038/s41467-020-17177-6] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 06/02/2020] [Indexed: 01/19/2023] Open
Abstract
TRIM family proteins play integral roles in the innate immune response to virus infection. MG53 (TRIM72) is essential for cell membrane repair and is believed to be a muscle-specific TRIM protein. Here we show human macrophages express MG53, and MG53 protein expression is reduced following virus infection. Knockdown of MG53 in macrophages leads to increases in type I interferon (IFN) upon infection. MG53 knockout mice infected with influenza virus show comparable influenza virus titres to wild type mice, but display increased morbidity accompanied by more accumulation of CD45+ cells and elevation of IFNβ in the lung. We find that MG53 knockdown results in activation of NFκB signalling, which is linked to an increase in intracellular calcium oscillation mediated by ryanodine receptor (RyR). MG53 inhibits IFNβ induction in an RyR-dependent manner. This study establishes MG53 as a new target for control of virus-induced morbidity and tissue injury.
Collapse
Affiliation(s)
- Matthew Sermersheim
- Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Adam D Kenney
- Department of Microbial Infection and Immunity, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Pei-Hui Lin
- Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Temet M McMichael
- Department of Microbial Infection and Immunity, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Chuanxi Cai
- Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Kristyn Gumpper
- Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - T M Ayodele Adesanya
- Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Haichang Li
- Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Xinyu Zhou
- Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Ki-Ho Park
- Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Jacob S Yount
- Department of Microbial Infection and Immunity, The Ohio State University Wexner Medical Center, Columbus, OH, USA.
| | - Jianjie Ma
- Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, OH, USA.
| |
Collapse
|
15
|
Abstract
Kawasaki disease is an acute febrile illness and systemic vasculitis of unknown aetiology that predominantly afflicts young children, causes coronary artery aneurysms and can result in long-term cardiovascular sequelae. Kawasaki disease is the leading cause of acquired heart disease among children in the USA. Coronary artery aneurysms develop in some untreated children with Kawasaki disease, leading to ischaemic heart disease and myocardial infarction. Although intravenous immunoglobulin (IVIG) treatment reduces the risk of development of coronary artery aneurysms, some children have IVIG-resistant Kawasaki disease and are at increased risk of developing coronary artery damage. In addition, the lack of specific diagnostic tests and biomarkers for Kawasaki disease make early diagnosis and treatment challenging. The use of experimental mouse models of Kawasaki disease vasculitis has considerably improved our understanding of the pathology of the disease and helped characterize the cellular and molecular immune mechanisms contributing to cardiovascular complications, in turn leading to the development of innovative therapeutic approaches. Here, we outline the pathophysiology of Kawasaki disease and summarize and discuss the progress gained from experimental mouse models and their potential therapeutic translation to human disease. This Review outlines the pathophysiology of Kawasaki disease and discusses the progress gained from experimental mouse models and their potential therapeutic translation to human disease. Kawasaki disease is a childhood systemic vasculitis leading to the development of coronary artery aneurysms; it is the leading cause of acquired heart disease in children in developed countries. The cause of Kawasaki disease is unknown, although it is suspected to be triggered by an unidentified infectious pathogen in genetically predisposed children. Kawasaki disease might not be a normal immune response to an unusual environmental stimulus, but rather a genetically determined unusual and uncontrolled immune response to a common stimulus. Although the aetiological agent in humans is unknown, mouse models of Kawasaki disease vasculitis demonstrate similar pathological features and have substantially accelerated discoveries in the field. Genetic and transcriptomic analysis of blood samples from patients with Kawasaki disease and experimental evidence generated using mouse models have demonstrated the critical role of IL-1β in the pathogenesis of this disease and the therapeutic potential of targeting this pathway (currently under investigation in clinical trials).
Collapse
|
16
|
Bhattacharyya ND, Feng CG. Regulation of T Helper Cell Fate by TCR Signal Strength. Front Immunol 2020; 11:624. [PMID: 32508803 PMCID: PMC7248325 DOI: 10.3389/fimmu.2020.00624] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Accepted: 03/19/2020] [Indexed: 12/16/2022] Open
Abstract
T cells are critical in orchestrating protective immune responses to cancer and an array of pathogens. The interaction between a peptide MHC (pMHC) complex on antigen presenting cells (APCs) and T cell receptors (TCRs) on T cells initiates T cell activation, division, and clonal expansion in secondary lymphoid organs. T cells must also integrate multiple T cell-intrinsic and extrinsic signals to acquire the effector functions essential for the defense against invading microbes. In the case of T helper cell differentiation, while innate cytokines have been demonstrated to shape effector CD4+ T lymphocyte function, the contribution of TCR signaling strength to T helper cell differentiation is less understood. In this review, we summarize the signaling cascades regulated by the strength of TCR stimulation. Various mechanisms in which TCR signal strength controls T helper cell expansion and differentiation are also discussed.
Collapse
Affiliation(s)
- Nayan D Bhattacharyya
- Immunology and Host Defense Group, Discipline of Infectious Diseases and Immunology, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia.,Tuberculosis Research Program, Centenary Institute, The University of Sydney, Sydney, NSW, Australia
| | - Carl G Feng
- Immunology and Host Defense Group, Discipline of Infectious Diseases and Immunology, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia.,Tuberculosis Research Program, Centenary Institute, The University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
17
|
Abstract
Calcium (Ca2+) signalling is of paramount importance to immunity. Regulated increases in cytosolic and organellar Ca2+ concentrations in lymphocytes control complex and crucial effector functions such as metabolism, proliferation, differentiation, antibody and cytokine secretion and cytotoxicity. Altered Ca2+ regulation in lymphocytes leads to various autoimmune, inflammatory and immunodeficiency syndromes. Several types of plasma membrane and organellar Ca2+-permeable channels are functional in T cells. They contribute highly localized spatial and temporal Ca2+ microdomains that are required for achieving functional specificity. While the mechanistic details of these Ca2+ microdomains are only beginning to emerge, it is evident that through crosstalk, synergy and feedback mechanisms, they fine-tune T cell signalling to match complex immune responses. In this article, we review the expression and function of various Ca2+-permeable channels in the plasma membrane, endoplasmic reticulum, mitochondria and endolysosomes of T cells and their role in shaping immunity and the pathogenesis of immune-mediated diseases.
Collapse
Affiliation(s)
- Mohamed Trebak
- Department of Cellular and Molecular Physiology, Pennsylvania State University College of Medicine, Hershey, PA, USA.
| | - Jean-Pierre Kinet
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
18
|
Kushnir A, Santulli G, Reiken SR, Coromilas E, Godfrey SJ, Brunjes DL, Colombo PC, Yuzefpolskaya M, Sokol SI, Kitsis RN, Marks AR. Ryanodine Receptor Calcium Leak in Circulating B-Lymphocytes as a Biomarker in Heart Failure. Circulation 2019; 138:1144-1154. [PMID: 29593014 DOI: 10.1161/circulationaha.117.032703] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
BACKGROUND Advances in congestive heart failure (CHF) management depend on biomarkers for monitoring disease progression and therapeutic response. During systole, intracellular Ca2+ is released from the sarcoplasmic reticulum into the cytoplasm through type-2 ryanodine receptor/Ca2+ release channels. In CHF, chronically elevated circulating catecholamine levels cause pathological remodeling of type-2 ryanodine receptor/Ca2+ release channels resulting in diastolic sarcoplasmic reticulum Ca2+ leak and decreased myocardial contractility. Similarly, skeletal muscle contraction requires sarcoplasmic reticulum Ca2+ release through type-1 ryanodine receptors (RyR1), and chronically elevated catecholamine levels in CHF cause RyR1-mediated sarcoplasmic reticulum Ca2+ leak, contributing to myopathy and weakness. Circulating B-lymphocytes express RyR1 and catecholamine-responsive signaling cascades, making them a potential surrogate for defects in intracellular Ca2+ handling because of leaky RyR channels in CHF. METHODS Whole blood was collected from patients with CHF, CHF following left-ventricular assist device implant, and controls. Blood was also collected from mice with ischemic CHF, ischemic CHF+S107 (a drug that specifically reduces RyR channel Ca2+ leak), and wild-type controls. Channel macromolecular complex was assessed by immunostaining RyR1 immunoprecipitated from lymphocyte-enriched preparations. RyR1 Ca2+ leak was assessed using flow cytometry to measure Ca2+ fluorescence in B-lymphocytes in the absence and presence of RyR1 agonists that empty RyR1 Ca2+ stores within the endoplasmic reticulum. RESULTS Circulating B-lymphocytes from humans and mice with CHF exhibited remodeled RyR1 and decreased endoplasmic reticulum Ca2+ stores, consistent with chronic intracellular Ca2+ leak. This Ca2+ leak correlated with circulating catecholamine levels. The intracellular Ca2+ leak was significantly reduced in mice treated with the Rycal S107. Patients with CHF treated with left-ventricular assist devices exhibited a heterogeneous response. CONCLUSIONS In CHF, B-lymphocytes exhibit remodeled leaky RyR1 channels and decreased endoplasmic reticulum Ca2+ stores consistent with chronic intracellular Ca2+ leak. RyR1-mediated Ca2+ leak in B-lymphocytes assessed using flow cytometry provides a surrogate measure of intracellular Ca2+ handling and systemic sympathetic burden, presenting a novel biomarker for monitoring response to pharmacological and mechanical CHF therapy.
Collapse
Affiliation(s)
- Alexander Kushnir
- Department of Physiology and Cellular Biophysics, Clyde and Helen Wu Center for Molecular Cardiology, Columbia University College of Physicians and Surgeons, New York (A.K., G.S., S.R.R., A.R.M.).,Department of Medicine, Division of Cardiology, Columbia University Medical Center, New York (A.K., E.C., S.J.G., D.L.B., P.C.C., M.Y., A.R.M.)
| | - Gaetano Santulli
- Department of Physiology and Cellular Biophysics, Clyde and Helen Wu Center for Molecular Cardiology, Columbia University College of Physicians and Surgeons, New York (A.K., G.S., S.R.R., A.R.M.)
| | - Steven R Reiken
- Department of Physiology and Cellular Biophysics, Clyde and Helen Wu Center for Molecular Cardiology, Columbia University College of Physicians and Surgeons, New York (A.K., G.S., S.R.R., A.R.M.)
| | - Ellie Coromilas
- Department of Medicine, Division of Cardiology, Columbia University Medical Center, New York (A.K., E.C., S.J.G., D.L.B., P.C.C., M.Y., A.R.M.)
| | - Sarah J Godfrey
- Department of Medicine, Division of Cardiology, Columbia University Medical Center, New York (A.K., E.C., S.J.G., D.L.B., P.C.C., M.Y., A.R.M.)
| | - Danielle L Brunjes
- Department of Medicine, Division of Cardiology, Columbia University Medical Center, New York (A.K., E.C., S.J.G., D.L.B., P.C.C., M.Y., A.R.M.)
| | - Paolo C Colombo
- Department of Medicine, Division of Cardiology, Columbia University Medical Center, New York (A.K., E.C., S.J.G., D.L.B., P.C.C., M.Y., A.R.M.)
| | - Melana Yuzefpolskaya
- Department of Medicine, Division of Cardiology, Columbia University Medical Center, New York (A.K., E.C., S.J.G., D.L.B., P.C.C., M.Y., A.R.M.)
| | - Seth I Sokol
- Department of Medicine, Division of Cardiology, Jacobi Medical Center, Bronx, NY (S.I.S.)
| | - Richard N Kitsis
- Departments of Medicine and Cell Biology, Wilf Family Cardiovascular Research Institute, Albert Einstein Cancer Center, Diabetes Research Center, Albert Einstein College of Medicine, Bronx, NY (R.N.K.)
| | - Andrew R Marks
- Department of Physiology and Cellular Biophysics, Clyde and Helen Wu Center for Molecular Cardiology, Columbia University College of Physicians and Surgeons, New York (A.K., G.S., S.R.R., A.R.M.).,Department of Medicine, Division of Cardiology, Columbia University Medical Center, New York (A.K., E.C., S.J.G., D.L.B., P.C.C., M.Y., A.R.M.)
| |
Collapse
|
19
|
Potapenko E, Negrão NW, Huang G, Docampo R. The acidocalcisome inositol-1,4,5-trisphosphate receptor of Trypanosoma brucei is stimulated by luminal polyphosphate hydrolysis products. J Biol Chem 2019; 294:10628-10637. [PMID: 31138655 DOI: 10.1074/jbc.ra119.007906] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 05/27/2019] [Indexed: 12/21/2022] Open
Abstract
Acidocalcisomes are acidic calcium stores rich in polyphosphate (polyP) and are present in trypanosomes and also in a diverse range of other organisms. Ca2+ is released from these organelles through a channel, inositol 1,4,5-trisphosphate receptor (TbIP3R), which is essential for growth and infectivity of the parasite Trypanosoma brucei However, the mechanism by which TbIP3R controls Ca2+ release is unclear. In this work, we expressed TbIP3R in a chicken B lymphocyte cell line in which the genes for all three vertebrate IP3Rs were stably ablated (DT40-3KO). We show that IP3-mediated Ca2+ release depends on Ca2+ but not on ATP concentration and is inhibited by heparin, caffeine, and 2-aminomethoxydiphenyl borate (2-APB). Excised patch clamp recordings from nuclear membranes of DT40 cells expressing only TbIP3R disclosed that luminal inorganic orthophosphate (Pi) or pyrophosphate (PPi), and neutral or alkaline pH can stimulate IP3-generated currents. In contrast, polyP or acidic pH did not induce these currents, and nuclear membranes obtained from cells expressing rat IP3R were unresponsive to polyP or its hydrolysis products. Our results are consistent with the notion that polyP hydrolysis products within acidocalcisomes or alkalinization of their luminal pH activate TbIP3R and Ca2+ release. We conclude that TbIP3R is well-adapted to its role as the major Ca2+ release channel of acidocalcisomes in T. brucei.
Collapse
Affiliation(s)
| | - Núria W Negrão
- From the Center for Tropical and Emerging Global Diseases and.,Department of Cellular Biology, University of Georgia, Athens, Georgia 30602
| | - Guozhong Huang
- From the Center for Tropical and Emerging Global Diseases and
| | - Roberto Docampo
- From the Center for Tropical and Emerging Global Diseases and .,Department of Cellular Biology, University of Georgia, Athens, Georgia 30602
| |
Collapse
|
20
|
Diener C, Hart M, Alansary D, Poth V, Walch-Rückheim B, Menegatti J, Grässer F, Fehlmann T, Rheinheimer S, Niemeyer BA, Lenhof HP, Keller A, Meese E. Modulation of intracellular calcium signaling by microRNA-34a-5p. Cell Death Dis 2018; 9:1008. [PMID: 30262862 PMCID: PMC6160487 DOI: 10.1038/s41419-018-1050-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 09/10/2018] [Accepted: 09/10/2018] [Indexed: 12/21/2022]
Abstract
Adjusting intracellular calcium signaling is an important feature in the regulation of immune cell function and survival. Here we show that miR-34a-5p, a small non-coding RNA that is deregulated in many common diseases, is a regulator of store-operated Ca2+ entry (SOCE) and calcineurin signaling. Upon miR-34a-5p overexpression, we observed both a decreased depletion of ER calcium content and a decreased Ca2+ influx through Ca2+ release-activated Ca2+ channels. Based on an in silico target prediction we identified multiple miR-34a-5p target genes within both pathways that are implicated in the balance between T-cell activation and apoptosis including ITPR2, CAMLG, STIM1, ORAI3, RCAN1, PPP3R1, and NFATC4. Functional analysis revealed a decrease in Ca2+ activated calcineurin pathway activity measured by a reduced IL-2 secretion due to miR-34a-5p overexpression. Impacting SOCE and/or downstream calcineurin/NFAT signaling by miR-34a-5p offers a possible future approach to manipulate immune cells for clinical interventions.
Collapse
Affiliation(s)
- Caroline Diener
- Institute of Human Genetics, Saarland University, 66421, Homburg, Germany.
| | - Martin Hart
- Institute of Human Genetics, Saarland University, 66421, Homburg, Germany
| | - Dalia Alansary
- Molecular Biophysics, Center for Integrative Physiology and Molecular Medicine, School of Medicine, Saarland University, 66421, Homburg, Germany
| | - Vanessa Poth
- Molecular Biophysics, Center for Integrative Physiology and Molecular Medicine, School of Medicine, Saarland University, 66421, Homburg, Germany
| | - Barbara Walch-Rückheim
- Institute of Virology and Center of Human and Molecular Biology, Saarland University, 66421, Homburg, Germany
| | - Jennifer Menegatti
- Institute of Virology and Center of Human and Molecular Biology, Medical School, Saarland University, 66421, Homburg, Germany
| | - Friedrich Grässer
- Institute of Virology and Center of Human and Molecular Biology, Medical School, Saarland University, 66421, Homburg, Germany
| | - Tobias Fehlmann
- Chair for Clinical Bioinformatics, Saarland University, 66123, Saarbrücken, Germany
| | | | - Barbara A Niemeyer
- Molecular Biophysics, Center for Integrative Physiology and Molecular Medicine, School of Medicine, Saarland University, 66421, Homburg, Germany
| | - Hans-Peter Lenhof
- Center for Bioinformatics, Saarland Informatics Campus, Saarland University, 66123, Saarbrücken, Germany
| | - Andreas Keller
- Chair for Clinical Bioinformatics, Saarland University, 66123, Saarbrücken, Germany
| | - Eckart Meese
- Institute of Human Genetics, Saarland University, 66421, Homburg, Germany
| |
Collapse
|
21
|
ITPR3 gene haplotype is associated with cervical squamous cell carcinoma risk in Taiwanese women. Oncotarget 2018; 8:10085-10090. [PMID: 28036301 PMCID: PMC5354643 DOI: 10.18632/oncotarget.14341] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 12/15/2016] [Indexed: 11/25/2022] Open
Abstract
Host immunogenetic background plays an important role in human papillomavirus (HPV) infection and cervical cancer development. Inositol 1,4,5-triphosphate receptor type 3 (ITPR3) is essential for both immune activation and cancer pathogenesis. We aim to investigate if ITPR3 genetic polymorphisms are associated with the risk of cervical cancer in Taiwanese women. ITPR3 rs3748079 A/G and rs2229634 C/T polymorphisms were genotyped in a hospital-based study of 462 women with cervical squamous cell carcinoma (CSCC) and 921 age-matched healthy control women. The presence and genotypes of HPV in CSCC was determined. No significant association of individual ITPR3 variants were found among controls, CSCC, and HPV-16 positive CSCC. However, we found a significant association of haplotype AT between CSCC and controls (OR = 2.28, 95% CI 1.31-3.97, P = 2.83 × 10-3) and the OR increased further in CSCC patients infected with HPV-16 (OR = 2.89, 95% CI 1.55-5.37, P = 4.54 × 10-4). The linkage disequilibrium analysis demonstrated that ITPR3 association with CSCC was independent of HLA-DRB1 alleles. In conclusion, these findings suggest that AT haplotype in the ITPR3 gene may serve as a potential marker for genetic susceptibility to CSCC.
Collapse
|
22
|
Thaxton JE, Wallace C, Riesenberg B, Zhang Y, Paulos CM, Beeson CC, Liu B, Li Z. Modulation of Endoplasmic Reticulum Stress Controls CD4 + T-cell Activation and Antitumor Function. Cancer Immunol Res 2017. [PMID: 28642246 DOI: 10.1158/2326-6066.cir-17-0081] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The endoplasmic reticulum (ER) is an energy-sensing organelle with intimate ties to programming cell activation and metabolic fate. T-cell receptor (TCR) activation represents a form of acute cell stress and induces mobilization of ER Ca2+ stores. The role of the ER in programming T-cell activation and metabolic fate remains largely undefined. Gp96 is an ER protein with functions as a molecular chaperone and Ca2+ buffering protein. We hypothesized that the ER stress response may be important for CD4+ T-cell activation and that gp96 may be integral to this process. To test our hypothesis, we utilized genetic deletion of the gp96 gene Hsp90b1 in a CD4+ T cell-specific manner. We show that gp96-deficient CD4+ T cells cannot undergo activation-induced glycolysis due to defective Ca2+ mobilization upon TCR engagement. We found that activating naïve CD4+ T cells while inhibiting ER Ca2+ exchange, through pharmacological blockade of the ER Ca2+ channel inositol trisphosphate receptor (IP3R), led to a reduction in cytosolic Ca2+ content and generated a pool of CD62Lhigh/CD44low CD4+ T cells compared with wild-type (WT) matched controls. In vivo IP3R-inhibited CD4+ T cells exhibited elevated tumor control above WT T cells. Together, these data show that ER-modulated cytosolic Ca2+ plays a role in defining CD4+ T-cell phenotype and function. Factors associated with the ER stress response are suitable targets for T cell-based immunotherapies. Cancer Immunol Res; 5(8); 666-75. ©2017 AACR.
Collapse
Affiliation(s)
- Jessica E Thaxton
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, South Carolina.,Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina.,Department of Orthopaedic Surgery, Medical University of South Carolina, Charleston, South Carolina
| | - Caroline Wallace
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, South Carolina
| | - Brian Riesenberg
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, South Carolina
| | - Yongliang Zhang
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, South Carolina
| | - Chrystal M Paulos
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, South Carolina.,Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina
| | - Craig C Beeson
- Department of Pharmaceutical and Biomedical Sciences, Medical University of South Carolina, Charleston, South Carolina
| | - Bei Liu
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, South Carolina.,Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina
| | - Zihai Li
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, South Carolina. .,Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina.,First Affiliated Hospital, Zhengzhou University School of Medicine, Zhengzhou, China
| |
Collapse
|
23
|
Santulli G, Nakashima R, Yuan Q, Marks AR. Intracellular calcium release channels: an update. J Physiol 2017; 595:3041-3051. [PMID: 28303572 PMCID: PMC5430224 DOI: 10.1113/jp272781] [Citation(s) in RCA: 158] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 02/20/2017] [Indexed: 12/19/2022] Open
Abstract
Ryanodine receptors (RyRs) and inositol 1,4,5-trisphosphate receptors (IP3 Rs) are calcium (Ca2+ ) release channels on the endo/sarcoplasmic reticulum (ER/SR). Here we summarize the latest advances in the field, describing the recently discovered mechanistic roles of intracellular Ca2+ release channels in the regulation of mitochondrial fitness and endothelial function, providing novel therapeutic options for the treatment of heart failure, hypertension, and diabetes mellitus.
Collapse
Affiliation(s)
- Gaetano Santulli
- The Wu Center for Molecular CardiologyColumbia UniversityNew YorkNYUSA
- Department of Physiology and Cellular BiophysicsCollege of Physicians and SurgeonsColumbia University Medical CenterNew YorkNYUSA
| | - Ryutaro Nakashima
- The Wu Center for Molecular CardiologyColumbia UniversityNew YorkNYUSA
- Department of Physiology and Cellular BiophysicsCollege of Physicians and SurgeonsColumbia University Medical CenterNew YorkNYUSA
| | - Qi Yuan
- The Wu Center for Molecular CardiologyColumbia UniversityNew YorkNYUSA
- Department of Physiology and Cellular BiophysicsCollege of Physicians and SurgeonsColumbia University Medical CenterNew YorkNYUSA
| | - Andrew R. Marks
- The Wu Center for Molecular CardiologyColumbia UniversityNew YorkNYUSA
- Department of Physiology and Cellular BiophysicsCollege of Physicians and SurgeonsColumbia University Medical CenterNew YorkNYUSA
- Department of MedicineColumbia UniversityNew YorkNYUSA
| |
Collapse
|
24
|
Fos C, Becart S, Canonigo Balancio AJ, Boehning D, Altman A. Association of the EF-hand and PH domains of the guanine nucleotide exchange factor SLAT with IP₃ receptor 1 promotes Ca²⁺ signaling in T cells. Sci Signal 2014; 7:ra93. [PMID: 25270259 DOI: 10.1126/scisignal.2005565] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The guanine nucleotide exchange factor SLAT (SWAP-70-like adaptor of T cells) regulates T cell activation and differentiation by enabling Ca(2+) release from intracellular stores in response to stimulation of the T cell receptor (TCR). We found a TCR-induced association between SLAT and inositol 1,4,5-trisphosphate (IP3) receptor type 1 (IP3R1). The N-terminal region of SLAT, which contains two EF-hand motifs that we determined bound Ca(2+), and the SLAT pleckstrin homology (PH) domain independently bound to IP3R1 by associating with a conserved motif within the IP3R1 ligand-binding domain. Disruption of the SLAT-IP3R1 interaction with cell-permeable, IP3R1-based fusion peptides inhibited TCR-stimulated Ca(2+) signaling, activation of the transcription factor NFAT (nuclear factor of activated T cells), and production of cytokines, suggesting that this interaction is required for optimal T cell activation. The finding that SLAT is an IP3R1-interacting protein required for T cell activation suggests that this interaction could be a potential target for a selective immunosuppressive drug.
Collapse
Affiliation(s)
- Camille Fos
- Division of Cell Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037, USA
| | - Stephane Becart
- Division of Cell Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037, USA
| | - Ann J Canonigo Balancio
- Division of Cell Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037, USA
| | - Darren Boehning
- Department of Biochemistry and Molecular Biology, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Amnon Altman
- Division of Cell Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037, USA.
| |
Collapse
|
25
|
Alkylglycerols modulate the proliferation and differentiation of non-specific agonist and specific antigen-stimulated splenic lymphocytes. PLoS One 2014; 9:e96207. [PMID: 24763671 PMCID: PMC3999215 DOI: 10.1371/journal.pone.0096207] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2014] [Accepted: 04/04/2014] [Indexed: 12/02/2022] Open
Abstract
Alkylglycerols (AKGs) are ether-linked glycerols derived from shark liver oil and found in small amounts in human milk. Previous studies showed that oral AKGs administration significantly increased the immune response in mice. The aim of the present study was to investigate the in vitro immunomodulatory effect of AKGs on stimulating splenic lymphocyte responses. C57BL/6 mice were immunized with hepatitis B surface antigen (HBsAg). Splenic B cells were purified and stimulated with anti-BCR and anti-CD38. Meanwhile, splenic CD4+ T cells were purified and stimulated with anti-CD3 and anti-CD28. For antigen specific stimulation, the purified CD4+ T cells were cocultured with HBsAg -pulsed dendritic cells. The stimulated lymphocytes were treated with different concentrations of AKGs. The cell proliferation was assessed by [3H]-thymidine incorporation assay. The maturation of B cells was assessed by examining the germline (GL) transcription of IgG (γ1) mRNA expression, and the surface expressions of CD80/CD86 markers were examined by flow cytometry analysis. Th1/Th2 polarity was assessed by T-BET (Th1)/GATA-3 (Th2) flow cytometry assay and by characteristic cytokines ELISA assay (TNF-α and IFN-γ for Th1; IL-4 and IL-10 for Th2). It was found that AKGs significantly increased the BCR/CD38 -stimulated B cell proliferation. The T cell proliferation in response to CD3/CD28 or specific antigen stimulation was also increased by AKGs. The transcriptional level of IgG (γ1) and the expressions of CD80/CD86 molecules were markedly increased by AKGs in BCR/CD38 -stimulated B cells. Meanwhile, the results showed that AKGs increased the expression of T-BET transcriptional factor and the production of Th1 cytokines (TNF-α and IFN-γ) upon CD3/CD28 stimulation; whereas, levels of Th2 cytokines (IL-4 and IL-10) were decreased by AKGs. Our study demonstrated that AKGs can modulate immune responses by boosting the proliferation and maturation of murine lymphocytes in vitro.
Collapse
|
26
|
Brint E, O’Callaghan G, Houston A. Life in the Fas lane: differential outcomes of Fas signaling. Cell Mol Life Sci 2013; 70:4085-99. [PMID: 23579628 PMCID: PMC11113183 DOI: 10.1007/s00018-013-1327-z] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2012] [Revised: 03/14/2013] [Accepted: 03/18/2013] [Indexed: 12/11/2022]
Abstract
Fas, also known as CD95 or APO-1, is a member of the tumor necrosis factor/nerve growth factor superfamily. Although best characterized in terms of its apoptotic function, recent studies have identified several other cellular responses emanating from Fas. These responses include migration, invasion, inflammation, and proliferation. In this review, we focus on the diverse cellular outcomes of Fas signaling and the molecular switches identified to date that regulate its pro- and anti-apoptotic functions. Such switches occur at different levels of signal transduction, ranging from the receptor through to cross-talk with other signaling pathways. Factors identified to date including other extracellular signals, proteins recruited to the death-inducing signaling complex, and the availability of different intracellular components of signal transduction pathways. The success of therapeutically targeting Fas will require a better understanding of these pathways, as well as the regulatory mechanisms that determine cellular outcome following receptor activation.
Collapse
Affiliation(s)
- Elizabeth Brint
- Department of Pathology, University College Cork, National University of Ireland, Cork, Ireland
| | - Grace O’Callaghan
- Department of Medicine, University College Cork, National University of Ireland, Cork, Ireland
| | - Aileen Houston
- Department of Medicine, University College Cork, National University of Ireland, Cork, Ireland
| |
Collapse
|
27
|
Ets-1 facilitates nuclear entry of NFAT proteins and their recruitment to the IL-2 promoter. Proc Natl Acad Sci U S A 2013; 110:15776-81. [PMID: 24019486 DOI: 10.1073/pnas.1304343110] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
E26 transformation-specific sequence 1 (Ets-1), the prototype of the ETS family of transcription factors, is critical for the expression of IL-2 by murine Th cells; however, its mechanism of action is still unclear. Here we show that Ets-1 is also essential for optimal production of IL-2 by primary human Th cells. Although Ets-1 negatively regulates the expression of Blimp1, a known suppressor of IL-2 expression, ablation of B lymphocyte-induced maturation protein 1 (Blimp1) does not rescue the expression of IL-2 by Ets-1-deficient Th cells. Instead, Ets-1 physically and functionally interacts with the nuclear factor of activated T-cells (NFAT) and is required for the recruitment of NFAT to the IL-2 promoter. In addition, Ets-1 is located in both the nucleus and cytoplasm of resting Th cells. Nuclear Ets-1 quickly exits the nucleus in response to calcium-dependent signals and competes with NFAT proteins for binding to protein components of noncoding RNA repressor of NFAT complex (NRON), which serves as a cytoplasmic trap for phosphorylated NFAT proteins. This nuclear exit of Ets-1 precedes rapid nuclear entry of NFAT and Ets-1 deficiency results in impaired nuclear entry, but not dephosphorylation, of NFAT proteins. Thus, Ets-1 promotes the expression of IL-2 by modulating the activity of NFAT.
Collapse
|
28
|
Tespa1 is a novel component of mitochondria-associated endoplasmic reticulum membranes and affects mitochondrial calcium flux. Biochem Biophys Res Commun 2013; 433:322-6. [DOI: 10.1016/j.bbrc.2013.02.099] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2013] [Accepted: 02/28/2013] [Indexed: 12/20/2022]
|
29
|
Fujimoto T, Matsuzaki H, Tanaka M, Shirasawa S. Tespa1 protein is phosphorylated in response to store-operated calcium entry. Biochem Biophys Res Commun 2013; 434:162-5. [PMID: 23541577 DOI: 10.1016/j.bbrc.2013.02.128] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2013] [Accepted: 02/27/2013] [Indexed: 10/27/2022]
Abstract
We previously reported that Tespa1 (thymocyte-expressed, positive selection-associated gene 1) protein expressed in lymphocytes physically interacts with IP3R (Inositol 1,4,5-trisphosphate receptor), a Ca(2+) channel protein spanning endoplasmic reticulum (ER) membrane. However, the biochemical characterization of Tespa1 protein remains unknown. In this study, we have found that Tespa1 protein was posttranslationally modified upon intracellular Ca(2+) increase in thymocytes. Through the analyses using various inhibitors, store-operated Ca(2+) entry (SOCE) was found to be an essential factor for the Tespa1 protein modification induced by T cell receptor (TCR)-stimulation. Remarkably, the Ca(2+)-dependent Tespa1 protein modification was restored by in vitro protein phosphatase treatment, indicating that this modification was due to phosphorylation. Moreover, we examined whether Ca(2+)-dependent phosphorylation of Tespa1 protein would affect the physical association between Tespa1 and IP3R proteins, revealing that physical association of these proteins is maintained regardless of the presence or absence of phosphorylation of Tespa1. In addition, KRAP protein which represents substantial amino acid sequence homology to Tespa1 was also posttranslationally phosphorylated by intracellular Ca(2+) increase in HCT116 human colon cancer cells and HEK293 human embryonic kidney cells, suggesting that common signaling mechanism(s) may contribute to the molecular modification of Tespa1 and KRAP in different cellular processes. All these results suggested a novel molecular modification of Tespa1 and the existence of the regulatory pathway that SOCE affects the Tespa1-IP3R molecular complex.
Collapse
Affiliation(s)
- Takahiro Fujimoto
- Department of Cell Biology, Faculty of Medicine, Fukuoka University, 7-45-1 Nanakuma, Fukuoka 814-0180, Japan
| | | | | | | |
Collapse
|
30
|
Fujimoto T, Shirasawa S. Identification of KRAP-expressing cells and the functional relevance of KRAP to the subcellular localization of IP3R in the stomach and kidney. Int J Mol Med 2012; 30:1287-93. [PMID: 22992961 PMCID: PMC4042864 DOI: 10.3892/ijmm.2012.1126] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2012] [Accepted: 08/20/2012] [Indexed: 12/31/2022] Open
Abstract
KRAS-induced actin-interacting protein (KRAP), originally identified as
one of the deregulated genes expressed in colorectal cancer, participates under
physiological conditions in the regulation of systemic energy homeostasis and of the
exocrine system. We have recently found that KRAP is a molecule associated with inositol
1,4,5-trisphosphate receptor (IP3R) and is critical for the proper subcellular
localization of IP3R in the liver and the pancreas. However, the expression of
KRAP and its precise function in other tissues remain elusive. In this study, we aimed to
identify the KRAP-expressing cells in mouse stomach and kidneys and to examine the
relevance of KRAP expression in the regulation of IP3R localization in these
tissues. In the stomach, double immunohistochemical staining for KRAP and IP3R
demonstrated that KRAP was expressed along with the apical regions in the mucous cells and
the chief cells, and IP3R3 was dominantly co-localized with KRAP in these
cells. Furthermore, IP3R2 was also co-localized with IP3R3 in the
chief cells. It is of note that the proper localization of IP3R3 and
IP3R2 in the chief cells and of IP3R3 in the mucous cells were
significantly abrogated in KRAP-deficient mice. In the kidneys, KRAP was
expressed in both the apical and the basal regions of the proximal tubular cells.
Intriguingly, KRAP deficiency abrogated the localization of
IP3R1 in the proximal tubular cells. Finally, co-immunoprecipitation study in
the stomachs and the kidneys validated the physical association of KRAP with
IP3Rs. These findings demonstrate that KRAP physically associates with
IP3Rs and regulates the proper localization of IP3Rs in the mucous
cells and the chief cells of the stomach and in the proximal tubular cells of the
kidneys.
Collapse
Affiliation(s)
- Takahiro Fujimoto
- Department of Cell Biology, Faculty of Medicine, Fukuoka University, Fukuoka 814-0180, Japan
| | | |
Collapse
|
31
|
Matsuzaki H, Fujimoto T, Ota T, Ogawa M, Tsunoda T, Doi K, Hamabashiri M, Tanaka M, Shirasawa S. Tespa1 is a novel inositol 1,4,5-trisphosphate receptor binding protein in T and B lymphocytes. FEBS Open Bio 2012; 2:255-9. [PMID: 23650607 PMCID: PMC3642165 DOI: 10.1016/j.fob.2012.08.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2012] [Revised: 07/30/2012] [Accepted: 08/24/2012] [Indexed: 10/27/2022] Open
Abstract
Tespa1 has been recently reported to be a critical molecule in T-cell development, however, the precise molecular mechanisms of Tespa1 remain elusive. Here, we demonstrate that Tespa1 shows amino-acid sequence homology to KRAS-induced actin-interacting protein (KRAP), an inositol 1,4,5-trisphosphate receptor (IP3R) binding protein, and that Tespa1 physically associates with IP3R in T and B lymphocytes. Two-consecutive phenylalanine residues (Phe185/Phe186) in Tespa1, which are conserved between Tespa1 and KRAP, are indispensable for the association between Tespa1 and IP3R. These findings suggest that Tespa1 plays critical roles in the immune system through the regulation of the IP3R.
Collapse
Affiliation(s)
- Hiroshi Matsuzaki
- Department of Cell Biology, Faculty of Medicine, Fukuoka University, 7-45-1 Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Imbeault M, Giguère K, Ouellet M, Tremblay MJ. Exon level transcriptomic profiling of HIV-1-infected CD4(+) T cells reveals virus-induced genes and host environment favorable for viral replication. PLoS Pathog 2012; 8:e1002861. [PMID: 22876188 PMCID: PMC3410884 DOI: 10.1371/journal.ppat.1002861] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2012] [Accepted: 06/30/2012] [Indexed: 01/01/2023] Open
Abstract
HIV-1 is extremely specialized since, even amongst CD4+ T lymphocytes (its major natural reservoir in peripheral blood), the virus productively infects only a small proportion of cells under an activated state. As the percentage of HIV-1-infected cells is very low, most studies have so far failed to capture the precise transcriptomic profile at the whole-genome scale of cells highly susceptible to virus infection. Using Affymetrix Exon array technology and a reporter virus allowing the magnetic isolation of HIV-1-infected cells, we describe the host cell factors most favorable for virus establishment and replication along with an overview of virus-induced changes in host gene expression occurring exclusively in target cells productively infected with HIV-1. We also establish that within a population of activated CD4+ T cells, HIV-1 has no detectable effect on the transcriptome of uninfected bystander cells at early time points following infection. The data gathered in this study provides unique insights into the biology of HIV-1-infected CD4+ T cells and identifies genes thought to play a determinant role in the interplay between the virus and its host. Furthermore, it provides the first catalogue of alternative splicing events found in primary human CD4+ T cells productively infected with HIV-1. Some previous studies have monitored HIV-1-induced gene expression in various host cell targets and tissues but the discrimination between productively infected cells and uninfected bystander cells represents a technical challenge yet to be solved. Consequently, data interpretation has always been biased towards the transcriptional response of a majority of uninfected bystander cells that were exposed to soluble factors released by virus-infected cells. Following the design of a unique and innovative molecular tool to identify cells productively infected with HIV-1 and the description of an efficient magnetic beads-based technique to separate them from uninfected bystander cells, we undertake this challenge and perform the first comparative whole-genome transcriptomic and large-scale proteomic profiling of both HIV-1-infected and uninfected bystander CD4+ T cells. We demonstrate herein that HIV-1- infected and uninfected bystander cells display distinctive transcriptomic signatures which might permit to identify new susceptibility and resistance factors.
Collapse
Affiliation(s)
- Michaël Imbeault
- Centre de Recherche en Infectiologie, Centre Hospitalier Universitaire de Québec - CHUL, Faculté de Médecine, Université Laval, Québec City, Québec, Canada
| | - Katia Giguère
- Centre de Recherche en Infectiologie, Centre Hospitalier Universitaire de Québec - CHUL, Faculté de Médecine, Université Laval, Québec City, Québec, Canada
| | - Michel Ouellet
- Centre de Recherche en Infectiologie, Centre Hospitalier Universitaire de Québec - CHUL, Faculté de Médecine, Université Laval, Québec City, Québec, Canada
| | - Michel J. Tremblay
- Centre de Recherche en Infectiologie, Centre Hospitalier Universitaire de Québec - CHUL, Faculté de Médecine, Université Laval, Québec City, Québec, Canada
- Département de Microbiologie-Infectiologie et Immunologie, Faculté de Médecine, Université Laval, Québec City, Québec, Canada
- * E-mail:
| |
Collapse
|
33
|
|
34
|
Ion channels in autoimmune neurodegeneration. FEBS Lett 2011; 585:3836-42. [DOI: 10.1016/j.febslet.2011.03.065] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2011] [Revised: 03/26/2011] [Accepted: 03/28/2011] [Indexed: 11/23/2022]
|
35
|
Kwon J, Shatynski KE, Chen H, Morand S, de Deken X, Miot F, Leto TL, Williams MS. The nonphagocytic NADPH oxidase Duox1 mediates a positive feedback loop during T cell receptor signaling. Sci Signal 2010; 3:ra59. [PMID: 20682913 DOI: 10.1126/scisignal.2000976] [Citation(s) in RCA: 100] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Production of reactive oxygen species, often by NADPH (reduced form of nicotinamide adenine dinucleotide phosphate) oxidases, plays a role in the signaling responses of cells to many receptor stimuli. Here, we describe the function of the calcium-dependent, nonphagocytic NADPH oxidase Duox1 in primary human CD4(+) T cells and cultured T cell lines. Duox1 bound to inositol 1,4,5-trisphosphate receptor 1 and was required for early T cell receptor (TCR)-stimulated production of hydrogen peroxide (H(2)O(2)) through a pathway that was dependent on TCR-proximal kinases. Transient or stable knockdown of Duox1 inhibited TCR signaling, especially phosphorylation of tyrosine-319 of zeta chain-associated protein kinase of 70 kilodaltons (ZAP-70), store-operated entry of calcium ions (Ca(2+)), and activation of extracellular signal-regulated kinase. The production of cytokines was also inhibited by knockdown of Duox1. Duox1-mediated inactivation of Src homology 2 domain-containing protein tyrosine phosphatase 2 promoted the phosphorylation of ZAP-70 and its association with the Src family tyrosine kinase Lck and the CD3zeta chain of the TCR complex. Thus, we suggest that activation of Duox1, downstream of proximal TCR signals, generates H(2)O(2) that acts in a positive feedback loop to enhance and sustain further TCR signaling.
Collapse
Affiliation(s)
- Jaeyul Kwon
- Center for Vascular and Inflammatory Diseases, Department of Microbiology and Immunology, University of Maryland School of Medicine, 800 West Baltimore Street, Baltimore, MD 21201, USA.
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Hogan PG, Lewis RS, Rao A. Molecular basis of calcium signaling in lymphocytes: STIM and ORAI. Annu Rev Immunol 2010; 28:491-533. [PMID: 20307213 DOI: 10.1146/annurev.immunol.021908.132550] [Citation(s) in RCA: 601] [Impact Index Per Article: 42.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Ca(2+) entry into cells of the peripheral immune system occurs through highly Ca(2+)-selective channels known as CRAC (calcium release-activated calcium) channels. CRAC channels are a very well-characterized example of store-operated Ca(2+) channels, so designated because they open when the endoplasmic reticulum (ER) Ca(2+) store becomes depleted. Physiologically, Ca(2+) is released from the ER lumen into the cytoplasm when activated receptors couple to phospholipase C and trigger production of the second messenger inositol 1,4,5-trisphosphate (IP(3)). IP(3) binds to IP(3) receptors in the ER membrane and activates Ca(2+) release. The proteins STIM and ORAI were discovered through limited and genome-wide RNAi screens, respectively, performed in Drosophila cells and focused on identifying modulators of store-operated Ca(2+) entry. STIM1 and STIM2 sense the depletion of ER Ca(2+) stores, whereas ORAI1 is a pore subunit of the CRAC channel. In this review, we discuss selected aspects of Ca(2+) signaling in cells of the immune system, focusing on the roles of STIM and ORAI proteins in store-operated Ca(2+) entry.
Collapse
Affiliation(s)
- Patrick G Hogan
- Department of Pathology, Harvard Medical School, Immune Disease Institute, Children's Hospital Boston, Massachusetts 02115, USA.
| | | | | |
Collapse
|
37
|
Yehuda-Shnaidman E, Kalderon B, Azazmeh N, Bar-Tana J. Gating of the mitochondrial permeability transition pore by thyroid hormone. FASEB J 2009; 24:93-104. [PMID: 19723706 DOI: 10.1096/fj.09-133538] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The calorigenic-thermogenic activity of thyroid hormone (T3) has long been ascribed to uncoupling of mitochondrial oxidative phosphorylation. However, the mode of action of T3 in promoting mitochondrial proton leak is still unresolved. Mitochondrial uncoupling by T3 is reported here to be transduced in vivo in rats and in cultured Jurkat cells by gating of the mitochondrial permeability transition pore (PTP). T3-induced PTP gating is shown here to be abrogated in inositol 1,4,5-trisphosphate (IP(3)) receptor 1 (IP(3)R1)(-/-) cells, indicating that the endoplasmic reticulum IP(3)R1 may serve as upstream target for the mitochondrial activity of T3. IP(3)R1 gating by T3 is due to its increased expression and truncation into channel-only peptides, resulting in IP(3)-independent Ca(2+) efflux. Increased cytosolic Ca(2+) results in activation of protein phosphatase 2B, dephosphorylation and depletion of mitochondrial Bcl2 (S70), and increase in mitochondrial free Bax leading to low-conductance PTP gating. The T3 transduction pathway integrates genomic and nongenomic activities of T3 in regulating mitochondrial energetics and may offer novel targets for thyromimetics designed to modulate energy expenditure.
Collapse
Affiliation(s)
- Einav Yehuda-Shnaidman
- Department of Human Nutrition and Metabolism, Hebrew University Medical School, Jerusalem, Israel
| | | | | | | |
Collapse
|
38
|
Abstract
Calcium acts as a second messenger in many cell types, including lymphocytes. Resting lymphocytes maintain a low concentration of Ca2+. However, engagement of antigen receptors induces calcium influx from the extracellular space by several routes. A chief mechanism of Ca2+ entry in lymphocytes is through store-operated calcium (SOC) channels. The identification of two important molecular components of SOC channels, CRACM1 (the pore-forming subunit) and STIM1 (the sensor of stored calcium), has allowed genetic and molecular manipulation of the SOC entry pathway. In this review, we highlight advances in the understanding of Ca2+ signaling in lymphocytes with special emphasis on SOC entry. We also discuss outstanding questions and probable future directions of the field.
Collapse
Affiliation(s)
- Monika Vig
- Laboratory of Allergy and Immunology, Department of Pathology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts 02115, USA.
| | | |
Collapse
|
39
|
Nagaleekar VK, Diehl SA, Juncadella I, Charland C, Muthusamy N, Eaton S, Haynes L, Garrett-Sinha LA, Anguita J, Rincón M. IP3 receptor-mediated Ca2+ release in naive CD4 T cells dictates their cytokine program. THE JOURNAL OF IMMUNOLOGY 2009; 181:8315-22. [PMID: 19050248 DOI: 10.4049/jimmunol.181.12.8315] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
IP(3) (inositol 1,4,5-trisphosphate) receptors (IP(3)Rs) regulate the release of Ca(2+) from intracellular stores in response to IP(3). Little is known about regulation of the expression of IP(3)Rs and their role during the activation of CD4 T cells. In this study we show that mouse naive CD4 T cells express IP(3)R1, IP(3)R2, and IP(3)R3, but that gene expression of IP(3)R3 primarily is down-regulated upon activation due to loss of the Ets-1 transcription factor. Down-regulation of IP(3)R expression in activated CD4 T cells is associated with the failure of TCR ligation to trigger Ca(2+) release in these cells. We also show that down-regulation of specific IP(3)Rs in activated CD4 T cells correlates with the requirement of IP(3)R-mediated Ca(2+) release only for the induction of, but not for the maintenance of, IL-2 and IFN-gamma expression. Interestingly, while inhibition of IP(3)R function early during activation blocks IL-2 and IFN-gamma production, it promotes the production of IL-17 by CD4 T cells. Thus, IP(3)Rs play a key role in the activation and differentiation of CD4 T cells. The immunosuppressive effect of pharmacological blockers of these receptors may be complicated by promoting the development of inflammatory CD4 T cells.
Collapse
Affiliation(s)
- Viswas K Nagaleekar
- Department of Medicine, Immunobiology Program, University of Vermont, Burlington, VT 05405, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Thippeswamy G, Sheela ML, Salimath BP. Octacosanol isolated from Tinospora cordifolia downregulates VEGF gene expression by inhibiting nuclear translocation of NF-<kappa>B and its DNA binding activity. Eur J Pharmacol 2008; 588:141-50. [PMID: 18513715 DOI: 10.1016/j.ejphar.2008.04.027] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2007] [Revised: 03/26/2008] [Accepted: 04/02/2008] [Indexed: 10/22/2022]
Abstract
Octacosanol is a long-chain aliphatic alcohol, which is the main component of policosanol used as a normolipidemic agent. It is known that angiogenesis is involved in tumor growth and metastasis. The present study identified octacosanol isolated from the plant Tinospora cordifolia as a new antiangiogenic compound with inhibitory effects on in vivo angiogenesis assays. Our results showed that octacosanol (i) inhibits proliferation of endothelial cells and Ehrlich ascites tumor cells, (ii) inhibits neovascularization induced by angiogenic factors in chick chorioallantoic membrane and rat cornea in vivo angiogenesis assays, (iii) inhibits secretion of ascites fluid in the growing tumor cells in vivo. Concerning the mechanism of action, octacosanol inhibited secretion of vascular endothelial growth factor into ascites fluid by the tumor cells. At the molecular level octacosanol markedly inhibits activity of matrix metalloproteinases (MMPs) and translocation of transcription factor nuclear factor-<kappa>B to nucleus. The mechanism of inhibition of angiogenesis by octacosanol reflects on its effect on tumor angiogenesis and metastasis.
Collapse
Affiliation(s)
- G Thippeswamy
- Department of Studies in Applied Botany and Biotechnology, University of Mysore, Manasagangotri, Mysore-570006, India
| | | | | |
Collapse
|
41
|
Dadsetan S, Zakharova L, Molinski TF, Fomina AF. Store-operated Ca2+ influx causes Ca2+ release from the intracellular Ca2+ channels that is required for T cell activation. J Biol Chem 2008; 283:12512-9. [PMID: 18316371 DOI: 10.1074/jbc.m709330200] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The precise control of many T cell functions relies on cytosolic Ca(2+) dynamics that is shaped by the Ca(2+) release from the intracellular store and extracellular Ca(2+) influx. The Ca(2+) influx activated following T cell receptor (TCR)-mediated store depletion is considered to be a major mechanism for sustained elevation in cytosolic Ca(2+) concentration ([Ca(2+)](i)) necessary for T cell activation, whereas the role of intracellular Ca(2+) release channels is believed to be minor. We found, however, that in Jurkat T cells [Ca(2+)](i) elevation observed upon activation of the store-operated Ca(2+) entry (SOCE) by passive store depletion with cyclopiazonic acid, a reversible blocker of sarco-endoplasmic reticulum Ca(2+)-ATPase, inversely correlated with store refilling. This indicated that intracellular Ca(2+) release channels were activated in parallel with SOCE and contributed to global [Ca(2+)](i) elevation. Pretreating cells with (-)-xestospongin C (10 microM) or ryanodine (400 microM), the antagonists of inositol 1,4,5-trisphosphate receptor (IP3R) or ryanodine receptor (RyR), respectively, facilitated store refilling and significantly reduced [Ca(2+)](i) elevation evoked by the passive store depletion or TCR ligation. Although the Ca(2+) release from the IP3R can be activated by TCR stimulation, the Ca(2+) release from the RyR was not inducible via TCR engagement and was exclusively activated by the SOCE. We also established that inhibition of IP3R or RyR down-regulated T cell proliferation and T-cell growth factor interleukin 2 production. These studies revealed a new aspect of [Ca(2+)](i) signaling in T cells, that is SOCE-dependent Ca(2+) release via IP3R and/or RyR, and identified the IP3R and RyR as potential targets for manipulation of Ca(2+)-dependent functions of T lymphocytes.
Collapse
Affiliation(s)
- Sepehr Dadsetan
- Department of Physiology and Membrane Biology, University of California, Davis, Davis, California 95616, USA
| | | | | | | |
Collapse
|
42
|
Reut TM, Mattan L, Dafna T, Ruth KK, Ruth S. The role of Src family kinases in egg activation. Dev Biol 2007; 312:77-89. [PMID: 17949706 DOI: 10.1016/j.ydbio.2007.09.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2007] [Revised: 08/14/2007] [Accepted: 09/05/2007] [Indexed: 01/06/2023]
Abstract
The Src family kinases (SFKs) are believed to mediate some of the early events of egg activation at fertilization--intracellular Ca2+ increase and resumption of the second meiotic division (RMII). SFKs are both necessary and sufficient for triggering intracellular Ca2+ increase in eggs of sea urchin, sea star, Xenopus etc, but their role in mammalian eggs is not entirely determined. In this study we examined the involvement of SFKs in the events leading to Ca2+ increase in rat eggs and demonstrated their involvement in RMII. Microinjecting mRNAs of active forms of Fyn or c-Yes but not of c-Src, into ovulated eggs, triggered RMII without evoking Ca2+ increase. A specific SFKs inhibitor (SU6656) or dominant-negative (DN) forms of Fyn or c-Yes were unable to block Ca2+ oscillations rather, modulated them, in fertilized eggs or in parthenogenetically activated eggs. Moreover, inhibiting SFKs activity blocked RMII and decreased the level of cyclin B1 degradation. Our results imply participation of SFKs in the signal transduction pathway leading to egg activation, but not in the one leading to Ca2+ increase. We propose that SFKs act downstream to Ca2+ increase at the level of M-phase promoting factor (MPF).
Collapse
Affiliation(s)
- Tomashov-Matar Reut
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel-Aviv University, Ramat-Aviv 69978, Tel-Aviv, Israel
| | | | | | | | | |
Collapse
|
43
|
Zhou Z, Yin J, Dou Z, Tang J, Zhang C, Cao Y. The calponin homology domain of Vav1 associates with calmodulin and is prerequisite to T cell antigen receptor-induced calcium release in Jurkat T lymphocytes. J Biol Chem 2007; 282:23737-44. [PMID: 17550897 DOI: 10.1074/jbc.m702975200] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Vav1 is a guanine nucleotide exchange factor that is expressed specifically in hematopoietic cells and plays important roles in T cell development and activation. Vav1 consists of multiple structural domains so as to facilitate both its guanine nucleotide exchange activity and scaffold function following T cell antigen receptor (TCR) engagement. Previous studies demonstrated that the calponin homology (CH) domain of Vav1 is required for TCR-stimulated calcium mobilization and thus downstream activation of nuclear factor of activated T cells. However, it remained obscure how Vav1 functions in regulating calcium flux. In an effort to explore molecules interacting with Vav1, we found that calmodulin bound to Vav1 in a calcium-dependent and TCR activation-independent manner. The binding site was mapped to the CH domain of Vav1. Reconstitution of vav1-null Jurkat T cells (J.Vav1) with CH-deleted Vav1 exhibited a severe deficiency in calcium release to the same extent as that of Jurkat cells treated with the calmodulin inhibitor or J.Vav1 cells. The defect persisted even when phospholipase-Cgamma1 was fully activated, indicating a prerequisite role of Vav1 CH domain in calcium signaling. The results suggest that Vav1 and calmodulin function cooperatively to potentiate TCR-induced calcium release. This study unveiled a mechanism by which the Vav1 CH domain is involved in calcium signaling and provides insight into our understanding of the role of Vav1 in T cell activation.
Collapse
Affiliation(s)
- Zhuo Zhou
- Department of Biochemistry and Molecular Biology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | | | | | | | | | | |
Collapse
|
44
|
Afroze T, Sadi AM, Momen MA, Gu S, Heximer S, Husain M. c-Myb-dependent inositol 1,4,5-trisphosphate receptor type-1 expression in vascular smooth muscle cells. Arterioscler Thromb Vasc Biol 2007; 27:1305-11. [PMID: 17363689 DOI: 10.1161/atvbaha.107.142059] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE The IP3 receptor-1 (IP3R1) mediates Ca2+ signals critical to vascular smooth muscle cell (VSMC) proliferation. The cell cycle-associated transcription factor c-Myb increases Ca2+ at the G1/S transition. Here we show the mechanism through which c-Myb regulates expression of IP3R1. METHODS & RESULTS Ribonuclease protection confirmed transcriptional start (TS), and qRT-PCR revealed a 6-fold increase in IP3R1 mRNA as immortalized VSMC progress from G0 to G1/S. A c-Myb neutralizing antibody decreased IP3R1 mRNA expression 3-fold, and abolished the 3.4-fold increase in IP3R1 protein observed at G1/S. Primary aortic VSMCs in culture and proliferating carotid VSMCs in vivo showed similar regulation of IP3R1 mRNA and protein. Sequence analysis of a 3.1-Kb mouse IP3R1 promoter revealed 17 putative c-Myb binding sites. Reporter assays demonstrated a 2-fold increase in promoter activity in G1/S- versus G0-synchronized VSMCs, which was abolished by functional c-Myb knockdown or deletion of promoter sequences upstream and downstream of TS. Point mutations in Myb sites-13 or -15 significantly blunted G1/S-specific promoter induction in both immortalized and primary VSMCs. Gel shift and ChIP confirmed binding of c-Myb to sites-13 and -15 in G1/S stage VSMCs. CONCLUSION c-Myb regulates cell cycle-associated IP3R1 transcription in VSMCs via specific highly conserved Myb-binding sites in the IP3R1 promoter.
Collapse
MESH Headings
- Animals
- Calcium/metabolism
- Calcium Channels/genetics
- Calcium Channels/metabolism
- Carotid Arteries/surgery
- Carotid Artery Diseases/genetics
- Carotid Artery Diseases/metabolism
- Carotid Artery Diseases/physiopathology
- Cell Cycle/physiology
- Cell Line
- Cell Proliferation
- Chromatin Immunoprecipitation
- Conserved Sequence
- DNA/metabolism
- Disease Models, Animal
- Electrophoretic Mobility Shift Assay
- Genes, Reporter
- Inositol 1,4,5-Trisphosphate Receptors
- Luciferases
- Membrane Glycoproteins/genetics
- Membrane Glycoproteins/metabolism
- Mice
- Mice, Inbred C57BL
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/metabolism
- Mutation
- Myocytes, Smooth Muscle/metabolism
- Promoter Regions, Genetic
- Proto-Oncogene Proteins c-myb/metabolism
- RNA, Messenger/metabolism
- Receptors, Cytoplasmic and Nuclear/genetics
- Receptors, Cytoplasmic and Nuclear/metabolism
- Sequence Analysis, DNA
- Transcription, Genetic
- Transfection
- Up-Regulation
Collapse
Affiliation(s)
- Talat Afroze
- Division of Cell and Molecular Biology, Toronto General Hospital Research Institute, Toronto, Ontario, Canada
| | | | | | | | | | | |
Collapse
|
45
|
Chakrabarti R, Chakrabarti R. Calcium signaling in non-excitable cells: Ca2+ release and influx are independent events linked to two plasma membrane Ca2+ entry channels. J Cell Biochem 2007; 99:1503-16. [PMID: 17031847 DOI: 10.1002/jcb.21102] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
The regulatory mechanism of Ca2+ influx into the cytosol from the extracellular space in non-excitable cells is not clear. The "capacitative calcium entry" (CCE) hypothesis suggested that Ca2+ influx is triggered by the IP(3)-mediated emptying of the intracellular Ca2+ stores. However, there is no clear evidence for CCE and its mechanism remains elusive. In the present work, we have provided the reported evidences to show that inhibition of IP(3)-dependent Ca2+ release does not affect Ca2+ influx, and the experimental protocols used to demonstrate CCE can stimulate Ca2+ influx by means other than emptying of the Ca2+ stores. In addition, we have presented the reports showing that IP(3)-mediated Ca2+ release is linked to a Ca2+ entry from the extracellular space, which does not increase cytosolic [Ca2+] prior to Ca2+ release. Based on these and other reports, we have provided a model of Ca2+ signaling in non-excitable cells, in which IP(3)-mediated emptying of the intracellular Ca2+ store triggers entry of Ca2+ directly into the store, through a plasma membrane TRPC channel. Thus, emptying and direct refilling of the Ca2+ stores are repeated in the presence of IP(3), giving rise to the transient phase of oscillatory Ca2+ release. Direct Ca2+ entry into the store is regulated by its filling status in a negative and positive manner through a Ca2+ -binding protein and Stim1/Orai complex, respectively. The sustained phase of Ca2+ influx is triggered by diacylglycerol (DAG) through the activation of another TRPC channel, independent of Ca2+ release. The plasma membrane IP(3) receptor (IP(3)R) plays an essential role in Ca2+ influx, by interacting with the DAG-activated TRPC, without the requirement of binding to IP(3).
Collapse
Affiliation(s)
- Ranjana Chakrabarti
- Department of Laboratory Medicine, St. Michael's Hospital, 30 Bond Street, Toronto, Ontario, Canada M6N 4C5
| | | |
Collapse
|
46
|
Manicassamy S, Sadim M, Ye RD, Sun Z. Differential roles of PKC-theta in the regulation of intracellular calcium concentration in primary T cells. J Mol Biol 2005; 355:347-59. [PMID: 16309697 DOI: 10.1016/j.jmb.2005.10.043] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2005] [Revised: 10/10/2005] [Accepted: 10/17/2005] [Indexed: 12/13/2022]
Abstract
Activation of T lymphocytes requires protein kinase C theta (PKC-theta) and an appropriately elevated free intracellular Ca2+ concentration ([Ca2+]i). Here, we show that phorbol 12 myristate 13-acetate (PMA) inhibited Ca2+ influx in wild-type but not PKC-theta-/- T cells, suggesting that PKC-theta plays a role in PMA-mediated inhibition of Ca2+ influx. In contrast, T cell receptor (TCR) crosslinking in the same PKC-theta-/- T cells did result in significantly decreased [Ca2+]i compared to wild-type T cells, suggesting a positive role for PKC-theta in TCR-mediated Ca2+ mobilization. In PKC-theta-/- mice, peripheral mature T cells, but not developing thymocytes, displayed significantly decreased TCR-induced Ca2+ influx and nuclear factor of activated T cells (NFAT) translocation upon sub-optimal TCR crosslinking. The decreased intracellular free Ca2+ was due to changes in Ca2+ influx but not efflux, as observed in extracellular and intracellular Ca2+ mobilization studies. However, these differences in Ca2+ influx and nuclear factor of activated T cells (NFAT) translocation disappeared with increasing intensity of TCR crosslinking. The enhancing effect of PKC-theta on Ca2+ influx is not only dependent on the strength of TCR crosslinking but also on the developmental stage of T cells. The underlying mechanism involved phospholipase Cgamma1 activation and inositol triphosphate production. Furthermore, knockdown of endogenous PKC-theta expression in Jurkat cells resulted in significant inhibition of TCR-induced activation of NFAT, as evidenced from NFAT reporter studies. Forced expression of a constitutively active form of calcineurin in PKC-theta-/- Jurkat cells could readily overcome the above inhibition. Thus, PKC-theta can both positively and negatively regulate the Ca2+ influx that is critical for NFAT activity.
Collapse
Affiliation(s)
- Santhakumar Manicassamy
- Department of Microbiology and Immunology, College of Medicine, University of Illinois, Chicago, IL 60612, USA
| | | | | | | |
Collapse
|
47
|
Woodcock EA, Matkovich SJ. Ins(1,4,5)P3 receptors and inositol phosphates in the heart-evolutionary artefacts or active signal transducers? Pharmacol Ther 2005; 107:240-51. [PMID: 15908009 DOI: 10.1016/j.pharmthera.2005.04.002] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
The generation of the second messenger inositol 1,4,5-trisphosphate (Ins(1,4,5)P(3)) and its associated release of Ca(2+) from internal stores is a highly conserved module in intracellular signaling from Drosophila to mammals. Many cell types, often nonexcitable cells, depend on this pathway to couple external signals to intracellular Ca(2+) release. However, despite the presence of the requisite Ins(1,4,5)P(3) signaling machinery, excitable cells such as cardiac myocytes employ a robust alternate system of intracellular Ca(2+) release, namely, a coupled system of Ca(2+) influx, followed by Ca(2+) release via the IP(3)R-related ryanodine receptors. In these systems, Ins(1,4,5)P(3) signaling pathways appear to be largely dormant. In this review, we consider the general features of inositol phosphate (InsP) responses in cardiac myocytes and the molecules mediating these responses. The spatial localization of Ins(1,4,5)P(3) generation and Ins(1,4,5)P(3) receptor (IP(3)Rs) is likely of key importance, and we examine the state of knowledge in atrial, ventricular, and Purkinje myocytes. Several studies have implicated Ins(1,4,5)P(3) generation in both arrhythmogenic and hypertrophic responses, and possible mechanisms involving Ins(1,4,5)P(3) are discussed. While Ins(1,4,5)P(3) is unlikely to be a key player in cardiac excitation-contraction (EC) coupling, its potential role in an alternate Ca(2+) release system to signal changes in gene transcription warrants further investigation. Such studies will help to determine whether cardiac Ins(1,4,5)P(3) generation represents a vestigial pathway or plays an active role in cardiac signaling.
Collapse
Affiliation(s)
- Elizabeth A Woodcock
- Cellular Biochemistry Laboratory, Baker Heart Research Institute, Commercial Road, Melbourne, Australia.
| | | |
Collapse
|
48
|
Soghoian D, Jayaraman V, Silane M, Berenstein A, Jayaraman T. Inositol 1,4,5-trisphosphate receptor phosphorylation in breast cancer. Tumour Biol 2005; 26:207-12. [PMID: 16006774 DOI: 10.1159/000086954] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2005] [Indexed: 11/19/2022] Open
Abstract
The aim of this study was to establish the type(s) of inositol 1,4,5-trisphosphate receptors (IP3Rs) in T47D breast cancer cells that regulate intracellular calcium (Ca2+) and whether they interact with cyclin (Cy), an important regulator of cyclin-dependent kinases (cdk), during cell cycle progression. Immunoblotting, immunoprecipitation, and pull-down assays were used to identify IP3R expression and interaction with Cy. The relative IP3R3 expression, as compared to IP3R1, was higher in these cells. Pull-down analysis showed that IP3R3 interacted with both CyA and CyB. The interaction with Cys and the phosphorylation of IP3Rs by Cy/cdk complexes provide a novel mechanism of regulating intracellular Ca2+ release and Ca2+-dependent signaling events in breast cancer.
Collapse
Affiliation(s)
- Damien Soghoian
- Vascular Biology Laboratory, Department of Neurosurgery, St. Luke's Roosevelt Hospital Center, New York, N.Y. 10025, USA
| | | | | | | | | |
Collapse
|
49
|
Pawlak J, Singh J, Lea RW, Skwarlo-Sonta K. Effect of melatonin on phagocytic activity and intracellular free calcium concentration in testicular macrophages from normal and streptozotocin-induced diabetic rats. Mol Cell Biochem 2005; 275:207-13. [PMID: 16335800 DOI: 10.1007/s11010-005-1995-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
This study examined the effect of melatonin (MLT) on in vitro phagocytosis of testicular macrophages taken from control and streptozotocin (STZ)-induced diabetic rats and the possible mechanism of its action. The phagocytic activity was measured as a number of latex beads ingested by 100 macrophages (PI, phagocytic index) in consecutive time points of the incubation. Changes in intracellular free calcium level [Ca2+]i in isolated macrophages in vitro were measured with the use of ratio-image fluorescence microscopy (fluorescent dye: Fura2/AM). Phagocytic index in macrophages isolated from healthy rats was 20% higher than in those from diabetic animals. Melatonin in physiological concentration (10(-7) M) significantly (p < 0.05) increased the PI in testicular macrophages from control animals (PI = 68 +/- 5 with MLT compared to PI = 46 +/- 7 without MLT) while no such effect was observed in the cells from diabetic rats (PI = 36 +/- 23 with MLT compared to PI = 31 +/- 11 without MLT). Basal [Ca2+]i was significantly (p < 0.01) higher in macrophages from diabetic rats compared to control. Stimulation of both control and diabetic testicular macrophages with 10(-7) M MLT resulted in a significant (p < 0.05) increase in [Ca2+]i in cells incubated in 2.5 mM calcium solution while no such response was observed in calcium-free Tyrode solution. However, MLT evoked [Ca2+]i response in macrophages isolated from diabetic animals was much lower than in macrophages isolated from age-matched controls and the time needed for maximal response was much longer. Lack of response in calcium-free solution suggests that extracellular calcium may be necessary to trigger MLT response and in its progression.
Collapse
Affiliation(s)
- Joanna Pawlak
- Department of Vertebrate Physiology, Faculty of Biology, Warsaw University, ul. Miecznikowa 1, 02-096 Warsaw, Poland.
| | | | | | | |
Collapse
|
50
|
Camandola S, Cutler RG, Gary DS, Milhavet O, Mattson MP. Suppression of calcium release from inositol 1,4,5-trisphosphate-sensitive stores mediates the anti-apoptotic function of nuclear factor-kappaB. J Biol Chem 2005; 280:22287-96. [PMID: 15814613 DOI: 10.1074/jbc.m410923200] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
The activation of the transcription factor nuclear factor-kappaB (NF-kappaB) by growth factors, cytokines, and cellular stress can prevent apoptosis, but the underlying mechanism is unknown. Here we provide evidence for an action of NF-kappaB on calcium signaling that accounts for its anti-apoptotic function. Embryonic fibroblasts lacking the transactivating subunit of NF-kappaB RelA (p65) exhibit enhanced inositol 1,4,5-trisphosphate (IP(3)) receptor-mediated calcium release and increased sensitivity to apoptosis, which are restored upon re-expression of RelA. The size of the endoplasmic reticulum (ER) calcium pool and the number of IP(3) receptors per cell are decreased in response to stimuli that activate NF-kappaB and are increased when NF-kappaB activity is suppressed. The selective antagonism of IP(3) receptors blocks apoptosis in RelA-deficient cells, whereas activation of NF-kappaB in normal cells leads to decreased levels of the type 1 IP(3) receptor and decreased calcium release. Overexpression of Bcl-2 normalizes ER calcium homeostasis and prevents calcium-mediated apoptosis in RelA-deficient cells. These findings establish an ER calcium channel as a pivotal target for NF-kappaB-mediated cell survival signaling.
Collapse
MESH Headings
- Adenosine Triphosphate/metabolism
- Animals
- Apoptosis
- Blotting, Western
- Calcium/metabolism
- Calcium Channels/metabolism
- Calcium-Transporting ATPases/metabolism
- Cell Survival
- Ceramides/pharmacology
- Cytosol/metabolism
- DNA/metabolism
- Endoplasmic Reticulum/metabolism
- Immunoblotting
- Immunohistochemistry
- Inositol 1,4,5-Trisphosphate/metabolism
- Inositol 1,4,5-Trisphosphate Receptors
- Lipid Metabolism
- Mice
- Mice, Transgenic
- Microscopy, Fluorescence
- Microsomes/metabolism
- NF-kappa B/chemistry
- NF-kappa B/metabolism
- Oligonucleotides, Antisense/chemistry
- Oxidative Stress
- RNA, Messenger/metabolism
- Receptors, Cytoplasmic and Nuclear/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Sarcoplasmic Reticulum Calcium-Transporting ATPases
- Signal Transduction
- Time Factors
- Transcription Factor RelA
- Transcriptional Activation
Collapse
Affiliation(s)
- Simonetta Camandola
- Laboratory of Neurosciences, National Institute on Aging/NIH, Gerontology Research Center, 5600 Nathan Shock Drive, Baltimore, MD 21224, USA.
| | | | | | | | | |
Collapse
|