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
|
Wang Q, Yin X, Huang X, Zhang L, Lu H. Impact of mitochondrial dysfunction on the antitumor effects of immune cells. Front Immunol 2024; 15:1428596. [PMID: 39464876 PMCID: PMC11502362 DOI: 10.3389/fimmu.2024.1428596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 09/24/2024] [Indexed: 10/29/2024] Open
Abstract
Mitochondrial dysfunction, a hallmark of immune cell failure, affects the antitumor effects of immune cells through metabolic reprogramming, fission, fusion, biogenesis, and immune checkpoint signal transduction of mitochondria. According to researchers, restoring damaged mitochondrial function can enhance the efficacy of immune cells. Nevertheless, the mechanism of mitochondrial dysfunction in immune cells in patients with cancer is unclear. In this review, we recapitulate the impact of mitochondrial dysfunction on the antitumor effects of T cells, natural killer cells, dendritic cells, and tumor-associated macrophage and propose that targeting mitochondria can provide new strategies for antitumor therapy.
Collapse
Affiliation(s)
- Quan Wang
- Department of Radiation Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiangzhi Yin
- Department of Orthopaedics, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiaotong Huang
- Department of Radiation Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Lu Zhang
- Department of Radiation Oncology, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Haijun Lu
- Department of Radiation Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
3
|
Yazicioglu YF, Mitchell RJ, Clarke AJ. Mitochondrial control of lymphocyte homeostasis. Semin Cell Dev Biol 2024; 161-162:42-53. [PMID: 38608498 DOI: 10.1016/j.semcdb.2024.03.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 03/11/2024] [Accepted: 03/26/2024] [Indexed: 04/14/2024]
Abstract
Mitochondria play a multitude of essential roles within mammalian cells, and understanding how they control immunity is an emerging area of study. Lymphocytes, as integral cellular components of the adaptive immune system, rely on mitochondria for their function, and mitochondria can dynamically instruct their differentiation and activation by undergoing rapid and profound remodelling. Energy homeostasis and ATP production are often considered the primary functions of mitochondria in immune cells; however, their importance extends across a spectrum of other molecular processes, including regulation of redox balance, signalling pathways, and biosynthesis. In this review, we explore the dynamic landscape of mitochondrial homeostasis in T and B cells, and discuss how mitochondrial disorders compromise adaptive immunity.
Collapse
|
4
|
Moraly J, Kondo T, Benzaoui M, DuSold J, Talluri S, Pouzolles MC, Chien C, Dardalhon V, Taylor N. Metabolic dialogues: regulators of chimeric antigen receptor T cell function in the tumor microenvironment. Mol Oncol 2024; 18:1695-1718. [PMID: 38922759 PMCID: PMC11223614 DOI: 10.1002/1878-0261.13691] [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: 12/05/2023] [Revised: 02/23/2024] [Accepted: 06/11/2024] [Indexed: 06/28/2024] Open
Abstract
Tumor-infiltrating lymphocytes (TILs) and chimeric antigen receptor (CAR) T cells have demonstrated remarkable success in the treatment of relapsed/refractory melanoma and hematological malignancies, respectively. These treatments have marked a pivotal shift in cancer management. However, as "living drugs," their effectiveness is dependent on their ability to proliferate and persist in patients. Recent studies indicate that the mechanisms regulating these crucial functions, as well as the T cell's differentiation state, are conditioned by metabolic shifts and the distinct utilization of metabolic pathways. These metabolic shifts, conditioned by nutrient availability as well as cell surface expression of metabolite transporters, are coupled to signaling pathways and the epigenetic landscape of the cell, modulating transcriptional, translational, and post-translational profiles. In this review, we discuss the processes underlying the metabolic remodeling of activated T cells, the impact of a tumor metabolic environment on T cell function, and potential metabolic-based strategies to enhance T cell immunotherapy.
Collapse
Affiliation(s)
- Josquin Moraly
- Pediatric Oncology Branch, National Cancer InstituteNational Institutes of HealthBethesdaMDUSA
- Université Sorbonne Paris CitéParisFrance
| | - Taisuke Kondo
- Pediatric Oncology Branch, National Cancer InstituteNational Institutes of HealthBethesdaMDUSA
| | - Mehdi Benzaoui
- Pediatric Oncology Branch, National Cancer InstituteNational Institutes of HealthBethesdaMDUSA
- Université de Montpellier, Institut de Génétique Moléculaire de Montpellier, CNRSMontpellierFrance
| | - Justyn DuSold
- Pediatric Oncology Branch, National Cancer InstituteNational Institutes of HealthBethesdaMDUSA
| | - Sohan Talluri
- Pediatric Oncology Branch, National Cancer InstituteNational Institutes of HealthBethesdaMDUSA
| | - Marie C. Pouzolles
- Pediatric Oncology Branch, National Cancer InstituteNational Institutes of HealthBethesdaMDUSA
| | - Christopher Chien
- Pediatric Oncology Branch, National Cancer InstituteNational Institutes of HealthBethesdaMDUSA
| | - Valérie Dardalhon
- Université de Montpellier, Institut de Génétique Moléculaire de Montpellier, CNRSMontpellierFrance
| | - Naomi Taylor
- Pediatric Oncology Branch, National Cancer InstituteNational Institutes of HealthBethesdaMDUSA
- Université de Montpellier, Institut de Génétique Moléculaire de Montpellier, CNRSMontpellierFrance
| |
Collapse
|
5
|
Gao Y, Liu S, Huang Y, Li F, Zhang Y. Regulation of anti-tumor immunity by metal ion in the tumor microenvironment. Front Immunol 2024; 15:1379365. [PMID: 38915413 PMCID: PMC11194341 DOI: 10.3389/fimmu.2024.1379365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 05/29/2024] [Indexed: 06/26/2024] Open
Abstract
Metal ions play an essential role in regulating the functions of immune cells by transmitting intracellular and extracellular signals in tumor microenvironment (TME). Among these immune cells, we focused on the impact of metal ions on T cells because they can recognize and kill cancer cells and play an important role in immune-based cancer treatment. Metal ions are often used in nanomedicines for tumor immunotherapy. In this review, we discuss seven metal ions related to anti-tumor immunity, elucidate their roles in immunotherapy, and provide novel insights into tumor immunotherapy and clinical applications.
Collapse
Affiliation(s)
- Yaoxin Gao
- Biotherapy Center & Cancer Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Shasha Liu
- Biotherapy Center & Cancer Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yifan Huang
- Biotherapy Center & Cancer Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Feng Li
- Biotherapy Center & Cancer Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yi Zhang
- Biotherapy Center & Cancer Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- School of Life Sciences, Zhengzhou University, Zhengzhou, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, China
- School of Public Health, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
6
|
Diercks BP. The importance of Ca 2+ microdomains for the adaptive immune response. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119710. [PMID: 38522726 DOI: 10.1016/j.bbamcr.2024.119710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 03/12/2024] [Accepted: 03/17/2024] [Indexed: 03/26/2024]
Abstract
Calcium signaling stands out as the most widespread and universally used signaling system and is of utmost importance for immunity. Controlled elevations in cytosolic and organellar Ca2+ concentrations in T cells control complex and essential effector functions including proliferation, differentiation, cytokine secretion, and cytotoxicity, among others. Additionally, disruptions in Ca2+ regulation in T cells contribute to diverse autoimmune, inflammatory, and immunodeficiency conditions. Among the initial intracellular signals, which occurring even before T cell receptor (TCR) stimulation are highly localized, spatially and temporally restricted so-called Ca2+ microdomains, caused by adhesion to extracellular matrix proteins (ECM proteins). The Ca2+ microdomains present both before and within the initial seconds following TCR stimulation are likely to play a crucial role in fine-tuning the downstream activity of T cell activation and thus, shaping an adaptive immune response. In this review, the emphasis is on the recent advances of adhesion-dependent Ca2+ microdomains (ADCM) in the absence of TCR stimulation, initial Ca2+ microdomains evoked by TCR stimulation (TDCM), the downstream signaling processes as well as possible therapeutic targets for interventions.
Collapse
Affiliation(s)
- Björn-Philipp Diercks
- The Calcium Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany.
| |
Collapse
|
7
|
Li CJ, Tzeng YDT, Hsiao JH, Tseng LM, Hsu TS, Chu PY. Spatial and single-cell explorations uncover prognostic significance and immunological functions of mitochondrial calcium uniporter in breast cancer. Cancer Cell Int 2024; 24:140. [PMID: 38632642 PMCID: PMC11022417 DOI: 10.1186/s12935-024-03327-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 04/12/2024] [Indexed: 04/19/2024] Open
Abstract
The mitochondrial calcium uniporter (MCU) is a transmembrane protein facilitating the entry of calcium ions into mitochondria from the cell cytosol. Maintaining calcium balance is crucial for enhancing cellular energy supply and regulating cell death. The interplay of calcium balance through MCU and the sodium-calcium exchanger is known, but its regulation in the breast cancer tumor microenvironment remains elusive. Further investigations are warranted to explore MCU's potential in BRCA clinical pathology, tumor immune microenvironment, and precision oncology. Our study, employing a multi-omics approach, identifies MCU as an independent diagnostic biomarker for breast cancer (BRCA), correlated with advanced clinical status and poor overall survival. Utilizing public datasets from GEO and TCGA, we discern differentially expressed genes in BRCA and examine their associations with immune gene expression, overall survival, tumor stage, gene mutation status, and infiltrating immune cells. Spatial transcriptomics is employed to investigate MCU gene expression in various regions of BRCA, while spatial transcriptomics and single-cell RNA-sequencing methods explore the correlation between MCUs and immune cells. Our findings are validated through the analysis of 59 BRCA patient samples, utilizing immunohistochemistry and bioinformatics to examine the relationship between MCU expression, clinicopathological features, and prognosis. The study uncovers the expression of key gene regulators in BRCA associated with genetic variations, deletions, and the tumor microenvironment. Mutations in these regulators positively correlate with different immune cells in six immune datasets, playing a pivotal role in immune cell infiltration in BRCA. Notably, high MCU performance is linked to CD8 + T cells infiltration in BRCA. Furthermore, pharmacogenomic analysis of BRCA cell lines indicates that MCU inactivation is associated with increased sensitivity to specific small molecule drugs. Our findings suggest that MCU alterations may be linked to BRCA progression, unveiling new diagnostic and prognostic implications for MCU in BRCA. The study underscores MCU's role in the tumor immune microenvironment and cell cycle progression, positioning it as a potential tool for BRCA precision medicine and drug screening.
Collapse
Affiliation(s)
- Chia-Jung Li
- Department of Obstetrics and Gynecology, Kaohsiung Veterans General Hospital, Kaohsiung, 813, Taiwan
- Institute of BioPharmaceutical Sciences, National Sun Yat-sen University, Kaohsiung, 804, Taiwan
| | - Yen-Dun Tony Tzeng
- Department of Surgery, Kaohsiung Veterans General Hospital, Kaohsiung, 813, Taiwan
- Institute of Biomedical Sciences, National Sun Yat-sen University, Kaohsiung, 804, Taiwan
| | - Jui-Hu Hsiao
- Department of Surgery, Kaohsiung Municipal Minsheng Hospital, Kaohsiung, 802, Taiwan
| | - Ling-Ming Tseng
- School of Medicine, National Yang-Ming University, Taipei, 112, Taiwan
- Comprehensive Breast Health Center, Taipei Veterans General Hospital, Taipei, 112, Taiwan
| | - Tzu-Sheng Hsu
- Institute of Molecular and Cellular Biology, College of Life Sciences and Medicine, National Tsing Hua University, Hsinchu, 300, Taiwan
| | - Pei-Yi Chu
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung, 402, Taiwan.
- School of Medicine, College of Medicine, Fu Jen Catholic University, New Taipei City, 242, Taiwan.
- Department of Pathology, Show Chwan Memorial Hospital, Changhua, 500, Taiwan.
- National Institute of Cancer Research, National Health Research Institutes, Tainan, 704, Taiwan.
| |
Collapse
|
8
|
Cong J, Liu P, Han Z, Ying W, Li C, Yang Y, Wang S, Yang J, Cao F, Shen J, Zeng Y, Bai Y, Zhou C, Ye L, Zhou R, Guo C, Cang C, Kasper DL, Song X, Dai L, Sun L, Pan W, Zhu S. Bile acids modified by the intestinal microbiota promote colorectal cancer growth by suppressing CD8 + T cell effector functions. Immunity 2024; 57:876-889.e11. [PMID: 38479384 DOI: 10.1016/j.immuni.2024.02.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 12/10/2023] [Accepted: 02/15/2024] [Indexed: 04/12/2024]
Abstract
Concentrations of the secondary bile acid, deoxycholic acid (DCA), are aberrantly elevated in colorectal cancer (CRC) patients, but the consequences remain poorly understood. Here, we screened a library of gut microbiota-derived metabolites and identified DCA as a negative regulator for CD8+ T cell effector function. Mechanistically, DCA suppressed CD8+ T cell responses by targeting plasma membrane Ca2+ ATPase (PMCA) to inhibit Ca2+-nuclear factor of activated T cells (NFAT)2 signaling. In CRC patients, CD8+ T cell effector function negatively correlated with both DCA concentration and expression of a bacterial DCA biosynthetic gene. Bacteria harboring DCA biosynthetic genes suppressed CD8+ T cells effector function and promoted tumor growth in mice. This effect was abolished by disrupting bile acid metabolism via bile acid chelation, genetic ablation of bacterial DCA biosynthetic pathway, or specific bacteriophage. Our study demonstrated causation between microbial DCA metabolism and anti-tumor CD8+ T cell response in CRC, suggesting potential directions for anti-tumor therapy.
Collapse
Affiliation(s)
- Jingjing Cong
- Key Laboratory of Immune Response and Immunotherapy, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China; Department of Digestive Disease, The First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China; School of Pharmacy, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei 230032, China
| | - Pianpian Liu
- Key Laboratory of Immune Response and Immunotherapy, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China; Department of Digestive Disease, The First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China
| | - Zili Han
- Key Laboratory of Immune Response and Immunotherapy, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China; Department of Digestive Disease, The First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China
| | - Wei Ying
- Key Laboratory of Immune Response and Immunotherapy, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Chaoliang Li
- Key Laboratory of Immune Response and Immunotherapy, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China; Department of Digestive Disease, The First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China
| | - Yifei Yang
- Key Laboratory of Immune Response and Immunotherapy, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China; School of Data Science, University of Science and Technology of China, Hefei 230027, China
| | - Shuling Wang
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Jianbo Yang
- Key Laboratory of Immune Response and Immunotherapy, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China; Department of Digestive Disease, The First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China
| | - Fei Cao
- Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Juntao Shen
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Yu Zeng
- Key Laboratory of Immune Response and Immunotherapy, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Yu Bai
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Congzhao Zhou
- Key Laboratory of Immune Response and Immunotherapy, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Lilin Ye
- Institute of Immunology, Third Military Medical University, Chongqing 400038, China
| | - Rongbin Zhou
- Key Laboratory of Immune Response and Immunotherapy, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Chunjun Guo
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY 10021, USA
| | - Chunlei Cang
- Key Laboratory of Immune Response and Immunotherapy, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Dennis L Kasper
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Xinyang Song
- Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Lei Dai
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Linfeng Sun
- Key Laboratory of Immune Response and Immunotherapy, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Wen Pan
- Key Laboratory of Immune Response and Immunotherapy, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China; Department of Digestive Disease, The First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China.
| | - Shu Zhu
- Key Laboratory of Immune Response and Immunotherapy, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China; Department of Digestive Disease, The First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China; School of Data Science, University of Science and Technology of China, Hefei 230027, China.
| |
Collapse
|
9
|
Bacsa B, Hopl V, Derler I. Synthetic Biology Meets Ca 2+ Release-Activated Ca 2+ Channel-Dependent Immunomodulation. Cells 2024; 13:468. [PMID: 38534312 DOI: 10.3390/cells13060468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 02/27/2024] [Accepted: 03/05/2024] [Indexed: 03/28/2024] Open
Abstract
Many essential biological processes are triggered by the proximity of molecules. Meanwhile, diverse approaches in synthetic biology, such as new biological parts or engineered cells, have opened up avenues to precisely control the proximity of molecules and eventually downstream signaling processes. This also applies to a main Ca2+ entry pathway into the cell, the so-called Ca2+ release-activated Ca2+ (CRAC) channel. CRAC channels are among other channels are essential in the immune response and are activated by receptor-ligand binding at the cell membrane. The latter initiates a signaling cascade within the cell, which finally triggers the coupling of the two key molecular components of the CRAC channel, namely the stromal interaction molecule, STIM, in the ER membrane and the plasma membrane Ca2+ ion channel, Orai. Ca2+ entry, established via STIM/Orai coupling, is essential for various immune cell functions, including cytokine release, proliferation, and cytotoxicity. In this review, we summarize the tools of synthetic biology that have been used so far to achieve precise control over the CRAC channel pathway and thus over downstream signaling events related to the immune response.
Collapse
Affiliation(s)
- Bernadett Bacsa
- Division of Medical Physics und Biophysics, Medical University of Graz, A-8010 Graz, Austria
| | - Valentina Hopl
- Institute of Biophysics, JKU Life Science Center, Johannes Kepler University Linz, A-4020 Linz, Austria
| | - Isabella Derler
- Institute of Biophysics, JKU Life Science Center, Johannes Kepler University Linz, A-4020 Linz, Austria
| |
Collapse
|
10
|
Kodakandla G, Akimzhanov AM, Boehning D. Regulatory mechanisms controlling store-operated calcium entry. Front Physiol 2023; 14:1330259. [PMID: 38169682 PMCID: PMC10758431 DOI: 10.3389/fphys.2023.1330259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 12/08/2023] [Indexed: 01/05/2024] Open
Abstract
Calcium influx through plasma membrane ion channels is crucial for many events in cellular physiology. Cell surface stimuli lead to the production of inositol 1,4,5-trisphosphate (IP3), which binds to IP3 receptors (IP3R) in the endoplasmic reticulum (ER) to release calcium pools from the ER lumen. This leads to the depletion of ER calcium pools, which has been termed store depletion. Store depletion leads to the dissociation of calcium ions from the EF-hand motif of the ER calcium sensor Stromal Interaction Molecule 1 (STIM1). This leads to a conformational change in STIM1, which helps it to interact with the plasma membrane (PM) at ER:PM junctions. At these ER:PM junctions, STIM1 binds to and activates a calcium channel known as Orai1 to form calcium release-activated calcium (CRAC) channels. Activation of Orai1 leads to calcium influx, known as store-operated calcium entry (SOCE). In addition to Orai1 and STIM1, the homologs of Orai1 and STIM1, such as Orai2/3 and STIM2, also play a crucial role in calcium homeostasis. The influx of calcium through the Orai channel activates a calcium current that has been termed the CRAC current. CRAC channels form multimers and cluster together in large macromolecular assemblies termed "puncta". How CRAC channels form puncta has been contentious since their discovery. In this review, we will outline the history of SOCE, the molecular players involved in this process, as well as the models that have been proposed to explain this critical mechanism in cellular physiology.
Collapse
Affiliation(s)
- Goutham Kodakandla
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ, United States
| | - Askar M. Akimzhanov
- Department of Biochemistry and Molecular Biology, McGovern Medical School, Houston, TX, United States
| | - Darren Boehning
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ, United States
| |
Collapse
|
11
|
Kodakandla G, Akimzhanov AM, Boehning D. Regulatory mechanisms controlling store-operated calcium entry. ARXIV 2023:arXiv:2309.06907v3. [PMID: 37744466 PMCID: PMC10516112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 09/26/2023]
Abstract
Calcium influx through plasma membrane ion channels is crucial for many events in cellular physiology. Cell surface stimuli lead to the production of inositol 1,4,5-trisphosphate (IP3), which binds to IP3 receptors (IP3R) in the endoplasmic reticulum (ER) to release calcium pools from the ER lumen. This leads to the depletion of ER calcium pools, which has been termed store depletion. Store depletion leads to the dissociation of calcium ions from the EF-hand motif of the ER calcium sensor Stromal Interaction Molecule 1 (STIM1). This leads to a conformational change in STIM1, which helps it to interact with the plasma membrane (PM) at ER:PM junctions. At these ER:PM junctions, STIM1 binds to and activates a calcium channel known as Orai1 to form calcium-release activated calcium (CRAC) channels. Activation of Orai1 leads to calcium influx, known as store-operated calcium entry (SOCE). In addition to Orai1 and STIM1, the homologs of Orai1 and STIM1, such as Orai2/3 and STIM2, also play a crucial role in calcium homeostasis. The influx of calcium through the Orai channel activates a calcium current that has been termed the CRAC current. CRAC channels form multimers and cluster together in large macromolecular assemblies termed "puncta". How CRAC channels form puncta has been contentious since their discovery. In this review, we will outline the history of SOCE, the molecular players involved in this process, as well as the models that have been proposed to explain this critical mechanism in cellular physiology.
Collapse
Affiliation(s)
- Goutham Kodakandla
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ, USA, 08103
| | - Askar M. Akimzhanov
- Department of Biochemistry and Molecular Biology, McGovern Medical School, Houston, Texas, USA, 77030
| | - Darren Boehning
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ, USA, 08103
| |
Collapse
|
12
|
Gross S, Womer L, Kappes DJ, Soboloff J. Multifaceted control of T cell differentiation by STIM1. Trends Biochem Sci 2023; 48:1083-1097. [PMID: 37696713 PMCID: PMC10787584 DOI: 10.1016/j.tibs.2023.08.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 08/08/2023] [Accepted: 08/18/2023] [Indexed: 09/13/2023]
Abstract
In T cells, stromal interaction molecule (STIM) and Orai are dispensable for conventional T cell development, but critical for activation and differentiation. This review focuses on novel STIM-dependent mechanisms for control of Ca2+ signals during T cell activation and its impact on mitochondrial function and transcriptional activation for control of T cell differentiation and function. We highlight areas that require further work including the roles of plasma membrane Ca2+ ATPase (PMCA) and partner of STIM1 (POST) in controlling Orai function. A major knowledge gap also exists regarding the independence of T cell development from STIM and Orai, despite compelling evidence that it requires Ca2+ signals. Resolving these and other outstanding questions ensures that the field will remain active for many years to come.
Collapse
Affiliation(s)
- Scott Gross
- Fels Cancer Institute for Personalized Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, 19140, USA
| | - Lauren Womer
- Fels Cancer Institute for Personalized Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, 19140, USA
| | | | - Jonathan Soboloff
- Fels Cancer Institute for Personalized Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, 19140, USA; Department of Cancer and Cellular Biology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, 19140, USA.
| |
Collapse
|
13
|
Criado Santos N, Bouvet S, Cruz Cobo M, Mandavit M, Bermont F, Castelbou C, Mansour F, Azam M, Giordano F, Nunes-Hasler P. Sec22b regulates phagosome maturation by promoting ORP8-mediated lipid exchange at endoplasmic reticulum-phagosome contact sites. Commun Biol 2023; 6:1008. [PMID: 37794132 PMCID: PMC10550925 DOI: 10.1038/s42003-023-05382-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 09/21/2023] [Indexed: 10/06/2023] Open
Abstract
Phagosome maturation is critical for immune defense, defining whether ingested material is destroyed or converted into antigens. Sec22b regulates phagosome maturation, yet how has remained unclear. Here we show Sec22b tethers endoplasmic reticulum-phagosome membrane contact sites (MCS) independently of the known tether STIM1. Sec22b knockdown increases calcium signaling, phagolysosome fusion and antigen degradation and alters phagosomal phospholipids PI(3)P, PS and PI(4)P. Levels of PI(4)P, a lysosome docking lipid, are rescued by Sec22b re-expression and by expression of the artificial tether MAPPER but not the MCS-disrupting mutant Sec22b-P33. Moreover, Sec22b co-precipitates with the PS/PI(4)P exchange protein ORP8. Wild-type, but not mutant ORP8 rescues phagosomal PI(4)P and reduces antigen degradation. Sec22b, MAPPER and ORP8 but not P33 or mutant-ORP8 restores phagolysosome fusion in knockdown cells. These findings clarify an alternative mechanism through which Sec22b controls phagosome maturation and beg a reassessment of the relative contribution of Sec22b-mediated fusion versus tethering to phagosome biology.
Collapse
Affiliation(s)
- Nina Criado Santos
- Department of Pathology and Immunology, Geneva Center for Inflammation Research, Faculty of Medicine, University of Geneva, Centre Médicale Universitaire, 1 Rue Michel-Servet, Geneva, Switzerland
| | - Samuel Bouvet
- Department of Cellular Physiology and Metabolism, Faculty of Medicine, University of Geneva, Centre Médicale Universitaire, 1 Rue Michel-Servet, Geneva, Switzerland
| | - Maria Cruz Cobo
- Department of Pathology and Immunology, Geneva Center for Inflammation Research, Faculty of Medicine, University of Geneva, Centre Médicale Universitaire, 1 Rue Michel-Servet, Geneva, Switzerland
| | - Marion Mandavit
- Department of Pathology and Immunology, Geneva Center for Inflammation Research, Faculty of Medicine, University of Geneva, Centre Médicale Universitaire, 1 Rue Michel-Servet, Geneva, Switzerland
| | - Flavien Bermont
- Department of Cellular Physiology and Metabolism, Faculty of Medicine, University of Geneva, Centre Médicale Universitaire, 1 Rue Michel-Servet, Geneva, Switzerland
| | - Cyril Castelbou
- Department of Cellular Physiology and Metabolism, Faculty of Medicine, University of Geneva, Centre Médicale Universitaire, 1 Rue Michel-Servet, Geneva, Switzerland
| | - Farah Mansour
- Department of Pathology and Immunology, Geneva Center for Inflammation Research, Faculty of Medicine, University of Geneva, Centre Médicale Universitaire, 1 Rue Michel-Servet, Geneva, Switzerland
| | - Maral Azam
- Department of Pathology and Immunology, Geneva Center for Inflammation Research, Faculty of Medicine, University of Geneva, Centre Médicale Universitaire, 1 Rue Michel-Servet, Geneva, Switzerland
| | - Francesca Giordano
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Université Paris-Saclay, Gif-sur-Yvette cedex, 91198, France
- Inserm U1280, Gif-sur-Yvette cedex, 91198, France
| | - Paula Nunes-Hasler
- Department of Pathology and Immunology, Geneva Center for Inflammation Research, Faculty of Medicine, University of Geneva, Centre Médicale Universitaire, 1 Rue Michel-Servet, Geneva, Switzerland.
| |
Collapse
|
14
|
Gil Montoya DC, Ornelas-Guevara R, Diercks BP, Guse AH, Dupont G. T cell Ca 2+ microdomains through the lens of computational modeling. Front Immunol 2023; 14:1235737. [PMID: 37860008 PMCID: PMC10582754 DOI: 10.3389/fimmu.2023.1235737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 09/18/2023] [Indexed: 10/21/2023] Open
Abstract
Cellular Ca2+ signaling is highly organized in time and space. Locally restricted and short-lived regions of Ca2+ increase, called Ca2+ microdomains, constitute building blocks that are differentially arranged to create cellular Ca2+ signatures controlling physiological responses. Here, we focus on Ca2+ microdomains occurring in restricted cytosolic spaces between the plasma membrane and the endoplasmic reticulum, called endoplasmic reticulum-plasma membrane junctions. In T cells, these microdomains have been finely characterized. Enough quantitative data are thus available to develop detailed computational models of junctional Ca2+ dynamics. Simulations are able to predict the characteristics of Ca2+ increases at the level of single channels and in junctions of different spatial configurations, in response to various signaling molecules. Thanks to the synergy between experimental observations and computational modeling, a unified description of the molecular mechanisms that create Ca2+ microdomains in the first seconds of T cell stimulation is emerging.
Collapse
Affiliation(s)
- Diana C. Gil Montoya
- The Calcium Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Roberto Ornelas-Guevara
- Unit of Theoretical Chronobiology, Faculté des Sciences CP231, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Björn-Philipp Diercks
- The Calcium Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Andreas H. Guse
- The Calcium Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Geneviève Dupont
- Unit of Theoretical Chronobiology, Faculté des Sciences CP231, Université Libre de Bruxelles (ULB), Brussels, Belgium
| |
Collapse
|
15
|
Ogier C, Solomon AMC, Lu Z, Recoules L, Klochkova A, Gabitova-Cornell L, Bayarmagnai B, Restifo D, Surumbayeva A, Vendramini-Costa DB, Deneka AY, Francescone R, Lilly AC, Sipman A, Gardiner JC, Luong T, Franco-Barraza J, Ibeme N, Cai KQ, Einarson MB, Nicolas E, Efimov A, Megill E, Snyder NW, Bousquet C, Cros J, Zhou Y, Golemis EA, Gligorijevic B, Soboloff J, Fuchs SY, Cukierman E, Astsaturov I. Trogocytosis of cancer-associated fibroblasts promotes pancreatic cancer growth and immune suppression via phospholipid scramblase anoctamin 6 (ANO6). BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.15.557802. [PMID: 37745612 PMCID: PMC10515956 DOI: 10.1101/2023.09.15.557802] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/26/2023]
Abstract
In pancreatic ductal adenocarcinoma (PDAC), the fibroblastic stroma constitutes most of the tumor mass and is remarkably devoid of functional blood vessels. This raises an unresolved question of how PDAC cells obtain essential metabolites and water-insoluble lipids. We have found a critical role for cancer-associated fibroblasts (CAFs) in obtaining and transferring lipids from blood-borne particles to PDAC cells via trogocytosis of CAF plasma membranes. We have also determined that CAF-expressed phospholipid scramblase anoctamin 6 (ANO6) is an essential CAF trogocytosis regulator required to promote PDAC cell survival. During trogocytosis, cancer cells and CAFs form synapse-like plasma membranes contacts that induce cytosolic calcium influx in CAFs via Orai channels. This influx activates ANO6 and results in phosphatidylserine exposure on CAF plasma membrane initiating trogocytosis and transfer of membrane lipids, including cholesterol, to PDAC cells. Importantly, ANO6-dependent trogocytosis also supports the immunosuppressive function of pancreatic CAFs towards cytotoxic T cells by promoting transfer of excessive amounts of cholesterol. Further, blockade of ANO6 antagonizes tumor growth via disruption of delivery of exogenous cholesterol to cancer cells and reverses immune suppression suggesting a potential new strategy for PDAC therapy.
Collapse
|
16
|
Petersen OH. What Does Physiological Mean? FUNCTION 2023; 4:zqad042. [PMID: 37601812 PMCID: PMC10433090 DOI: 10.1093/function/zqad042] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 08/01/2023] [Accepted: 08/01/2023] [Indexed: 08/22/2023] Open
Affiliation(s)
- Ole H Petersen
- School of Biosciences, Sir Martin Evans Building, Cardiff University, Wales CF10 3AX, UK
| |
Collapse
|
17
|
Gómez-Morón Á, Requena S, Pertusa C, Lozano-Prieto M, Calzada-Fraile D, Scagnetti C, Sánchez-García I, Calero-García AA, Izquierdo M, Martín-Cófreces NB. End-binding protein 1 regulates the metabolic fate of CD4 + T lymphoblasts and Jurkat T cells and the organization of the mitochondrial network. Front Immunol 2023; 14:1197289. [PMID: 37520527 PMCID: PMC10374013 DOI: 10.3389/fimmu.2023.1197289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 06/23/2023] [Indexed: 08/01/2023] Open
Abstract
The organization of the mitochondrial network is relevant for the metabolic fate of T cells and their ability to respond to TCR stimulation. This arrangement depends on cytoskeleton dynamics in response to TCR and CD28 activation, which allows the polarization of the mitochondria through their change in shape, and their movement along the microtubules towards the immune synapse. This work focus on the role of End-binding protein 1 (EB1), a protein that regulates tubulin polymerization and has been previously identified as a regulator of intracellular transport of CD3-enriched vesicles. EB1-interferred cells showed defective intracellular organization and metabolic strength in activated T cells, pointing to a relevant connection of the cytoskeleton and metabolism in response to TCR stimulation, which leads to increased AICD. By unifying the organization of the tubulin cytoskeleton and mitochondria during CD4+ T cell activation, this work highlights the importance of this connection for critical cell asymmetry together with metabolic functions such as glycolysis, mitochondria respiration, and cell viability.
Collapse
Affiliation(s)
- Álvaro Gómez-Morón
- Immunology Service, Instituto de Investigación Sanitaria del Hospital Universitario La Princesa (IIS-Princesa), Madrid, Spain
- Immunology, Oftalmology and Otorrinolaryngology Dept., School of Medicine, Universidad Complutense de Madrid (UCM), Madrid, Spain
| | - Silvia Requena
- Immunology Service, Instituto de Investigación Sanitaria del Hospital Universitario La Princesa (IIS-Princesa), Madrid, Spain
| | - Clara Pertusa
- Immunology Service, Instituto de Investigación Sanitaria del Hospital Universitario La Princesa (IIS-Princesa), Madrid, Spain
| | - Marta Lozano-Prieto
- Immunology Service, Instituto de Investigación Sanitaria del Hospital Universitario La Princesa (IIS-Princesa), Madrid, Spain
| | - Diego Calzada-Fraile
- Vascular Pathophysiology, Laboratory of Intercellular Communication, Fundación Centro Nacional de Investigaciones Cardiovasculares-Carlos III (CNIC), Madrid, Spain
| | - Camila Scagnetti
- Immunology Service, Instituto de Investigación Sanitaria del Hospital Universitario La Princesa (IIS-Princesa), Madrid, Spain
- Videomicroscopy Unit, Instituto de Investigación Sanitaria del Hospital Universitario La Princesa, IIS-Princesa, Madrid, Spain
| | - Inés Sánchez-García
- Immunology Service, Instituto de Investigación Sanitaria del Hospital Universitario La Princesa (IIS-Princesa), Madrid, Spain
| | | | - Manuel Izquierdo
- Instituto de Investigaciones Biomédicas "Alberto Sols", Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), Madrid, Spain
| | - Noa B Martín-Cófreces
- Immunology Service, Instituto de Investigación Sanitaria del Hospital Universitario La Princesa (IIS-Princesa), Madrid, Spain
- Vascular Pathophysiology, Laboratory of Intercellular Communication, Fundación Centro Nacional de Investigaciones Cardiovasculares-Carlos III (CNIC), Madrid, Spain
- Videomicroscopy Unit, Instituto de Investigación Sanitaria del Hospital Universitario La Princesa, IIS-Princesa, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| |
Collapse
|
18
|
Weiß M, Hernandez LC, Gil Montoya DC, Löhndorf A, Krüger A, Kopdag M, Uebler L, Landwehr M, Nawrocki M, Huber S, Woelk LM, Werner R, Failla AV, Flügel A, Dupont G, Guse AH, Diercks BP. Adhesion to laminin-1 and collagen IV induces the formation of Ca 2+ microdomains that sensitize mouse T cells for activation. Sci Signal 2023; 16:eabn9405. [PMID: 37339181 DOI: 10.1126/scisignal.abn9405] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2022] [Accepted: 05/31/2023] [Indexed: 06/22/2023]
Abstract
During an immune response, T cells migrate from blood vessel walls into inflamed tissues by migrating across the endothelium and through extracellular matrix (ECM). Integrins facilitate T cell binding to endothelial cells and ECM proteins. Here, we report that Ca2+ microdomains observed in the absence of T cell receptor (TCR)/CD3 stimulation are initial signaling events triggered by adhesion to ECM proteins that increase the sensitivity of primary murine T cells to activation. Adhesion to the ECM proteins collagen IV and laminin-1 increased the number of Ca2+ microdomains in a manner dependent on the kinase FAK, phospholipase C (PLC), and all three inositol 1,4,5-trisphosphate receptor (IP3R) subtypes and promoted the nuclear translocation of the transcription factor NFAT-1. Mathematical modeling predicted that the formation of adhesion-dependent Ca2+ microdomains required the concerted activity of two to six IP3Rs and ORAI1 channels to achieve the increase in the Ca2+ concentration in the ER-plasma membrane junction that was observed experimentally and that required SOCE. Further, adhesion-dependent Ca2+ microdomains were important for the magnitude of the TCR-induced activation of T cells on collagen IV as assessed by the global Ca2+ response and NFAT-1 nuclear translocation. Thus, adhesion to collagen IV and laminin-1 sensitizes T cells through a mechanism involving the formation of Ca2+ microdomains, and blocking this low-level sensitization decreases T cell activation upon TCR engagement.
Collapse
Affiliation(s)
- Mariella Weiß
- Calcium Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Lola C Hernandez
- Calcium Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Diana C Gil Montoya
- Calcium Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Anke Löhndorf
- Calcium Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Aileen Krüger
- Calcium Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Miriam Kopdag
- Calcium Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Liana Uebler
- Calcium Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Marie Landwehr
- Calcium Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Mikolaj Nawrocki
- Section of Molecular Immunology und Gastroenterology, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Samuel Huber
- Section of Molecular Immunology und Gastroenterology, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Lena-Marie Woelk
- Department of Computational Neuroscience, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - René Werner
- Department of Computational Neuroscience, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Antonio V Failla
- Microscopy Imaging Facility (UMIF), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Alexander Flügel
- Institute for Neuroimmunology and Multiple Sclerosis Research, University Medical Centre Göttingen, 37075 Göttingen, Germany
| | - Geneviève Dupont
- Unité de Chronobiologie Théorique, Faculté des Sciences, CP231, Université Libre de Bruxelles (ULB), B-1050 Brussels, Belgium
| | - Andreas H Guse
- Calcium Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Björn-Philipp Diercks
- Calcium Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| |
Collapse
|
19
|
Chang HF, Schirra C, Pattu V, Krause E, Becherer U. Lytic granule exocytosis at immune synapses: lessons from neuronal synapses. Front Immunol 2023; 14:1177670. [PMID: 37275872 PMCID: PMC10233144 DOI: 10.3389/fimmu.2023.1177670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 05/09/2023] [Indexed: 06/07/2023] Open
Abstract
Regulated exocytosis is a central mechanism of cellular communication. It is not only the basis for neurotransmission and hormone release, but also plays an important role in the immune system for the release of cytokines and cytotoxic molecules. In cytotoxic T lymphocytes (CTLs), the formation of the immunological synapse is required for the delivery of the cytotoxic substances such as granzymes and perforin, which are stored in lytic granules and released via exocytosis. The molecular mechanisms of their fusion with the plasma membrane are only partially understood. In this review, we discuss the molecular players involved in the regulated exocytosis of CTL, highlighting the parallels and differences to neuronal synaptic transmission. Additionally, we examine the strengths and weaknesses of both systems to study exocytosis.
Collapse
|
20
|
Garcia-Casas P, Rossini M, Filadi R, Pizzo P. Mitochondrial Ca 2+ signaling and Alzheimer's disease: Too much or too little? Cell Calcium 2023; 113:102757. [PMID: 37192560 DOI: 10.1016/j.ceca.2023.102757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 05/08/2023] [Accepted: 05/09/2023] [Indexed: 05/18/2023]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease, caused by poorly known pathogenic mechanisms and aggravated by delayed therapeutic intervention, that still lacks an effective cure. However, it is clear that some important neurophysiological processes are altered years before the onset of clinical symptoms, offering the possibility of identifying biological targets useful for implementation of new therapies. Of note, evidence has been provided suggesting that mitochondria, pivotal organelles in sustaining neuronal energy demand and modulating synaptic activity, are dysfunctional in AD samples. In particular, alterations in mitochondrial Ca2+ signaling have been proposed as causal events for neurodegeneration, although the exact outcomes and molecular mechanisms of these defects, as well as their longitudinal progression, are not always clear. Here, we discuss the importance of a correct mitochondrial Ca2+ handling for neuronal physiology and summarize the latest findings on dysfunctional mitochondrial Ca2+ pathways in AD, analysing possible consequences contributing to the neurodegeneration that characterizes the disease.
Collapse
Affiliation(s)
- Paloma Garcia-Casas
- Department of Biomedical Sciences, University of Padova, 35131 Padua, Italy; Department of Biochemistry and Molecular Biology and Physiology, School of Medicine, University of Valladolid, 47003 Valladolid, Spain
| | - Michela Rossini
- Department of Biomedical Sciences, University of Padova, 35131 Padua, Italy
| | - Riccardo Filadi
- Department of Biomedical Sciences, University of Padova, 35131 Padua, Italy; Neuroscience Institute, National Research Council (CNR), 35131 Padua, Italy.
| | - Paola Pizzo
- Department of Biomedical Sciences, University of Padova, 35131 Padua, Italy; Neuroscience Institute, National Research Council (CNR), 35131 Padua, Italy; Study Centre for Neurodegeneration (CESNE), University of Padova, 35131 Padua, Italy.
| |
Collapse
|
21
|
Jost P, Klein F, Brand B, Wahl V, Wyatt A, Yildiz D, Boehm U, Niemeyer BA, Vaeth M, Alansary D. Acute Downregulation but Not Genetic Ablation of Murine MCU Impairs Suppressive Capacity of Regulatory CD4 T Cells. Int J Mol Sci 2023; 24:ijms24097772. [PMID: 37175478 PMCID: PMC10178810 DOI: 10.3390/ijms24097772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 04/17/2023] [Accepted: 04/20/2023] [Indexed: 05/15/2023] Open
Abstract
By virtue of mitochondrial control of energy production, reactive oxygen species (ROS) generation, and maintenance of Ca2+ homeostasis, mitochondria play an essential role in modulating T cell function. The mitochondrial Ca2+ uniporter (MCU) is the pore-forming unit in the main protein complex mediating mitochondrial Ca2+ uptake. Recently, MCU has been shown to modulate Ca2+ signals at subcellular organellar interfaces, thus fine-tuning NFAT translocation and T cell activation. The mechanisms underlying this modulation and whether MCU has additional T cell subpopulation-specific effects remain elusive. However, mice with germline or tissue-specific ablation of Mcu did not show impaired T cell responses in vitro or in vivo, indicating that 'chronic' loss of MCU can be functionally compensated in lymphocytes. The current work aimed to specifically investigate whether and how MCU influences the suppressive potential of regulatory CD4 T cells (Treg). We show that, in contrast to genetic ablation, acute siRNA-mediated downregulation of Mcu in murine Tregs results in a significant reduction both in mitochondrial Ca2+ uptake and in the suppressive capacity of Tregs, while the ratios of Treg subpopulations and the expression of hallmark transcription factors were not affected. These findings suggest that permanent genetic inactivation of MCU may result in compensatory adaptive mechanisms, masking the effects on the suppressive capacity of Tregs.
Collapse
Affiliation(s)
- Priska Jost
- Molecular Biophysics, Saarland University, 66421 Homburg, Germany
| | - Franziska Klein
- Molecular Biophysics, Saarland University, 66421 Homburg, Germany
| | - Benjamin Brand
- Würzburg Institute of Systems Immunology, Max Planck Research Group at Julius-Maximilians University of Würzburg, 97078 Würzburg, Germany
| | - Vanessa Wahl
- Experimental Pharmacology, Center for Molecular Signaling (PZMS), School of Medicine, Saarland University, 66421 Homburg, Germany
| | - Amanda Wyatt
- Experimental Pharmacology, Center for Molecular Signaling (PZMS), School of Medicine, Saarland University, 66421 Homburg, Germany
| | - Daniela Yildiz
- Experimental Pharmacology, Center for Molecular Signaling (PZMS), School of Medicine, Saarland University, 66421 Homburg, Germany
| | - Ulrich Boehm
- Experimental Pharmacology, Center for Molecular Signaling (PZMS), School of Medicine, Saarland University, 66421 Homburg, Germany
| | | | - Martin Vaeth
- Würzburg Institute of Systems Immunology, Max Planck Research Group at Julius-Maximilians University of Würzburg, 97078 Würzburg, Germany
| | - Dalia Alansary
- Molecular Biophysics, Saarland University, 66421 Homburg, Germany
| |
Collapse
|
22
|
Beckmann D, Langnaese K, Gottfried A, Hradsky J, Tedford K, Tiwari N, Thomas U, Fischer KD, Korthals M. Ca 2+ Homeostasis by Plasma Membrane Ca 2+ ATPase (PMCA) 1 Is Essential for the Development of DP Thymocytes. Int J Mol Sci 2023; 24:ijms24021442. [PMID: 36674959 PMCID: PMC9865543 DOI: 10.3390/ijms24021442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 12/22/2022] [Accepted: 12/30/2022] [Indexed: 01/13/2023] Open
Abstract
The strength of Ca2+ signaling is a hallmark of T cell activation, yet the role of Ca2+ homeostasis in developing T cells before expressing a mature T cell receptor is poorly understood. We aimed to unveil specific functions of the two plasma membrane Ca2+ ATPases expressed in T cells, PMCA1 and PMCA4. On a transcriptional and protein level we found that PMCA4 was expressed at low levels in CD4-CD8- double negative (DN) thymocytes and was even downregulated in subsequent stages while PMCA1 was present throughout development and upregulated in CD4+CD8+ double positive (DP) thymocytes. Mice with a targeted deletion of Pmca1 in DN3 thymocytes had an almost complete block of DP thymocyte development with an accumulation of DN4 thymocytes but severely reduced numbers of CD8+ immature single positive (ISP) thymocytes. The DN4 thymocytes of these mice showed strongly elevated basal cytosolic Ca2+ levels and a pre-mature CD5 expression, but in contrast to the DP thymocytes they were only mildly prone to apoptosis. Surprisingly, mice with a germline deletion of Pmca4 did not show any signs of altered progression through the developmental thymocyte stages, nor altered Ca2+ homeostasis throughout this process. PMCA1 is, therefore, non-redundant in keeping cellular Ca2+ levels low in the early thymocyte development required for the DN to DP transition.
Collapse
Affiliation(s)
- David Beckmann
- Institute for Biochemistry and Cell Biology, Medical Faculty, Otto-von-Guericke-University Magdeburg, 39120 Magdeburg, Germany
| | - Kristina Langnaese
- Institute for Biochemistry and Cell Biology, Medical Faculty, Otto-von-Guericke-University Magdeburg, 39120 Magdeburg, Germany
| | - Anna Gottfried
- Institute for Biochemistry and Cell Biology, Medical Faculty, Otto-von-Guericke-University Magdeburg, 39120 Magdeburg, Germany
| | - Johannes Hradsky
- Institute for Biochemistry and Cell Biology, Medical Faculty, Otto-von-Guericke-University Magdeburg, 39120 Magdeburg, Germany
| | - Kerry Tedford
- Institute for Biochemistry and Cell Biology, Medical Faculty, Otto-von-Guericke-University Magdeburg, 39120 Magdeburg, Germany
| | - Nikhil Tiwari
- Department of Cellular Neuroscience, Leibniz Institute for Neurobiology, 39120 Magdeburg, Germany
| | - Ulrich Thomas
- Department of Cellular Neuroscience, Leibniz Institute for Neurobiology, 39120 Magdeburg, Germany
| | - Klaus-Dieter Fischer
- Institute for Biochemistry and Cell Biology, Medical Faculty, Otto-von-Guericke-University Magdeburg, 39120 Magdeburg, Germany
- Correspondence:
| | - Mark Korthals
- Institute for Biochemistry and Cell Biology, Medical Faculty, Otto-von-Guericke-University Magdeburg, 39120 Magdeburg, Germany
| |
Collapse
|
23
|
Sanchez GM, Incedal TC, Prada J, O'Callaghan P, Dyachok O, Echeverry S, Dumral Ö, Nguyen PM, Xie B, Barg S, Kreuger J, Dandekar T, Idevall-Hagren O. The β-cell primary cilium is an autonomous Ca2+ compartment for paracrine GABA signaling. J Cell Biol 2023; 222:213674. [PMID: 36350286 DOI: 10.1083/jcb.202108101] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 04/11/2022] [Accepted: 10/12/2022] [Indexed: 11/11/2022] Open
Abstract
The primary cilium is an organelle present in most adult mammalian cells that is considered as an antenna for sensing the local microenvironment. Here, we use intact mouse pancreatic islets of Langerhans to investigate signaling properties of the primary cilium in insulin-secreting β-cells. We find that GABAB1 receptors are strongly enriched at the base of the cilium, but are mobilized to more distal locations upon agonist binding. Using cilia-targeted Ca2+ indicators, we find that activation of GABAB1 receptors induces selective Ca2+ influx into primary cilia through a mechanism that requires voltage-dependent Ca2+ channel activation. Islet β-cells utilize cytosolic Ca2+ increases as the main trigger for insulin secretion, yet we find that increases in cytosolic Ca2+ fail to propagate into the cilium, and that this isolation is largely due to enhanced Ca2+ extrusion in the cilium. Our work reveals local GABA action on primary cilia that involves Ca2+ influx and depends on restricted Ca2+ diffusion between the cilium and cytosol.
Collapse
Affiliation(s)
| | | | - Juan Prada
- Department of Bioinformatics, University of Würzburg, Würzburg, Germany
| | - Paul O'Callaghan
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Oleg Dyachok
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | | | - Özge Dumral
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Phuoc My Nguyen
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Beichen Xie
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Sebastian Barg
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Johan Kreuger
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Thomas Dandekar
- Department of Bioinformatics, University of Würzburg, Würzburg, Germany
| | | |
Collapse
|
24
|
Colussi DM, Stathopulos PB. From passage to inhibition: Uncovering the structural and physiological inhibitory mechanisms of MCUb in mitochondrial calcium regulation. FASEB J 2023; 37:e22678. [PMID: 36538269 PMCID: PMC10107711 DOI: 10.1096/fj.202201080r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 10/14/2022] [Accepted: 11/21/2022] [Indexed: 12/24/2022]
Abstract
Mitochondrial calcium (Ca2+ ) regulation is critically implicated in the regulation of bioenergetics and cell fate. Ca2+ , a universal signaling ion, passively diffuses into the mitochondrial intermembrane space (IMS) through voltage-dependent anion channels (VDAC), where uptake into the matrix is tightly regulated across the inner mitochondrial membrane (IMM) by the mitochondrial Ca2+ uniporter complex (mtCU). In recent years, immense progress has been made in identifying and characterizing distinct structural and physiological mechanisms of mtCU component function. One of the main regulatory components of the Ca2+ selective mtCU channel is the mitochondrial Ca2+ uniporter dominant-negative beta subunit (MCUb). The structural mechanisms underlying the inhibitory effect(s) exerted by MCUb are poorly understood, despite high homology to the main mitochondrial Ca2+ uniporter (MCU) channel-forming subunits. In this review, we provide an overview of the structural differences between MCUb and MCU, believed to contribute to the inhibition of mitochondrial Ca2+ uptake. We highlight the possible structural rationale for the absent interaction between MCUb and the mitochondrial Ca2+ uptake 1 (MICU1) gatekeeping subunit and a potential widening of the pore upon integration of MCUb into the channel. We discuss physiological and pathophysiological information known about MCUb, underscoring implications in cardiac function and arrhythmia as a basis for future therapeutic discovery. Finally, we discuss potential post-translational modifications on MCUb as another layer of important regulation.
Collapse
Affiliation(s)
- Danielle M Colussi
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Peter B Stathopulos
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| |
Collapse
|
25
|
Xia Y, Gao B, Zhang X. Targeting mitochondrial quality control of T cells: Regulating the immune response in HCC. Front Oncol 2022; 12:993437. [PMID: 36212470 PMCID: PMC9539266 DOI: 10.3389/fonc.2022.993437] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 09/06/2022] [Indexed: 11/30/2022] Open
Abstract
Most of the primary hepatocellular carcinoma (HCC) develops from Viral Hepatitis including Hepatitis B virus, Hepatitis C Virus, and Nonalcoholic Steatohepatitis. Herein, T cells play crucial roles combined with chronic inflammation and chronic viral infection. However, T cells are gradually exhausted under chronic antigenic stimulation, which leads to T cell exhaustion in the tumor microenvironment, and the exhaustion is associated with mitochondrial dysfunction in T cells. Meanwhile, mitochondria play a crucial role in altering T cells’ metabolism modes to achieve desirable immunological responses, wherein mitochondria maintain quality control (MQC) and promote metabolism regulation in the microenvironment. Although immune checkpoint inhibitors have been widely used in clinical practice, there are some limitations in the therapeutic effect, thus combining immune checkpoint inhibitors with targeting mitochondrial biogenesis may enhance cellular metabolic adaptation and reverse the exhausted state. At present, several studies on mitochondrial quality control in HCC have been reported, however, there are gaps in the regulation of immune cell function by mitochondrial metabolism, particularly the modulating of T cell immune function. Hence, this review summarizes and discusses existing studies on the effects of MQC on T cell populations in liver diseases induced by HCC, it would be clued by mitochondrial quality control events.
Collapse
Affiliation(s)
- Yixue Xia
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
- Shanghai Key Lab of Human Performance, Shanghai University of Sport, Shanghai, China
| | - Binghong Gao
- School of Elite Sport, Shanghai University of Sport, Shanghai, China
- Shanghai Key Lab of Human Performance, Shanghai University of Sport, Shanghai, China
- *Correspondence: Binghong Gao, ; Xue Zhang,
| | - Xue Zhang
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
- School of Elite Sport, Shanghai University of Sport, Shanghai, China
- Shanghai Key Lab of Human Performance, Shanghai University of Sport, Shanghai, China
- *Correspondence: Binghong Gao, ; Xue Zhang,
| |
Collapse
|
26
|
Barak P, Kaur S, Scappini E, Tucker CJ, Parekh AB. Plasma Membrane Ca 2+ ATPase Activity Enables Sustained Store-operated Ca 2+ Entry in the Absence of a Bulk Cytosolic Ca 2+ Rise. FUNCTION 2022; 3:zqac040. [PMID: 38989036 PMCID: PMC11234650 DOI: 10.1093/function/zqac040] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 08/11/2022] [Accepted: 08/12/2022] [Indexed: 07/12/2024] Open
Abstract
In many cell types, the rise in cytosolic Ca2+ due to opening of Ca2+ release-activated Ca2+ (CRAC) channels drives a plethora of responses, including secretion, motility, energy production, and gene expression. The amplitude and time course of the cytosolic Ca2+ rise is shaped by the rates of Ca2+ entry into and removal from the cytosol. However, an extended bulk Ca2+ rise is toxic to cells. Here, we show that the plasma membrane Ca2+ ATPase (PMCA) pump plays a major role in preventing a prolonged cytosolic Ca2+ signal following CRAC channel activation. Ca2+ entry through CRAC channels leads to a sustained sub-plasmalemmal Ca2+ rise but bulk Ca2+ is kept low by the activity of PMCA4b. Despite the low cytosolic Ca2+, membrane permeability to Ca2+ is still elevated and Ca2+ continues to enter through CRAC channels. Ca2+-dependent NFAT activation, driven by Ca2+ nanodomains near the open channels, is maintained despite the return of bulk Ca2+ to near pre-stimulation levels. Our data reveal a central role for PMCA4b in determining the pattern of a functional Ca2+ signal and in sharpening local Ca2+ gradients near CRAC channels, whilst protecting cells from a toxic Ca2+ overload.
Collapse
Affiliation(s)
- Pradeep Barak
- Department of Physiology, Anatomy and Genetics, Oxford University, Oxford OX1 3PT, UK
- Oxford Nanoimaging, Linacre House, Jordan Hill Business Park Banbury Road, Oxford OX2 8TA, UK
| | - Suneet Kaur
- Laboratory of Signal Transduction, National Institute of Environmental Health Sciences, NIH, Research Triangle Park NC 27709, USA
| | - Erica Scappini
- Laboratory of Signal Transduction, National Institute of Environmental Health Sciences, NIH, Research Triangle Park NC 27709, USA
| | - Charles J Tucker
- Laboratory of Signal Transduction, National Institute of Environmental Health Sciences, NIH, Research Triangle Park NC 27709, USA
| | - Anant B Parekh
- Department of Physiology, Anatomy and Genetics, Oxford University, Oxford OX1 3PT, UK
- Laboratory of Signal Transduction, National Institute of Environmental Health Sciences, NIH, Research Triangle Park NC 27709, USA
| |
Collapse
|
27
|
Molon B, Liboni C, Viola A. CD28 and chemokine receptors: Signalling amplifiers at the immunological synapse. Front Immunol 2022; 13:938004. [PMID: 35983040 PMCID: PMC9379342 DOI: 10.3389/fimmu.2022.938004] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 07/08/2022] [Indexed: 01/14/2023] Open
Abstract
T cells are master regulators of the immune response tuning, among others, B cells, macrophages and NK cells. To exert their functions requiring high sensibility and specificity, T cells need to integrate different stimuli from the surrounding microenvironment. A finely tuned signalling compartmentalization orchestrated in dynamic platforms is an essential requirement for the proper and efficient response of these cells to distinct triggers. During years, several studies have depicted the pivotal role of the cytoskeleton and lipid microdomains in controlling signalling compartmentalization during T cell activation and functions. Here, we discuss mechanisms responsible for signalling amplification and compartmentalization in T cell activation, focusing on the role of CD28, chemokine receptors and the actin cytoskeleton. We also take into account the detrimental effect of mutations carried by distinct signalling proteins giving rise to syndromes characterized by defects in T cell functionality.
Collapse
Affiliation(s)
- Barbara Molon
- Pediatric Research Institute “Città della Speranza”, Corso Stati Uniti, Padova, Italy
- Department of Biomedical Sciences, University of Padova, Padova, Italy
- *Correspondence: Barbara Molon,
| | - Cristina Liboni
- Pediatric Research Institute “Città della Speranza”, Corso Stati Uniti, Padova, Italy
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Antonella Viola
- Pediatric Research Institute “Città della Speranza”, Corso Stati Uniti, Padova, Italy
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| |
Collapse
|
28
|
Chen Y, Ye Y, Krauß PL, Löwe P, Pfeiffenberger M, Damerau A, Ehlers L, Buttgereit T, Hoff P, Buttgereit F, Gaber T. Age-related increase of mitochondrial content in human memory CD4+ T cells contributes to ROS-mediated increased expression of proinflammatory cytokines. Front Immunol 2022; 13:911050. [PMID: 35935995 PMCID: PMC9353942 DOI: 10.3389/fimmu.2022.911050] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 06/29/2022] [Indexed: 11/18/2022] Open
Abstract
Cellular metabolism modulates effector functions in human CD4+ T (Th) cells by providing energy and building blocks. Conversely, cellular metabolic responses are modulated by various influences, e.g., age. Thus, we hypothesized that metabolic reprogramming in human Th cells during aging modulates effector functions and contributes to “inflammaging”, an aging-related, chronic, sterile, low-grade inflammatory state characterized by specific proinflammatory cytokines. Analyzing the metabolic response of human naive and memory Th cells from young and aged individuals, we observed that memory Th cells exhibit higher glycolytic and mitochondrial fluxes than naive Th cells. In contrast, the metabolism of the latter was not affected by donor age. Memory Th cells from aged donors showed a higher respiratory capacity, mitochondrial content, and intracellular ROS production than those from young donors without altering glucose uptake and cellular ATP levels, which finally resulted in higher secreted amounts of proinflammatory cytokines, e.g., IFN-γ, IP-10 from memory Th cells taken from aged donors after TCR-stimulation which were sensitive to ROS inhibition. These findings suggest that metabolic reprogramming in human memory Th cells during aging results in an increased expression of proinflammatory cytokines through enhanced ROS production, which may contribute to the pathogenesis of inflammaging.
Collapse
Affiliation(s)
- Yuling Chen
- Department of Rheumatology and Clinical Immunology, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
- German Rheumatism Research Centre (DRFZ) Berlin, A Leibniz Institute, Berlin, Germany
| | - Yuanchun Ye
- Department of Gastroenterology, Fujian Medical University Affiliated First Quanzhou Hospital, Quanzhou, China
- Department of Hematology, Oncology and Tumor Immunology, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Pierre-Louis Krauß
- Department of Rheumatology and Clinical Immunology, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
- German Rheumatism Research Centre (DRFZ) Berlin, A Leibniz Institute, Berlin, Germany
| | - Pelle Löwe
- Department of Rheumatology and Clinical Immunology, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
- German Rheumatism Research Centre (DRFZ) Berlin, A Leibniz Institute, Berlin, Germany
| | - Moritz Pfeiffenberger
- Department of Rheumatology and Clinical Immunology, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
- German Rheumatism Research Centre (DRFZ) Berlin, A Leibniz Institute, Berlin, Germany
| | - Alexandra Damerau
- Department of Rheumatology and Clinical Immunology, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
- German Rheumatism Research Centre (DRFZ) Berlin, A Leibniz Institute, Berlin, Germany
| | - Lisa Ehlers
- Department of Rheumatology and Clinical Immunology, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
- German Rheumatism Research Centre (DRFZ) Berlin, A Leibniz Institute, Berlin, Germany
| | - Thomas Buttgereit
- Department of Rheumatology and Clinical Immunology, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
- Department of Dermatology, Venerology, and Allergology, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Paula Hoff
- Department of Rheumatology and Clinical Immunology, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
- Rheumatologie, Endokrinologikum Berlin, Berlin, Germany
| | - Frank Buttgereit
- Department of Rheumatology and Clinical Immunology, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
- German Rheumatism Research Centre (DRFZ) Berlin, A Leibniz Institute, Berlin, Germany
| | - Timo Gaber
- Department of Rheumatology and Clinical Immunology, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
- German Rheumatism Research Centre (DRFZ) Berlin, A Leibniz Institute, Berlin, Germany
- *Correspondence: Timo Gaber,
| |
Collapse
|
29
|
Osorio C, Sfera A, Anton JJ, Thomas KG, Andronescu CV, Li E, Yahia RW, Avalos AG, Kozlakidis Z. Virus-Induced Membrane Fusion in Neurodegenerative Disorders. Front Cell Infect Microbiol 2022; 12:845580. [PMID: 35531328 PMCID: PMC9070112 DOI: 10.3389/fcimb.2022.845580] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 03/01/2022] [Indexed: 12/15/2022] Open
Abstract
A growing body of epidemiological and research data has associated neurotropic viruses with accelerated brain aging and increased risk of neurodegenerative disorders. Many viruses replicate optimally in senescent cells, as they offer a hospitable microenvironment with persistently elevated cytosolic calcium, abundant intracellular iron, and low interferon type I. As cell-cell fusion is a major driver of cellular senescence, many viruses have developed the ability to promote this phenotype by forming syncytia. Cell-cell fusion is associated with immunosuppression mediated by phosphatidylserine externalization that enable viruses to evade host defenses. In hosts, virus-induced immune dysfunction and premature cellular senescence may predispose to neurodegenerative disorders. This concept is supported by novel studies that found postinfectious cognitive dysfunction in several viral illnesses, including human immunodeficiency virus-1, herpes simplex virus-1, and SARS-CoV-2. Virus-induced pathological syncytia may provide a unified framework for conceptualizing neuronal cell cycle reentry, aneuploidy, somatic mosaicism, viral spreading of pathological Tau and elimination of viable synapses and neurons by neurotoxic astrocytes and microglia. In this narrative review, we take a closer look at cell-cell fusion and vesicular merger in the pathogenesis of neurodegenerative disorders. We present a "decentralized" information processing model that conceptualizes neurodegeneration as a systemic illness, triggered by cytoskeletal pathology. We also discuss strategies for reversing cell-cell fusion, including, TMEM16F inhibitors, calcium channel blockers, senolytics, and tubulin stabilizing agents. Finally, going beyond neurodegeneration, we examine the potential benefit of harnessing fusion as a therapeutic strategy in regenerative medicine.
Collapse
Affiliation(s)
- Carolina Osorio
- Department of Psychiatry, Loma Linda University, Loma Linda, CA, United States
| | - Adonis Sfera
- Department of Psychiatry, Loma Linda University, Loma Linda, CA, United States
- Department of Psychiatry, Patton State Hospital, San Bernardino, CA, United States
| | - Jonathan J. Anton
- Department of Psychiatry, Patton State Hospital, San Bernardino, CA, United States
| | - Karina G. Thomas
- Department of Psychiatry, Patton State Hospital, San Bernardino, CA, United States
| | - Christina V. Andronescu
- Medical Anthropology – Department of Anthropology, Stanford University, Stanford, CA, United States
| | - Erica Li
- School of Medicine, University of California, Riverside, Riverside, CA, United States
| | - Rayan W. Yahia
- School of Medicine, University of California, Riverside, Riverside, CA, United States
| | - Andrea García Avalos
- Universidad Nacional Autónoma de México (UNAM), Facultad de Medicina Campus, Ciudad de Mexico, Mexico
| | - Zisis Kozlakidis
- International Agency for Research on Cancer (IARC), Lyon, France
| |
Collapse
|
30
|
Wu H, Brand B, Eckstein M, Hochrein SM, Shumanska M, Dudek J, Nickel A, Maack C, Bogeski I, Vaeth M. Genetic Ablation of the Mitochondrial Calcium Uniporter (MCU) Does not Impair T Cell-Mediated Immunity In Vivo. Front Pharmacol 2022; 12:734078. [PMID: 34987384 PMCID: PMC8721163 DOI: 10.3389/fphar.2021.734078] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 11/22/2021] [Indexed: 12/11/2022] Open
Abstract
T cell activation and differentiation is associated with metabolic reprogramming to cope with the increased bioenergetic demand and to provide metabolic intermediates for the biosynthesis of building blocks. Antigen receptor stimulation not only promotes the metabolic switch of lymphocytes but also triggers the uptake of calcium (Ca2+) from the cytosol into the mitochondrial matrix. Whether mitochondrial Ca2+ influx through the mitochondrial Ca2+ uniporter (MCU) controls T cell metabolism and effector function remained, however, enigmatic. Using mice with T cell-specific deletion of MCU, we here show that genetic inactivation of mitochondrial Ca2+ uptake increased cytosolic Ca2+ levels following antigen receptor stimulation and store-operated Ca2+ entry (SOCE). However, ablation of MCU and the elevation of cytosolic Ca2+ did not affect mitochondrial respiration, differentiation and effector function of inflammatory and regulatory T cell subsets in vitro and in animal models of T cell-mediated autoimmunity and viral infection. These data suggest that MCU-mediated mitochondrial Ca2+ uptake is largely dispensable for murine T cell function. Our study has also important technical implications. Previous studies relied mostly on pharmacological inhibition or transient knockdown of mitochondrial Ca2+ uptake, but our results using mice with genetic deletion of MCU did not recapitulate these findings. The discrepancy of our study to previous reports hint at compensatory mechanisms in MCU-deficient mice and/or off-target effects of current MCU inhibitors.
Collapse
Affiliation(s)
- Hao Wu
- Würzburg Institute of Systems Immunology, Max Planck Research Group at the Julius-Maximilians-Universität Würzburg, Würzburg, Germany
| | - Benjamin Brand
- Würzburg Institute of Systems Immunology, Max Planck Research Group at the Julius-Maximilians-Universität Würzburg, Würzburg, Germany
| | - Miriam Eckstein
- Würzburg Institute of Systems Immunology, Max Planck Research Group at the Julius-Maximilians-Universität Würzburg, Würzburg, Germany
| | - Sophia M Hochrein
- Würzburg Institute of Systems Immunology, Max Planck Research Group at the Julius-Maximilians-Universität Würzburg, Würzburg, Germany
| | - Magdalena Shumanska
- Molecular Physiology, Institute of Cardiovascular Physiology, University Medical Center, Georg-August-University, Göttingen, Germany
| | - Jan Dudek
- Comprehensive Heart Failure Center (CHFC), University Hospital, Julius-Maximilians University of Würzburg, Würzburg, Germany
| | - Alexander Nickel
- Comprehensive Heart Failure Center (CHFC), University Hospital, Julius-Maximilians University of Würzburg, Würzburg, Germany
| | - Christoph Maack
- Comprehensive Heart Failure Center (CHFC), University Hospital, Julius-Maximilians University of Würzburg, Würzburg, Germany
| | - Ivan Bogeski
- Molecular Physiology, Institute of Cardiovascular Physiology, University Medical Center, Georg-August-University, Göttingen, Germany
| | - Martin Vaeth
- Würzburg Institute of Systems Immunology, Max Planck Research Group at the Julius-Maximilians-Universität Würzburg, Würzburg, Germany
| |
Collapse
|
31
|
Genetically encoded photo-switchable molecular sensors for optoacoustic and super-resolution imaging. Nat Biotechnol 2022; 40:598-605. [PMID: 34845372 PMCID: PMC9005348 DOI: 10.1038/s41587-021-01100-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 09/17/2021] [Indexed: 02/07/2023]
Abstract
Reversibly photo-switchable proteins are essential for many super-resolution fluorescence microscopic and optoacoustic imaging methods. However, they have yet to be used as sensors that measure the distribution of specific analytes at the nanoscale or in the tissues of live animals. Here we constructed the prototype of a photo-switchable Ca2+ sensor based on GCaMP5G that can be switched with 405/488-nm light and describe its molecular mechanisms at the structural level, including the importance of the interaction of the core barrel structure of the fluorescent protein with the Ca2+ receptor moiety. We demonstrate super-resolution imaging of Ca2+ concentration in cultured cells and optoacoustic Ca2+ imaging in implanted tumor cells in mice under controlled Ca2+ conditions. Finally, we show the generalizability of the concept by constructing examples of photo-switching maltose and dopamine sensors based on periplasmatic binding protein and G-protein-coupled receptor-based sensors.
Collapse
|
32
|
Carreras-Sureda A, Abrami L, Ji-Hee K, Wang WA, Henry C, Frieden M, Didier M, van der Goot FG, Demaurex N. S-acylation by ZDHHC20 targets ORAI1 channels to lipid rafts for efficient Ca 2+ signaling by Jurkat T cell receptors at the immune synapse. eLife 2021; 10:72051. [PMID: 34913437 PMCID: PMC8683079 DOI: 10.7554/elife.72051] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 11/29/2021] [Indexed: 12/29/2022] Open
Abstract
Efficient immune responses require Ca2+ fluxes across ORAI1 channels during engagement of T cell receptors (TCR) at the immune synapse (IS) between T cells and antigen presenting cells. Here, we show that ZDHHC20-mediated S-acylation of the ORAI1 channel at residue Cys143 promotes TCR recruitment and signaling at the IS. Cys143 mutations reduced ORAI1 currents and store-operated Ca2+ entry in HEK-293 cells and nearly abrogated long-lasting Ca2+ elevations, NFATC1 translocation, and IL-2 secretion evoked by TCR engagement in Jurkat T cells. The acylation-deficient channel remained in cholesterol-poor domains upon enforced ZDHHC20 expression and was recruited less efficiently to the IS along with actin and TCR. Our results establish S-acylation as a critical regulator of ORAI1 channel trafficking and function at the IS and reveal that ORAI1 S-acylation enhances TCR recruitment to the synapse.
Collapse
Affiliation(s)
| | - Laurence Abrami
- Faculty of Life Sciences, Global Health Institute, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Kim Ji-Hee
- Department of Physiology, Yonsei University Wonju College of Medicine, Wonju, Republic of Korea
| | - Wen-An Wang
- Department of Cell Physiology and Metabolism, Geneva, Switzerland
| | | | - Maud Frieden
- Department of Cell Physiology and Metabolism, Geneva, Switzerland
| | - Monica Didier
- Department of Cell Physiology and Metabolism, Geneva, Switzerland
| | - F Gisou van der Goot
- Faculty of Life Sciences, Global Health Institute, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Nicolas Demaurex
- Department of Cell Physiology and Metabolism, Geneva, Switzerland
| |
Collapse
|
33
|
Voros O, Panyi G, Hajdu P. Immune Synapse Residency of Orai1 Alters Ca 2+ Response of T Cells. Int J Mol Sci 2021; 22:ijms222111514. [PMID: 34768945 PMCID: PMC8583858 DOI: 10.3390/ijms222111514] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 10/11/2021] [Accepted: 10/18/2021] [Indexed: 12/19/2022] Open
Abstract
CRAC, which plays important role in Ca2+-dependent T-lymphocyte activation, is composed of the ER-resident STIM1 and the plasma membrane Orai1 pore-forming subunit. Both accumulate at the immunological synapse (IS) between a T cell and an antigen-presenting cell (APC). We hypothesized that adapter/interacting proteins regulate Orai1 residence in the IS. We could show that mGFP-tagged Orai1-Full channels expressed in Jurkat cells had a biphasic IS-accumulation kinetics peaked at 15 min. To understand the background of Orai1 IS-redistribution we knocked down STIM1 and SAP97 (adaptor protein with a short IS-residency (15 min) and ability to bind Orai1 N-terminus): the mGFP-Orai1-Full channels kept on accumulating in the IS up to the 60th minute in the STIM1- and SAP97-lacking Jurkat cells. Deletion of Orai1 N terminus (mGFP-Orai1-Δ72) resulted in the same time course as described for STIM1/SAP97 knock-down cells. Ca2+-imaging of IS-engaged T-cells revealed that of Orai1 residency modifies the Ca2+-response: cells expressing mGFP-Orai1-Δ72 construct or mGFP-Orai1-Full in SAP-97 knock-down cells showed higher number of Ca2+-oscillation up to the 90th minute after IS formation. Overall, these data suggest that SAP97 may contribute to the short-lived IS-residency of Orai1 and binding of STIM1 to Orai1 N-terminus is necessary for SAP97-Orai1 interaction.
Collapse
Affiliation(s)
- Orsolya Voros
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Debrecen, Egyetem tér 1, 4032 Debrecen, Hungary; (O.V.); (G.P.)
| | - György Panyi
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Debrecen, Egyetem tér 1, 4032 Debrecen, Hungary; (O.V.); (G.P.)
| | - Péter Hajdu
- Department of Biophysics and Cell Biology, Faculty of Dentistry, University of Debrecen, Nagyerdei krt. 98, 4032 Debrecen, Hungary
- Correspondence: ; Tel.: +36-52-258603
| |
Collapse
|
34
|
Bohmwald K, Gálvez NMS, Andrade CA, Mora VP, Muñoz JT, González PA, Riedel CA, Kalergis AM. Modulation of Adaptive Immunity and Viral Infections by Ion Channels. Front Physiol 2021; 12:736681. [PMID: 34690811 PMCID: PMC8531258 DOI: 10.3389/fphys.2021.736681] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 09/10/2021] [Indexed: 12/15/2022] Open
Abstract
Most cellular functions require of ion homeostasis and ion movement. Among others, ion channels play a crucial role in controlling the homeostasis of anions and cations concentration between the extracellular and intracellular compartments. Calcium (Ca2+) is one of the most relevant ions involved in regulating critical functions of immune cells, allowing the appropriate development of immune cell responses against pathogens and tumor cells. Due to the importance of Ca2+ in inducing the immune response, some viruses have evolved mechanisms to modulate intracellular Ca2+ concentrations and the mobilization of this cation through Ca2+ channels to increase their infectivity and to evade the immune system using different mechanisms. For instance, some viral infections require the influx of Ca2+ through ionic channels as a first step to enter the cell, as well as their replication and budding. Moreover, through the expression of viral proteins on the surface of infected cells, Ca2+ channels function can be altered, enhancing the pathogen evasion of the adaptive immune response. In this article, we review those ion channels and ion transporters that are essential for the function of immune cells. Specifically, cation channels and Ca2+ channels in the context of viral infections and their contribution to the modulation of adaptive immune responses.
Collapse
Affiliation(s)
- Karen Bohmwald
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Nicolás M. S. Gálvez
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Catalina A. Andrade
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Valentina P. Mora
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - José T. Muñoz
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Pablo A. González
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Claudia A. Riedel
- Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Millennium Institute on Immunology and Immunotherapy, Universidad Andres Bello, Santiago, Chile
| | - Alexis M. Kalergis
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
- Departamento de Endocrinología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
35
|
Nan J, Li J, Lin Y, Saif Ur Rahman M, Li Z, Zhu L. The interplay between mitochondria and store-operated Ca 2+ entry: Emerging insights into cardiac diseases. J Cell Mol Med 2021; 25:9496-9512. [PMID: 34564947 PMCID: PMC8505841 DOI: 10.1111/jcmm.16941] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 08/20/2021] [Accepted: 09/08/2021] [Indexed: 12/14/2022] Open
Abstract
Store‐operated Ca2+ entry (SOCE) machinery, including Orai channels, TRPCs, and STIM1, is key to cellular calcium homeostasis. The following characteristics of mitochondria are involved in the physiological and pathological regulation of cells: mitochondria mediate calcium uptake through calcium uniporters; mitochondria are regulated by mitochondrial dynamic related proteins (OPA1, MFN1/2, and DRP1) and form mitochondrial networks through continuous fission and fusion; mitochondria supply NADH to the electron transport chain through the Krebs cycle to produce ATP; under stress, mitochondria will produce excessive reactive oxygen species to regulate mitochondria‐endoplasmic reticulum interactions and the related signalling pathways. Both SOCE and mitochondria play critical roles in mediating cardiac hypertrophy, diabetic cardiomyopathy, and cardiac ischaemia‐reperfusion injury. All the mitochondrial characteristics mentioned above are determinants of SOCE activity, and vice versa. Ca2+ signalling dictates the reciprocal regulation between mitochondria and SOCE under the specific pathological conditions of cardiomyocytes. The coupling of mitochondria and SOCE is essential for various pathophysiological processes in the heart. Herein, we review the research focussing on the reciprocal regulation between mitochondria and SOCE and provide potential interplay patterns in cardiac diseases.
Collapse
Affiliation(s)
- Jinliang Nan
- Provincial Key Cardiovascular Research Laboratory, Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang Province, Hangzhou, China
| | - Jiamin Li
- Provincial Key Cardiovascular Research Laboratory, Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang Province, Hangzhou, China
| | - Yinuo Lin
- Wenzhou Municipal Key Cardiovascular Research Laboratory, Department of Cardiology, The First Affiliated Hospital, Wenzhou Medical University, Zhejiang Province, Wenzhou, China
| | - Muhammad Saif Ur Rahman
- Zhejiang University-University of Edinburgh Biomedical Institute, Haining, Zhejiang, China.,Clinical Research Center, The Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, China
| | - Zhengzheng Li
- Department of Neurology, Research Institute of Experimental Neurobiology, The First Affiliated Hospital, Wenzhou Medical University, Zhejiang Province, Wenzhou, China
| | - Lingjun Zhu
- Provincial Key Cardiovascular Research Laboratory, Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang Province, Hangzhou, China
| |
Collapse
|
36
|
Yoast RE, Emrich SM, Zhang X, Xin P, Arige V, Pathak T, Benson JC, Johnson MT, Abdelnaby AE, Lakomski N, Hempel N, Han JM, Dupont G, Yule DI, Sneyd J, Trebak M. The Mitochondrial Ca 2+ uniporter is a central regulator of interorganellar Ca 2+ transfer and NFAT activation. J Biol Chem 2021; 297:101174. [PMID: 34499925 PMCID: PMC8496184 DOI: 10.1016/j.jbc.2021.101174] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 08/31/2021] [Accepted: 09/03/2021] [Indexed: 12/12/2022] Open
Abstract
Mitochondrial Ca2+ uptake tailors the strength of stimulation of plasma membrane phospholipase C–coupled receptors to that of cellular bioenergetics. However, how Ca2+ uptake by the mitochondrial Ca2+ uniporter (MCU) shapes receptor-evoked interorganellar Ca2+ signaling is unknown. Here, we used CRISPR/Cas9 gene knockout, subcellular Ca2+ imaging, and mathematical modeling to show that MCU is a universal regulator of intracellular Ca2+ signaling across mammalian cell types. MCU activity sustains cytosolic Ca2+ signaling by preventing Ca2+-dependent inactivation of store-operated Ca2+ release–activated Ca2+ channels and by inhibiting Ca2+ extrusion. Paradoxically, MCU knockout (MCU-KO) enhanced cytosolic Ca2+ responses to store depletion. Physiological agonist stimulation in MCU-KO cells led to enhanced frequency of cytosolic Ca2+ oscillations, endoplasmic reticulum Ca2+ refilling, nuclear translocation of nuclear factor for activated T cells transcription factors, and cell proliferation, without altering inositol-1,4,5-trisphosphate receptor activity. Our data show that MCU has dual counterbalancing functions at the cytosol–mitochondria interface, whereby the cell-specific MCU-dependent cytosolic Ca2+ clearance and buffering capacity of mitochondria reciprocally regulate interorganellar Ca2+ transfer and nuclear factor for activated T cells nuclear translocation during receptor-evoked signaling. These findings highlight the critical dual function of the MCU not only in the acute Ca2+ buffering by mitochondria but also in shaping endoplasmic reticulum and cytosolic Ca2+ signals that regulate cellular transcription and function.
Collapse
Affiliation(s)
- Ryan E Yoast
- Department of Cellular and Molecular Physiology, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania, USA
| | - Scott M Emrich
- Department of Cellular and Molecular Physiology, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania, USA
| | - Xuexin Zhang
- Department of Cellular and Molecular Physiology, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania, USA
| | - Ping Xin
- Department of Cellular and Molecular Physiology, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania, USA; Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA; Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Vikas Arige
- Department of Pharmacology and Physiology, University of Rochester, Rochester, New York, USA
| | - Trayambak Pathak
- Department of Cellular and Molecular Physiology, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania, USA; Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA; Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - J Cory Benson
- Department of Cellular and Molecular Physiology, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania, USA; Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA; Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Martin T Johnson
- Department of Cellular and Molecular Physiology, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania, USA
| | - Ahmed Emam Abdelnaby
- Department of Cellular and Molecular Physiology, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania, USA; Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA; Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Natalia Lakomski
- Department of Cellular and Molecular Physiology, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania, USA
| | - Nadine Hempel
- Department of Pharmacology, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania, USA
| | - Jung Min Han
- Laboratory of Biological Modeling, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Geneviève Dupont
- Unité de Chronobiologie Théorique, Université Libre de Bruxelles, Brussels, Belgium
| | - David I Yule
- Department of Pharmacology and Physiology, University of Rochester, Rochester, New York, USA
| | - James Sneyd
- Department of Mathematics, The University of Auckland, Auckland, New Zealand
| | - Mohamed Trebak
- Department of Cellular and Molecular Physiology, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania, USA; Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA; Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.
| |
Collapse
|
37
|
Montes de Oca Balderas P. Mitochondria-plasma membrane interactions and communication. J Biol Chem 2021; 297:101164. [PMID: 34481840 PMCID: PMC8503596 DOI: 10.1016/j.jbc.2021.101164] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 08/24/2021] [Accepted: 09/01/2021] [Indexed: 11/28/2022] Open
Abstract
Mitochondria are known as the powerhouses of eukaryotic cells; however, they perform many other functions besides oxidative phosphorylation, including Ca2+ homeostasis, lipid metabolism, antiviral response, and apoptosis. Although other hypotheses exist, mitochondria are generally thought as descendants of an α-proteobacteria that adapted to the intracellular environment within an Asgard archaebacteria, which have been studied for decades as an organelle subdued by the eukaryotic cell. Nevertheless, several early electron microscopy observations hinted that some mitochondria establish specific interactions with certain plasma membrane (PM) domains in mammalian cells. Furthermore, recent findings have documented the direct physical and functional interaction of mitochondria and the PM, the organization of distinct complexes, and their communication through vesicular means. In yeast, some molecular players mediating this interaction have been elucidated, but only a few works have studied this interaction in mammalian cells. In addition, mitochondria can be translocated among cells through tunneling nanotubes or by other mechanisms, and free, intact, functional mitochondria have been reported in the blood plasma. Together, these findings challenge the conception of mitochondria as organelles subdued by the eukaryotic cell. This review discusses the evidence of the mitochondria interaction with the PM that has been long disregarded despite its importance in cell function, pathogenesis, and evolution. It also proposes a scheme of mitochondria–PM interactions with the intent to promote research and knowledge of this emerging pathway that promises to shift the current paradigms of cell biology.
Collapse
Affiliation(s)
- Pavel Montes de Oca Balderas
- Unidad de Neurobiología Dinámica, Department of Neurochemistry, Instituto Nacional de Neurología y Neurocirugía, Mexico City, Mexico; Lab. BL-305, Instituto de Fisiología Celular, Department of Cognitive Neuroscience, Universidad Nacional Autónoma de México, Mexico City, Mexico.
| |
Collapse
|
38
|
Capera J, Pérez-Verdaguer M, Navarro-Pérez M, Felipe A. Kv1.3 Controls Mitochondrial Dynamics during Cell Cycle Progression. Cancers (Basel) 2021; 13:cancers13174457. [PMID: 34503267 PMCID: PMC8431373 DOI: 10.3390/cancers13174457] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 08/27/2021] [Accepted: 09/03/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Voltage-dependent potassium channels control the proliferation of mammalian cells. In addition, mitochondria physiology is highly dynamic during the cell cycle. The aim of this work was to investigate whether the Kv1.3 channel participates in the mitochondrial control of cell cycle progression. Our data confirmed that Kv1.3 facilitates the proliferation of preadipocytes through the control of mitochondrial dynamics. In addition, adipogenesis was also dependent on Kv1.3 expression. We shed light on the role of Kv1.3 in mitochondria and adipose tissue metabolism, contributing further to the control of cell proliferation by Kv1.3. Abstract The voltage-gated potassium channel Kv1.3 is a potential therapeutic target for obesity and diabetes. The genetic ablation and pharmacological inhibition of Kv1.3 lead to a lean phenotype in rodents. The mechanism of regulation of body weight and energy homeostasis involves Kv1.3 expression in different organs, including white and brown adipose tissues. Here, we show that Kv1.3 promotes the proliferation of preadipocytes through the control of mitochondrial dynamics. Kv1.3 is expressed in mitochondria exhibiting high affinity for the perinuclear population. The mitochondrial network is highly dynamic during the cell cycle, showing continuous fusion-fission events. The formation of a hyperfused mitochondrial network at the G1/S phase of the cell cycle is dependent on Kv1.3 expression. Our results demonstrate that Kv1.3 promotes preadipocyte proliferation and differentiation by controlling mitochondrial membrane potential and mitochondrial dynamics at the G1 phase of the cell cycle.
Collapse
Affiliation(s)
- Jesusa Capera
- Molecular Physiology Laboratory, Departament de Bioquímica i Biomedicina Molecular, Institut de Biomedicina (IBUB), Universitat de Barcelona, 08028 Barcelona, Spain; (J.C.); (M.P.-V.); (M.N.-P.)
- Kennedy Institute of Rheumatology, University of Oxford, Oxford OX3 7FY, UK
| | - Mireia Pérez-Verdaguer
- Molecular Physiology Laboratory, Departament de Bioquímica i Biomedicina Molecular, Institut de Biomedicina (IBUB), Universitat de Barcelona, 08028 Barcelona, Spain; (J.C.); (M.P.-V.); (M.N.-P.)
- Department of Cell Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - María Navarro-Pérez
- Molecular Physiology Laboratory, Departament de Bioquímica i Biomedicina Molecular, Institut de Biomedicina (IBUB), Universitat de Barcelona, 08028 Barcelona, Spain; (J.C.); (M.P.-V.); (M.N.-P.)
| | - Antonio Felipe
- Molecular Physiology Laboratory, Departament de Bioquímica i Biomedicina Molecular, Institut de Biomedicina (IBUB), Universitat de Barcelona, 08028 Barcelona, Spain; (J.C.); (M.P.-V.); (M.N.-P.)
- Correspondence:
| |
Collapse
|
39
|
Zhao M, Quintana A, Zhang C, Andreyev AY, Kiosses W, Kuwana T, Murphy A, Hogan PG, Kronenberg M. Calcium signals regulate the functional differentiation of thymic iNKT cells. EMBO J 2021; 40:e107901. [PMID: 34169542 PMCID: PMC8365263 DOI: 10.15252/embj.2021107901] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 05/20/2021] [Accepted: 05/25/2021] [Indexed: 11/09/2022] Open
Abstract
How natural or innate-like lymphocytes generate the capacity to produce IL-4 and other cytokines characteristic of type 2 immunity remains unknown. Invariant natural killer T (iNKT) cells differentiate in the thymus into NKT1, NKT2, and NKT17 subsets, similar to mature, peripheral CD4+ T helper cells. The mechanism for this differentiation was not fully understood. Here, we show that NKT2 cells required higher and prolonged calcium (Ca2+ ) signals and continuing activity of the calcium release-activated calcium (CRAC) channel, than their NKT1 counterparts. The sustained Ca2+ entry via CRAC pathway in NKT2 cells was apparently mediated by ORAI and controlled in part by the large mitochondrial Ca2+ uptake. Unique properties of mitochondria in NKT2 cells, including high activity of oxidative phosphorylation, may regulate mitochondrial Ca2+ buffering in NKT2 cells. In addition, the low Ca2+ extrusion rate may also contribute to the higher Ca2+ level in NKT2 cells. Altogether, we identified ORAI-dependent Ca2+ signaling connected with mitochondria and cellular metabolism, as a central regulatory pathway for the differentiation of NKT2 cells.
Collapse
Affiliation(s)
- Meng Zhao
- Division of Developmental ImmunologyLa Jolla Institute for ImmunologyLa JollaCAUSA
- Arthritis and Clinical Immunology ProgramOklahoma Medical Research FoundationOklahoma CityOKUSA
- Department of Microbiology and ImmunologyUniversity of Oklahoma Health Science CenterOklahoma CityOKUSA
| | - Ariel Quintana
- Division of Signaling and Gene ExpressionLa Jolla Institute for ImmunologyLa JollaCAUSA
- Translational Science DivisionClinical Science DepartmentMoffitt Cancer Center Magnolia CampusTampaFLUSA
| | - Chen Zhang
- Division of Signaling and Gene ExpressionLa Jolla Institute for ImmunologyLa JollaCAUSA
| | | | - William Kiosses
- Core MicroscopyLa Jolla Institute for ImmunologyLa JollaCAUSA
| | - Tomomi Kuwana
- Division of Immune RegulationLa Jolla Institute for ImmunologyLa JollaCAUSA
| | | | - Patrick G Hogan
- Division of Signaling and Gene ExpressionLa Jolla Institute for ImmunologyLa JollaCAUSA
- Moores Cancer CenterUniversity of California San DiegoLa JollaCAUSA
| | - Mitchell Kronenberg
- Division of Developmental ImmunologyLa Jolla Institute for ImmunologyLa JollaCAUSA
- Division of Biological SciencesUniversity of California, San DiegoLa JollaCAUSA
| |
Collapse
|
40
|
Separation of presynaptic Ca v2 and Ca v1 channel function in synaptic vesicle exo- and endocytosis by the membrane anchored Ca 2+ pump PMCA. Proc Natl Acad Sci U S A 2021; 118:2106621118. [PMID: 34244444 PMCID: PMC8285953 DOI: 10.1073/pnas.2106621118] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Synaptic vesicle (SV) release, recycling, and plastic changes of release probability co-occur side by side within nerve terminals and rely on local Ca2+ signals with different temporal and spatial profiles. The mechanisms that guarantee separate regulation of these vital presynaptic functions during action potential (AP)-triggered presynaptic Ca2+ entry remain unclear. Combining Drosophila genetics with electrophysiology and imaging reveals the localization of two different voltage-gated calcium channels at the presynaptic terminals of glutamatergic neuromuscular synapses (the Drosophila Cav2 homolog, Dmca1A or cacophony, and the Cav1 homolog, Dmca1D) but with spatial and functional separation. Cav2 within active zones is required for AP-triggered neurotransmitter release. By contrast, Cav1 localizes predominantly around active zones and contributes substantially to AP-evoked Ca2+ influx but has a small impact on release. Instead, L-type calcium currents through Cav1 fine-tune short-term plasticity and facilitate SV recycling. Separate control of SV exo- and endocytosis by AP-triggered presynaptic Ca2+ influx through different channels demands efficient measures to protect the neurotransmitter release machinery against Cav1-mediated Ca2+ influx. We show that the plasma membrane Ca2+ ATPase (PMCA) resides in between active zones and isolates Cav2-triggered release from Cav1-mediated dynamic regulation of recycling and short-term plasticity, two processes which Cav2 may also contribute to. As L-type Cav1 channels also localize next to PQ-type Cav2 channels within axon terminals of some central mammalian synapses, we propose that Cav2, Cav1, and PMCA act as a conserved functional triad that enables separate control of SV release and recycling rates in presynaptic terminals.
Collapse
|
41
|
Shi S, Zhao Q, Ke C, Long S, Zhang F, Zhang X, Li Y, Liu X, Hu H, Yin S. Loureirin B Exerts its Immunosuppressive Effects by Inhibiting STIM1/Orai1 and K V1.3 Channels. Front Pharmacol 2021; 12:685092. [PMID: 34248635 PMCID: PMC8268022 DOI: 10.3389/fphar.2021.685092] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 05/11/2021] [Indexed: 11/16/2022] Open
Abstract
Loureirin B (LrB) is a constituent extracted from traditional Chinese medicine Resina Draconis. It has broad biological functions and an impressive immunosuppressive effect that has been supported by numerous studies. However, the molecular mechanisms underlying Loureirin B-induced immune suppression are not fully understood. We previously reported that Loureirin B inhibited KV1.3 channel, calcium ion (Ca2+) influx, and interleukin-2 (IL-2) secretion in Jurkat T cells. In this study, we applied CRISPR/Cas9 to edit KV1.3 coding gene KCNA3 and successfully generated a KV1.3 knockout (KO) cell model to determine whether KV1.3 KO was sufficient to block the Loureirin B-induced immunosuppressive effect. Surprisingly, we showed that Loureirin B could still inhibit Ca2+ influx and IL-2 secretion in the Jurkat T cells in the absence of KV1.3 although KO KV1.3 reduced about 50% of Ca2+ influx and 90% IL-2 secretion compared with that in the wild type cells. Further experiments showed that Loureirin B directly inhibited STIM1/Orai1 channel in a dose-dependent manner. Our results suggest that Loureirin B inhibits Ca2+ influx and IL-2 secretion in Jurkat T cells by inhibiting both KV1.3 and STIM1/Orai1 channels. These studies also revealed an additional molecular target for Loureirin B-induced immunosuppressive effect, which makes it a promising leading compound for treating autoimmune diseases.
Collapse
Affiliation(s)
- Shujuan Shi
- Department of Chemical Biology, School of Pharmaceutical Sciences, South-Central University for Nationalities, Wuhan, China
| | - Qianru Zhao
- Department of Chemical Biology, School of Pharmaceutical Sciences, South-Central University for Nationalities, Wuhan, China
| | - Caihua Ke
- Department of Chemical Biology, School of Pharmaceutical Sciences, South-Central University for Nationalities, Wuhan, China
| | - Siru Long
- Department of Chemical Biology, School of Pharmaceutical Sciences, South-Central University for Nationalities, Wuhan, China
| | - Feng Zhang
- Department of Chemical Biology, School of Pharmaceutical Sciences, South-Central University for Nationalities, Wuhan, China
| | - Xu Zhang
- Department of Chemical Biology, School of Pharmaceutical Sciences, South-Central University for Nationalities, Wuhan, China
| | - Yi Li
- Department of Chemical Biology, School of Pharmaceutical Sciences, South-Central University for Nationalities, Wuhan, China
| | - Xinqiao Liu
- Department of Chemical Biology, School of Pharmaceutical Sciences, South-Central University for Nationalities, Wuhan, China
| | - Hongzhen Hu
- Department of Anesthesiology, the Center for the Study of Itch & Sensory Disorders, Washington University School of Medicine, St. Louis, MO, United States
| | - Shijin Yin
- Department of Chemical Biology, School of Pharmaceutical Sciences, South-Central University for Nationalities, Wuhan, China
| |
Collapse
|
42
|
Pallafacchina G, Zanin S, Rizzuto R. From the Identification to the Dissection of the Physiological Role of the Mitochondrial Calcium Uniporter: An Ongoing Story. Biomolecules 2021; 11:biom11060786. [PMID: 34071006 PMCID: PMC8224590 DOI: 10.3390/biom11060786] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Revised: 05/14/2021] [Accepted: 05/20/2021] [Indexed: 12/16/2022] Open
Abstract
The notion of mitochondria being involved in the decoding and shaping of intracellular Ca2+ signals has been circulating since the end of the 19th century. Despite that, the molecular identity of the channel that mediates Ca2+ ion transport into mitochondria remained elusive for several years. Only in the last decade, the genes and pathways responsible for the mitochondrial uptake of Ca2+ began to be cloned and characterized. The gene coding for the pore-forming unit of the mitochondrial channel was discovered exactly 10 years ago, and its product was called mitochondrial Ca2+ uniporter or MCU. Before that, only one of its regulators, the mitochondria Ca2+ uptake regulator 1, MICU1, has been described in 2010. However, in the following years, the scientific interest in mitochondrial Ca2+ signaling regulation and physiological role has increased. This shortly led to the identification of many of its components, to the description of their 3D structure, and the characterization of the uniporter contribution to tissue physiology and pathology. In this review, we will summarize the most relevant achievements in the history of mitochondrial Ca2+ studies, presenting a chronological overview of the most relevant and landmarking discoveries. Finally, we will explore the impact of mitochondrial Ca2+ signaling in the context of muscle physiology, highlighting the recent advances in understanding the role of the MCU complex in the control of muscle trophism and metabolism.
Collapse
Affiliation(s)
- Giorgia Pallafacchina
- Department of Biomedical Sciences, University of Padua, 35131 Padua, Italy
- Neuroscience Institute, Italian National Research Council (CNR), 35131 Padua, Italy
- Correspondence: (G.P.); (R.R.); Tel.: +39-049-827-6029 (G.P.); +39-049-827-3001 (R.R.)
| | - Sofia Zanin
- Department of Immunology, Infectiology and Haematology, Institut Necker-Enfants Malades (INEM), INSERM U1151-CNRS UMR 8253, 75015 Paris, France;
| | - Rosario Rizzuto
- Neuroscience Institute, Italian National Research Council (CNR), 35131 Padua, Italy
- Correspondence: (G.P.); (R.R.); Tel.: +39-049-827-6029 (G.P.); +39-049-827-3001 (R.R.)
| |
Collapse
|
43
|
Gil D, Guse AH, Dupont G. Three-Dimensional Model of Sub-Plasmalemmal Ca 2+ Microdomains Evoked by the Interplay Between ORAI1 and InsP 3 Receptors. Front Immunol 2021; 12:659790. [PMID: 33995380 PMCID: PMC8113648 DOI: 10.3389/fimmu.2021.659790] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 04/06/2021] [Indexed: 11/21/2022] Open
Abstract
Ca2+ signaling plays an essential role in T cell activation, which is a key step to start an adaptive immune response. During the transition from a quiescent to a fully activated state, Ca2+ microdomains characterized by reduced spatial and temporal extents are observed in the junctions between the plasma membrane (PM) and the endoplasmic reticulum (ER). Such Ca2+ responses can also occur in response to T cell adhesion to other cells or extracellular matrix proteins in otherwise unstimulated T cells. These non-TCR/CD3-dependent Ca2+ microdomains rely on d-myo-inositol 1,4,5-trisphosphate (IP3) signaling and subsequent store operated Ca2+ entry (SOCE) via the ORAI/STIM system. The detailed molecular mechanism of adhesion-dependent Ca2+ microdomain formation remains to be fully elucidated. We used mathematical modeling to investigate the spatiotemporal characteristics of T cell Ca2+ microdomains and their molecular regulators. We developed a reaction-diffusion model using COMSOL Multiphysics to describe the evolution of cytosolic and ER Ca2+ concentrations in a three-dimensional ER-PM junction. Equations are based on a previously proposed realistic description of the junction, which is extended to take into account IP3 receptors (IP3R) that are located next to the junction. The first model only considered the ORAI channels and the SERCA pumps. Taking into account the existence of preformed clusters of ORAI1 and STIM2, ORAI1 slightly opens in conditions of a full ER. These simulated Ca2+ microdomains are too small as compared to those observed in unstimulated T cells. When considering the opening of the IP3Rs located near the junction, the local depletion of ER Ca2+ allows for larger Ca2+ fluxes through the ORAI1 channels and hence larger local Ca2+ concentrations. Computational results moreover show that Ca2+ diffusion in the ER has a major impact on the Ca2+ changes in the junction, by affecting the local Ca2+ gradients in the sub-PM ER. Besides pointing out the likely involvement of the spontaneous openings of IP3Rs in the activation of SOCE in conditions of T cell adhesion prior to full activation, the model provides a tool to investigate how Ca2+ microdomains extent and interact in response to T cell receptor activation.
Collapse
Affiliation(s)
- Diana Gil
- The Ca2+ Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Andreas H Guse
- The Ca2+ Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Geneviève Dupont
- Unit of Theoretical Chronobiology, Faculté des Sciences CP231, Université Libre de Bruxelles (ULB), Brussels, Belgium
| |
Collapse
|
44
|
Stafford N, Zi M, Baudoin F, Mohamed TMA, Prehar S, De Giorgio D, Cartwright EJ, Latini R, Neyses L, Oceandy D. PMCA4 inhibition does not affect cardiac remodelling following myocardial infarction, but may reduce susceptibility to arrhythmia. Sci Rep 2021; 11:1518. [PMID: 33452399 PMCID: PMC7810749 DOI: 10.1038/s41598-021-81170-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 01/04/2021] [Indexed: 12/03/2022] Open
Abstract
Ischaemic heart disease is the world's leading cause of mortality. Survival rates from acute myocardial infarction (MI) have improved in recent years; however, this has led to an increase in the prevalence of heart failure (HF) due to chronic remodelling of the infarcted myocardium, for which treatment options remain poor. We have previously shown that inhibition of isoform 4 of the plasma membrane calcium ATPase (PMCA4) prevents chronic remodelling and HF development during pressure overload, through fibroblast mediated Wnt signalling modulation. Given that Wnt signalling also plays a prominent role during remodelling of the infarcted heart, this study investigated the effect of genetic and functional loss of PMCA4 on cardiac outcomes following MI. Neither genetic deletion nor pharmacological inhibition of PMCA4 affected chronic remodelling of the post-MI myocardium. This was the case when PMCA4 was deleted globally, or specifically from cardiomyocytes or fibroblasts. PMCA4-ablated hearts were however less prone to acute arrhythmic events, which may offer a slight survival benefit. Overall, this study demonstrates that PMCA4 inhibition does not affect chronic outcomes following MI.
Collapse
Affiliation(s)
- Nicholas Stafford
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK
| | - Min Zi
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK
| | - Florence Baudoin
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK
| | - Tamer M A Mohamed
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK
- Department of Medicine, Institute of Molecular Cardiology, University of Louisville, Louisville, KY, USA
- Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| | - Sukhpal Prehar
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK
| | - Daria De Giorgio
- Department of Cardiovascular Medicine, Mario Negri Institute for Pharmacological Research, Milan, Italy
| | - Elizabeth J Cartwright
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK
| | - Roberto Latini
- Department of Cardiovascular Medicine, Mario Negri Institute for Pharmacological Research, Milan, Italy
| | - Ludwig Neyses
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK
- Simply Uni, Sète, France
| | - Delvac Oceandy
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK.
| |
Collapse
|
45
|
A calcium optimum for cytotoxic T lymphocyte and natural killer cell cytotoxicity. Semin Cell Dev Biol 2020; 115:10-18. [PMID: 33358089 DOI: 10.1016/j.semcdb.2020.12.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 11/24/2020] [Accepted: 12/08/2020] [Indexed: 02/07/2023]
Abstract
Cytotoxic T lymphocytes (CTL) and natural killer (NK) cells are required for host defense. They destroy malignant target cells like cancer cells. Among metal cations, Ca2+ plays a prescinded role for CTL and NK cytotoxicity as it is the only cation used as ubiquitous second messenger. Measuring intracellular Ca2+ concentrations [Ca2+]int in single cells has greatly changed our understanding of Ca2+ signaling. Yet, comparing the role of Ca2+ in the pre-[Ca2+]int and [Ca2+]int measurement era reveals that even in the pre-[Ca2+]int measurement era (before 1980), the functions of Ca2+ and some other metal cations for the cytotoxic immune response were well established. It was even shown that Ca2+ influx across the plasma membrane but not Ca2+ release from intracellular sources is relevant for lymphocyte cytotoxicity and that very little Ca2+ is needed for efficient lymphocyte cytotoxicity against cancer cells. In the [Ca2+]int measurement era after 1980, many of the important findings were better and more quantitatively refined and in addition the molecules important for Ca2+ transport were defined. The unexpected finding that there is a Ca2+ optimum of CTL and NK cell cytotoxicity deserves some attention and may be important for anti-cancer therapy.
Collapse
|
46
|
Zöphel D, Hof C, Lis A. Altered Ca 2+ Homeostasis in Immune Cells during Aging: Role of Ion Channels. Int J Mol Sci 2020; 22:ijms22010110. [PMID: 33374304 PMCID: PMC7794837 DOI: 10.3390/ijms22010110] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 12/21/2020] [Accepted: 12/22/2020] [Indexed: 12/29/2022] Open
Abstract
Aging is an unstoppable process and begins shortly after birth. Each cell of the organism is affected by the irreversible process, not only with equal density but also at varying ages and with different speed. Therefore, aging can also be understood as an adaptation to a continually changing cellular environment. One of these very prominent changes in age affects Ca2+ signaling. Especially immune cells highly rely on Ca2+-dependent processes and a strictly regulated Ca2+ homeostasis. The intricate patterns of impaired immune cell function may represent a deficit or compensatory mechanisms. Besides, altered immune function through Ca2+ signaling can profoundly affect the development of age-related disease. This review attempts to summarize changes in Ca2+ signaling due to channels and receptors in T cells and beyond in the context of aging.
Collapse
Affiliation(s)
| | | | - Annette Lis
- Correspondence: ; Tel.: +49-(0)-06841-1616318; Fax: +49-(0)-6841-1616302
| |
Collapse
|
47
|
Martin-Cofreces NB, Chichon FJ, Calvo E, Torralba D, Bustos-Moran E, Dosil SG, Rojas-Gomez A, Bonzon-Kulichenko E, Lopez JA, Otón J, Sorrentino A, Zabala JC, Vernos I, Vazquez J, Valpuesta JM, Sanchez-Madrid F. The chaperonin CCT controls T cell receptor-driven 3D configuration of centrioles. SCIENCE ADVANCES 2020; 6:eabb7242. [PMID: 33268369 PMCID: PMC7821906 DOI: 10.1126/sciadv.abb7242] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 10/19/2020] [Indexed: 05/17/2023]
Abstract
T lymphocyte activation requires the formation of immune synapses (IS) with antigen-presenting cells. The dynamics of membrane receptors, signaling scaffolds, microfilaments, and microtubules at the IS determine the potency of T cell activation and subsequent immune response. Here, we show that the cytosolic chaperonin CCT (chaperonin-containing TCP1) controls the changes in reciprocal orientation of the centrioles and polarization of the tubulin dynamics induced by T cell receptor in T lymphocytes forming an IS. CCT also controls the mitochondrial ultrastructure and the metabolic status of T cells, regulating the de novo synthesis of tubulin as well as posttranslational modifications (poly-glutamylation, acetylation, Δ1 and Δ2) of αβ-tubulin heterodimers, fine-tuning tubulin dynamics. These changes ultimately determine the function and organization of the centrioles, as shown by three-dimensional reconstruction of resting and stimulated primary T cells using cryo-soft x-ray tomography. Through this mechanism, CCT governs T cell activation and polarity.
Collapse
Affiliation(s)
- N B Martin-Cofreces
- Immunology Service, Hospital Universitario de la Princesa, UAM, IIS-IP. Madrid, 28006 Spain.
- Area of Vascular Pathophysiology, Laboratory of Intercellular Communication, Fundación Centro Nacional de Investigaciones Cardiovasculares-Carlos III, Madrid, 28029 Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), 28029 Spain
| | - F J Chichon
- Department of Macromolecular Structure, Computational Systems Biology Group, Centro Nacional de Biotecnología (CNB-CSIC), Madrid, 28049, Spain
| | - E Calvo
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), 28029 Spain
- Laboratory of Cardiovascular Proteomics. Fundación Centro Nacional de Investigaciones Cardiovasculares-Carlos III, Madrid, 28029 Spain
| | - D Torralba
- Immunology Service, Hospital Universitario de la Princesa, UAM, IIS-IP. Madrid, 28006 Spain
- Area of Vascular Pathophysiology, Laboratory of Intercellular Communication, Fundación Centro Nacional de Investigaciones Cardiovasculares-Carlos III, Madrid, 28029 Spain
| | - E Bustos-Moran
- Immunology Service, Hospital Universitario de la Princesa, UAM, IIS-IP. Madrid, 28006 Spain
- Area of Vascular Pathophysiology, Laboratory of Intercellular Communication, Fundación Centro Nacional de Investigaciones Cardiovasculares-Carlos III, Madrid, 28029 Spain
| | - S G Dosil
- Immunology Service, Hospital Universitario de la Princesa, UAM, IIS-IP. Madrid, 28006 Spain
- Area of Vascular Pathophysiology, Laboratory of Intercellular Communication, Fundación Centro Nacional de Investigaciones Cardiovasculares-Carlos III, Madrid, 28029 Spain
| | - A Rojas-Gomez
- Immunology Service, Hospital Universitario de la Princesa, UAM, IIS-IP. Madrid, 28006 Spain
- Area of Vascular Pathophysiology, Laboratory of Intercellular Communication, Fundación Centro Nacional de Investigaciones Cardiovasculares-Carlos III, Madrid, 28029 Spain
| | - E Bonzon-Kulichenko
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), 28029 Spain
- Laboratory of Cardiovascular Proteomics. Fundación Centro Nacional de Investigaciones Cardiovasculares-Carlos III, Madrid, 28029 Spain
| | - J A Lopez
- Laboratory of Cardiovascular Proteomics. Fundación Centro Nacional de Investigaciones Cardiovasculares-Carlos III, Madrid, 28029 Spain
| | - J Otón
- Structural Studies Division, MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge CB2 0QH, UK
| | - A Sorrentino
- ALBA Synchrotron Light Source, Cerdanyola del Vallès, Barcelona 08290, Spain
| | - J C Zabala
- Departament of Molecular Biology, Facultad de Medicina, Universidad de Cantabria, Santander, 39005 Spain
| | - I Vernos
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Dr Aiguader 88, Barcelona, 08003, Spain
- Universitat Pompeu Fabra (UPF), Dr Aiguader 88, Barcelona 08003, Spain
- ICREA, Pg. Lluis Companys 23, Barcelona 08010, Spain
| | - J Vazquez
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), 28029 Spain
- Laboratory of Cardiovascular Proteomics. Fundación Centro Nacional de Investigaciones Cardiovasculares-Carlos III, Madrid, 28029 Spain
| | - J M Valpuesta
- Department of Macromolecular Structure, Computational Systems Biology Group, Centro Nacional de Biotecnología (CNB-CSIC), Madrid, 28049, Spain.
| | - F Sanchez-Madrid
- Immunology Service, Hospital Universitario de la Princesa, UAM, IIS-IP. Madrid, 28006 Spain.
- Area of Vascular Pathophysiology, Laboratory of Intercellular Communication, Fundación Centro Nacional de Investigaciones Cardiovasculares-Carlos III, Madrid, 28029 Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), 28029 Spain
| |
Collapse
|
48
|
Zhou Y, Zhao R, Schwarz EC, Akbar R, Kaba M, Pattu V, Helms V, Rieger H, Nunes-Hasler P, Qu B. Interorganelle Tethering to Endocytic Organelles Determines Directional Cytokine Transport in CD4 + T Cells. THE JOURNAL OF IMMUNOLOGY 2020; 205:2988-3000. [PMID: 33106338 DOI: 10.4049/jimmunol.2000195] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 09/20/2020] [Indexed: 12/24/2022]
Abstract
Delivery of vesicles to their desired destinations plays a central role in maintaining proper cell functionality. In certain scenarios, depending on loaded cargos, the vesicles have spatially distinct destinations. For example, in T cells, some cytokines (e.g., IL-2) are polarized to the T cell-target cell interface, whereas the other cytokines are delivered multidirectionally (e.g., TNF-α). In this study, we show that in primary human CD4+ T cells, both TNF-α+ and IL-2+ vesicles can tether with endocytic organelles (lysosomes/late endosomes) by forming membrane contact sites. Tethered cytokine-containing vesicle (CytV)-endocytic organelle pairs are released sequentially. Only endocytic organelle-tethered CytVs are preferentially transported to their desired destination. Mathematical models suggest that endocytic organelle tethering could regulate the direction of cytokine transport by selectively attaching different microtubule motor proteins (such as kinesin and dynein) to the corresponding CytVs. These findings establish the previously unknown interorganelle tethering to endocytic organelles as a universal solution for directional cytokine transport in CD4+ T cells. Modulating tethering to endocytic organelles can, therefore, coordinately control directionally distinct cytokine transport.
Collapse
Affiliation(s)
- Yan Zhou
- Department of Biophysics, Center for Integrative Physiology and Molecular Medicine, School of Medicine, Saarland University, 66421 Homburg, Germany
| | - Renping Zhao
- Department of Biophysics, Center for Integrative Physiology and Molecular Medicine, School of Medicine, Saarland University, 66421 Homburg, Germany
| | - Eva C Schwarz
- Department of Biophysics, Center for Integrative Physiology and Molecular Medicine, School of Medicine, Saarland University, 66421 Homburg, Germany
| | - Rahmad Akbar
- Center for Bioinformatics, Saarland University, 66123 Saarbrücken, Germany
| | - Mayis Kaba
- Department of Cell Physiology and Metabolism, University Medical Center, University of Geneva, 1211 Geneva, Switzerland
| | - Varsha Pattu
- Department of Physiology, Center for Integrative Physiology and Molecular Medicine, School of Medicine, Saarland University, 66421 Homburg, Germany
| | - Volkhard Helms
- Center for Bioinformatics, Saarland University, 66123 Saarbrücken, Germany
| | - Heiko Rieger
- Department of Theoretical Physics, Saarland University, 66123 Saarbrücken, Germany
| | - Paula Nunes-Hasler
- Department of Cell Physiology and Metabolism, University Medical Center, University of Geneva, 1211 Geneva, Switzerland.,Department of Pathology and Immunology, University Medical Center, University of Geneva, 1211 Geneva, Switzerland; and
| | - Bin Qu
- Department of Biophysics, Center for Integrative Physiology and Molecular Medicine, School of Medicine, Saarland University, 66421 Homburg, Germany; .,Leibniz Institute for New Materials, 66123 Saarbrücken, Germany
| |
Collapse
|
49
|
Mastrogiovanni M, Juzans M, Alcover A, Di Bartolo V. Coordinating Cytoskeleton and Molecular Traffic in T Cell Migration, Activation, and Effector Functions. Front Cell Dev Biol 2020; 8:591348. [PMID: 33195256 PMCID: PMC7609836 DOI: 10.3389/fcell.2020.591348] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 09/24/2020] [Indexed: 12/28/2022] Open
Abstract
Dynamic localization of receptors and signaling molecules at the plasma membrane and within intracellular vesicular compartments is crucial for T lymphocyte sensing environmental cues, triggering membrane receptors, recruiting signaling molecules, and fine-tuning of intracellular signals. The orchestrated action of actin and microtubule cytoskeleton and intracellular vesicle traffic plays a key role in all these events that together ensure important steps in T cell physiology. These include extravasation and migration through lymphoid and peripheral tissues, T cell interactions with antigen-presenting cells, T cell receptor (TCR) triggering by cognate antigen-major histocompatibility complex (MHC) complexes, immunological synapse formation, cell activation, and effector functions. Cytoskeletal and vesicle traffic dynamics and their interplay are coordinated by a variety of regulatory molecules. Among them, polarity regulators and membrane-cytoskeleton linkers are master controllers of this interplay. Here, we review the various ways the T cell plasma membrane, receptors, and their signaling machinery interplay with the actin and microtubule cytoskeleton and with intracellular vesicular compartments. We highlight the importance of this fine-tuned crosstalk in three key stages of T cell biology involving cell polarization: T cell migration in response to chemokines, immunological synapse formation in response to antigen cues, and effector functions. Finally, we discuss two examples of perturbation of this interplay in pathological settings, such as HIV-1 infection and mutation of the polarity regulator and tumor suppressor adenomatous polyposis coli (Apc) that leads to familial polyposis and colorectal cancer.
Collapse
Affiliation(s)
- Marta Mastrogiovanni
- Ligue Nationale Contre le Cancer – Equipe Labellisée LIGUE 2018, Lymphocyte Cell Biology Unit, INSERM-U1221, Department of Immunology, Institut Pasteur, Paris, France
- Collège Doctoral, Sorbonne Université, Paris, France
| | - Marie Juzans
- Ligue Nationale Contre le Cancer – Equipe Labellisée LIGUE 2018, Lymphocyte Cell Biology Unit, INSERM-U1221, Department of Immunology, Institut Pasteur, Paris, France
| | - Andrés Alcover
- Ligue Nationale Contre le Cancer – Equipe Labellisée LIGUE 2018, Lymphocyte Cell Biology Unit, INSERM-U1221, Department of Immunology, Institut Pasteur, Paris, France
| | - Vincenzo Di Bartolo
- Ligue Nationale Contre le Cancer – Equipe Labellisée LIGUE 2018, Lymphocyte Cell Biology Unit, INSERM-U1221, Department of Immunology, Institut Pasteur, Paris, France
| |
Collapse
|
50
|
|