1
|
Yi C, Yang J, Zhang T, Xie Z, Xiong Q, Chen D, Jiang S. lncRNA signature mediates mitochondrial permeability transition-driven necrosis in regulating the tumor immune microenvironment of cervical cancer. Sci Rep 2024; 14:17406. [PMID: 39075098 PMCID: PMC11286791 DOI: 10.1038/s41598-024-65990-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 06/26/2024] [Indexed: 07/31/2024] Open
Abstract
Mitochondrial permeability transition (MPT)-driven necrosis (MPTDN) was a regulated variant of cell death triggered by specific stimuli. It played a crucial role in the development of organisms and the pathogenesis of diseases, and may provide new strategies for treating various diseases. However, there was limited research on the mechanisms of MPTDN in cervical cancer (CESC) at present. In this study, Weighted Gene Co-expression Network Analysis (WGCNA) was performed on differentially expressed genes in CESC. The module MEyellow, which showed the highest correlation with the phenotype, was selected for in-depth analysis. It was found that the genes in the MEyellow module may be associated with the tumor immune microenvironment (TIME). Through COX univariate regression and LASSO regression analysis, 6 key genes were identified. These genes were further investigated from multiple perspectives, including their independent diagnostic value, prognostic value, specific regulatory mechanisms in the tumor immune microenvironment, drug sensitivity analysis, and somatic mutation analysis. This study provided a comprehensive exploration of the mechanisms of action of these 6 key genes in CESC patients. And qRT-PCR validation was also conducted. Through COX univariate regression and LASSO coefficient screening of the MEyellow module, 6 key genes were identified: CHRM3-AS2, AC096734.1, BISPR, LINC02446, LINC00944, and DGUOK-AS1. Evaluation of the independent diagnostic value of these 6 key genes revealed that they can serve as independent diagnostic biomarkers. Through correlation analysis among these 6 genes, a potential regulatory mechanism among them was identified. Therefore, a risk prognostic model was established based on the collective action of these 6 genes, and the model showed good performance in predicting the survival period of CESC patients. By studying the relationship between these 6 key genes and the tumor microenvironment of CESC patients from multiple angles, it was found that these 6 genes are key regulatory factors in the tumor immune microenvironment of CESC patients. Additionally, 16 drugs that are associated with these 6 key genes were identified, and 8 small molecule drugs were predicted based on the lncRNA-mRNA network. The 6 key genes can serve as independent biomarkers for diagnosis, and the Risk score of these genes when acting together can be used as an indicator for predicting the clinical survival period of CESC patients. Additionally, these 6 key genes were closely related to the tumor immune microenvironment of CESC patients and were the important regulatory factors in the tumor immune microenvironment of CESC patients.
Collapse
Affiliation(s)
- Chen Yi
- Department of Biomedical Engineering, Nanchang Hang Kong University, Nanchang, 330063, Jiangxi, China
| | - Jun Yang
- Department of Biomedical Engineering, Nanchang Hang Kong University, Nanchang, 330063, Jiangxi, China
| | - Ting Zhang
- Department of Biomedical Engineering, Nanchang Hang Kong University, Nanchang, 330063, Jiangxi, China
| | - Zilu Xie
- Department of Biomedical Engineering, Nanchang Hang Kong University, Nanchang, 330063, Jiangxi, China
| | - Qiliang Xiong
- Department of Biomedical Engineering, Nanchang Hang Kong University, Nanchang, 330063, Jiangxi, China
| | - Dongjuan Chen
- Department of Laboratory Medicine, Maternal and Child Health Hospital of Hubei Province, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430070, China.
| | - Shaofeng Jiang
- Department of Biomedical Engineering, Nanchang Hang Kong University, Nanchang, 330063, Jiangxi, China.
| |
Collapse
|
2
|
Castaneda Ruan P, Benson JC, Trebak M, Kirk V, Sneyd J. A Model for the Coexistence of Competing Mechanisms for Ca 2 + Oscillations in T-lymphocytes. Bull Math Biol 2024; 86:86. [PMID: 38869652 PMCID: PMC11176111 DOI: 10.1007/s11538-024-01317-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 05/29/2024] [Indexed: 06/14/2024]
Abstract
Ca 2 + is a ubiquitous signaling mechanism across different cell types. In T-cells, it is associated with cytokine production and immune function. Benson et al. have shown the coexistence of competing Ca 2 + oscillations during antigen stimulation of T-cell receptors, depending on the presence of extracellular Ca 2 + influx through the Ca 2 + release-activated Ca 2 + channel (Benson in J Biol Chem 29:105310, 2023). In this paper, we construct a mathematical model consisting of five ordinary differential equations and analyze the relationship between the competing oscillatory mechanisms.. We perform bifurcation analysis on two versions of our model, corresponding to the two oscillatory types, to find the defining characteristics of these two families.
Collapse
Affiliation(s)
- Paco Castaneda Ruan
- Department of Mathematics, University of Auckland, Auckland Central, 1142, Auckland, New Zealand.
| | - J Cory Benson
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, 1526, USA
- Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, 1526, USA
- Graduate Program in Cellular and Molecular Physiology, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania, 17033, USA
| | - Mohamed Trebak
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, 1526, USA
- Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, 1526, USA
| | - Vivien Kirk
- Department of Mathematics, University of Auckland, Auckland Central, 1142, Auckland, New Zealand
| | - James Sneyd
- Department of Mathematics, University of Auckland, Auckland Central, 1142, Auckland, New Zealand
| |
Collapse
|
3
|
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
|
4
|
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
|
5
|
Benson JC, Romito O, Abdelnaby AE, Xin P, Pathak T, Weir SE, Kirk V, Castaneda F, Yoast RE, Emrich SM, Tang PW, Yule DI, Hempel N, Potier-Cartereau M, Sneyd J, Trebak M. A multiple-oscillator mechanism underlies antigen-induced Ca 2+ oscillations in Jurkat T-cells. J Biol Chem 2023; 299:105310. [PMID: 37778728 PMCID: PMC10641176 DOI: 10.1016/j.jbc.2023.105310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Revised: 09/11/2023] [Accepted: 09/20/2023] [Indexed: 10/03/2023] Open
Abstract
T-cell receptor stimulation triggers cytosolic Ca2+ signaling by inositol-1,4,5-trisphosphate (IP3)-mediated Ca2+ release from the endoplasmic reticulum (ER) and Ca2+ entry through Ca2+ release-activated Ca2+ (CRAC) channels gated by ER-located stromal-interacting molecules (STIM1/2). Physiologically, cytosolic Ca2+ signaling manifests as regenerative Ca2+ oscillations, which are critical for nuclear factor of activated T-cells-mediated transcription. In most cells, Ca2+ oscillations are thought to originate from IP3 receptor-mediated Ca2+ release, with CRAC channels indirectly sustaining them through ER refilling. Here, experimental and computational evidence support a multiple-oscillator mechanism in Jurkat T-cells whereby both IP3 receptor and CRAC channel activities oscillate and directly fuel antigen-evoked Ca2+ oscillations, with the CRAC channel being the major contributor. KO of either STIM1 or STIM2 significantly reduces CRAC channel activity. As such, STIM1 and STIM2 synergize for optimal Ca2+ oscillations and activation of nuclear factor of activated T-cells 1 and are essential for ER refilling. The loss of both STIM proteins abrogates CRAC channel activity, drastically reduces ER Ca2+ content, severely hampers cell proliferation and enhances cell death. These results clarify the mechanism and the contribution of STIM proteins to Ca2+ oscillations in T-cells.
Collapse
Affiliation(s)
- J Cory Benson
- 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; Graduate Program in Cellular and Molecular Physiology, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania, USA
| | - Olivier Romito
- 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; Inserm UMR 1069, Nutrition Croissance Cancer, Faculté de Médecine, Université de Tours, Tours, France
| | - Ahmed Emam Abdelnaby
- 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
| | - Ping Xin
- 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
| | - Trayambak Pathak
- 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
| | - Sierra E Weir
- 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
| | - Vivien Kirk
- Department of Mathematics, University of Auckland, Auckland, New Zealand
| | | | - Ryan E Yoast
- Graduate Program in Cellular and Molecular Physiology, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania, USA
| | - Scott M Emrich
- Graduate Program in Cellular and Molecular Physiology, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania, USA
| | - Priscilla W Tang
- Division of Hematology/Oncology, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA; UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - David I Yule
- Department of Pharmacology and Physiology, University of Rochester, Rochester, New York, USA
| | - Nadine Hempel
- Division of Hematology/Oncology, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA; UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Marie Potier-Cartereau
- Inserm UMR 1069, Nutrition Croissance Cancer, Faculté de Médecine, Université de Tours, Tours, France
| | - James Sneyd
- Department of Mathematics, University of Auckland, Auckland, New Zealand
| | - Mohamed Trebak
- 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; UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.
| |
Collapse
|
6
|
Howard E, Hurrell BP, Helou DG, Shafiei-Jahani P, Hasiakos S, Painter J, Srikanth S, Gwack Y, Akbari O. Orai inhibition modulates pulmonary ILC2 metabolism and alleviates airway hyperreactivity in murine and humanized models. Nat Commun 2023; 14:5989. [PMID: 37752127 PMCID: PMC10522697 DOI: 10.1038/s41467-023-41065-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 08/22/2023] [Indexed: 09/28/2023] Open
Abstract
Ca2+ entry via Ca2+ release-activated Ca2+ (CRAC) channels is a predominant mechanism of intracellular Ca2+ elevation in immune cells. Here we show the immunoregulatory role of CRAC channel components Orai1 and Orai2 in Group 2 innate lymphoid cells (ILC2s), that play crucial roles in the induction of type 2 inflammation. We find that blocking or genetic ablation of Orai1 and Orai2 downregulates ILC2 effector function and cytokine production, consequently ameliorating the development of ILC2-mediated airway inflammation in multiple murine models. Mechanistically, ILC2 metabolic and mitochondrial homeostasis are inhibited and lead to the upregulation of reactive oxygen species production. We confirm our findings in human ILC2s, as blocking Orai1 and Orai2 prevents the development of airway hyperreactivity in humanized mice. Our findings have a broad impact on the basic understanding of Ca2+ signaling in ILC2 biology, providing potential insights into the development of therapies for the treatment of allergic and atopic inflammatory diseases.
Collapse
Affiliation(s)
- Emily Howard
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Benjamin P Hurrell
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Doumet Georges Helou
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Pedram Shafiei-Jahani
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Spyridon Hasiakos
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Jacob Painter
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Sonal Srikanth
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Yousang Gwack
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Omid Akbari
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
7
|
Lu S, Sun X, Zhou Z, Tang H, Xiao R, Lv Q, Wang B, Qu J, Yu J, Sun F, Deng Z, Tian Y, Li C, Yang Z, Yang P, Rao B. Mechanism of Bazhen decoction in the treatment of colorectal cancer based on network pharmacology, molecular docking, and experimental validation. Front Immunol 2023; 14:1235575. [PMID: 37799727 PMCID: PMC10548240 DOI: 10.3389/fimmu.2023.1235575] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 08/31/2023] [Indexed: 10/07/2023] Open
Abstract
Objective Bazhen Decoction (BZD) is a common adjuvant therapy drug for colorectal cancer (CRC), although its anti-tumor mechanism is unknown. This study aims to explore the core components, key targets, and potential mechanisms of BZD treatment for CRC. Methods The Traditional Chinese Medicine Systems Pharmacology (TCMSP) was employed to acquire the BZD's active ingredient and targets. Meanwhile, the Drugbank, Therapeutic Target Database (TTD), DisGeNET, and GeneCards databases were used to retrieve pertinent targets for CRC. The Venn plot was used to obtain intersection targets. Cytoscape software was used to construct an "herb-ingredient-target" network and identify core targets. GO and KEGG pathway enrichment analyses were conducted using R language software. Molecular docking of key ingredients and core targets of drugs was accomplished using PyMol and Autodock Vina software. Cell and animal research confirmed Bazhen Decoction efficacy and mechanism in treating colorectal cancer. Results BZD comprises 173 effective active ingredients. Using four databases, 761 targets related to CRC were identified. The intersection of BZD and CRC yielded 98 targets, which were utilized to construct the "herb-ingredient-target" network. The four key effector components with the most targets were quercetin, kaempferol, licochalcone A, and naringenin. Protein-protein interaction (PPI) analysis revealed that the core targets of BZD in treating CRC were AKT1, MYC, CASP3, ESR1, EGFR, HIF-1A, VEGFR, JUN, INS, and STAT3. The findings from molecular docking suggest that the core ingredient exhibits favorable binding potential with the core target. Furthermore, the GO and KEGG enrichment analysis demonstrates that BZD can modulate multiple signaling pathways related to CRC, like the T cell receptor, PI3K-Akt, apoptosis, P53, and VEGF signaling pathway. In vitro, studies have shown that BZD dose-dependently inhibits colon cancer cell growth and invasion and promotes apoptosis. Animal experiments have shown that BZD treatment can reverse abnormal expression of PI3K, AKT, MYC, EGFR, HIF-1A, VEGFR, JUN, STAT3, CASP3, and TP53 genes. BZD also increases the ratio of CD4+ T cells to CD8+ T cells in the spleen and tumor tissues, boosting IFN-γ expression, essential for anti-tumor immunity. Furthermore, BZD has the potential to downregulate the PD-1 expression on T cell surfaces, indicating its ability to effectively restore T cell function by inhibiting immune checkpoints. The results of HE staining suggest that BZD exhibits favorable safety profiles. Conclusion BZD treats CRC through multiple components, targets, and metabolic pathways. BZD can reverse the abnormal expression of genes such as PI3K, AKT, MYC, EGFR, HIF-1A, VEGFR, JUN, STAT3, CASP3, and TP53, and suppresses the progression of colorectal cancer by regulating signaling pathways such as PI3K-AKT, P53, and VEGF. Furthermore, BZD can increase the number of T cells and promote T cell activation in tumor-bearing mice, enhancing the immune function against colorectal cancer. Among them, quercetin, kaempferol, licochalcone A, naringenin, and formaronetin are more highly predictive components related to the T cell activation in colorectal cancer mice. This study is of great significance for the development of novel anti-cancer drugs. It highlights the importance of network pharmacology-based approaches in studying complex traditional Chinese medicine formulations.
Collapse
Affiliation(s)
- Shuai Lu
- Key Laboratory of Cancer Foods for Special Medical Purpose (FSMP) for State Market Regulation, Department of Gastrointestinal Surgery/Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical University, Beijing International Science and Technology Cooperation Base for Cancer Metabolism and Nutrition, Beijing, China
| | - Xibo Sun
- Key Laboratory of Cancer Foods for Special Medical Purpose (FSMP) for State Market Regulation, Department of Gastrointestinal Surgery/Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical University, Beijing International Science and Technology Cooperation Base for Cancer Metabolism and Nutrition, Beijing, China
- Department of Breast Surgery, The Second Affiliated Hospital of Shandong First Medical University, Shandong, China
| | - Zhongbao Zhou
- Department of Urology, Beijing TianTan Hospital, Capital Medical University, Beijing, China
| | - Huazhen Tang
- Key Laboratory of Cancer Foods for Special Medical Purpose (FSMP) for State Market Regulation, Department of Gastrointestinal Surgery/Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical University, Beijing International Science and Technology Cooperation Base for Cancer Metabolism and Nutrition, Beijing, China
| | - Ruixue Xiao
- Key Laboratory of Molecular Pathology, Inner Mongolia Medical University, Hohhot, China
| | - Qingchen Lv
- Medical Laboratory College, Hebei North University, Zhangjiakou, China
| | - Bing Wang
- Key Laboratory of Cancer Foods for Special Medical Purpose (FSMP) for State Market Regulation, Department of Gastrointestinal Surgery/Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical University, Beijing International Science and Technology Cooperation Base for Cancer Metabolism and Nutrition, Beijing, China
| | - Jinxiu Qu
- Key Laboratory of Cancer Foods for Special Medical Purpose (FSMP) for State Market Regulation, Department of Gastrointestinal Surgery/Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical University, Beijing International Science and Technology Cooperation Base for Cancer Metabolism and Nutrition, Beijing, China
| | - Jinxuan Yu
- First Clinical Medical College, Binzhou Medical University, Yantai, China
| | - Fang Sun
- Institute of Hepatobiliary Surgery, The First Medical Center of Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Zhuoya Deng
- Institute of Hepatobiliary Surgery, The First Medical Center of Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Yuying Tian
- Key Laboratory of Molecular Pathology, Inner Mongolia Medical University, Hohhot, China
| | - Cong Li
- Key Laboratory of Molecular Pathology, Inner Mongolia Medical University, Hohhot, China
| | - Zhenpeng Yang
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, China
| | - Penghui Yang
- Institute of Hepatobiliary Surgery, The First Medical Center of Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Benqiang Rao
- Key Laboratory of Cancer Foods for Special Medical Purpose (FSMP) for State Market Regulation, Department of Gastrointestinal Surgery/Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical University, Beijing International Science and Technology Cooperation Base for Cancer Metabolism and Nutrition, Beijing, China
| |
Collapse
|
8
|
Benson JC, Trebak M. Too much of a good thing: The case of SOCE in cellular apoptosis. Cell Calcium 2023; 111:102716. [PMID: 36931194 PMCID: PMC10481469 DOI: 10.1016/j.ceca.2023.102716] [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: 02/22/2023] [Revised: 03/06/2023] [Accepted: 03/09/2023] [Indexed: 03/13/2023]
Abstract
Intracellular calcium (Ca2+) is an essential second messenger in eukaryotic cells regulating numerous cellular functions such as contraction, secretion, immunity, growth, and metabolism. Ca2+ signaling is also a key signal transducer in the intrinsic apoptosis pathway. The store-operated Ca2+ entry pathway (SOCE) is ubiquitously expressed in eukaryotic cells, and is the primary Ca2+ influx pathway in non-excitable cells. SOCE is mediated by the endoplasmic reticulum Ca2+ sensing STIM proteins, and the plasma membrane Ca2+-selective Orai channels. A growing number of studies have implicated SOCE in regulating cell death primarily via the intrinsic apoptotic pathway in a variety of tissues and in response to physiological stressors such as traumatic brain injury, ischemia reperfusion injury, sepsis, and alcohol toxicity. Notably, the literature points to excessive cytosolic Ca2+ influx through SOCE in vulnerable cells as a key factor tipping the balance towards cellular apoptosis. While the literature primarily addresses the functions of STIM1 and Orai1, STIM2, Orai2 and Orai3 are also emerging as potential regulators of cell death. Here, we review the functions of STIM and Orai proteins in regulating cell death and the implications of this regulation to human pathologies.
Collapse
Affiliation(s)
- J Cory Benson
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, 200 Lothrop Street, Pittsburgh, PA 1526, USA; Vascular Medicine Institute, University of Pittsburgh School of Medicine, 200 Lothrop Street, Pittsburgh, PA 1526, USA; Department of Cellular and Molecular Physiology, Graduate Program, The Pennsylvania State University College of Medicine, 500 University Drive, Hershey, PA 17033, USA
| | - Mohamed Trebak
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, 200 Lothrop Street, Pittsburgh, PA 1526, USA; Vascular Medicine Institute, University of Pittsburgh School of Medicine, 200 Lothrop Street, Pittsburgh, PA 1526, USA; UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, 200 Lothrop Street, Pittsburgh, PA 1526, USA.
| |
Collapse
|
9
|
Goriounova AS, Gilmore RC, Wrennall JA, Tarran R. Super resolution microscopy analysis reveals increased Orai1 activity in asthma and cystic fibrosis lungs. J Cyst Fibros 2023; 22:161-171. [PMID: 35961837 PMCID: PMC9982747 DOI: 10.1016/j.jcf.2022.07.003] [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: 03/09/2022] [Revised: 07/01/2022] [Accepted: 07/04/2022] [Indexed: 11/28/2022]
Abstract
QUESTION In diseases such as asthma and cystic fibrosis (CF), the immune response is dysregulated and the lung is chronically inflamed. Orai1 activation is required for the initiation and persistence of inflammation. However, Orai1 expression in the lung is poorly understood. We therefore tested the hypothesis that Orai1 expression was upregulated in asthmatic and CF lungs. MATERIALS AND METHODS We used LungMAP to analyze single-cell RNAseq data of Orai1 and stromal interaction molecule 1 (STIM1) expression in normal human lungs. We then performed RNAscope analysis and immunostaining on lung sections from normal, asthma, and CF donors. We imaged sections by confocal and super resolution microscopy, and analyzed Orai1 and STIM1 expression in different pulmonary cell types. RESULTS Orai1 was broadly-expressed, but expression was greatest in immune cells. At mRNA and protein levels, there were no consistent trends in expression levels between the three phenotypes. Orai1 must interact with STIM1 in order to activate and conduct Ca2+. We therefore used STIM1/Orai1 co-localization as a marker of Orai1 activity. Using this approach, we found significantly increased co-localization between these proteins in epithelia, interstitial and luminal immune cells, but not alveoli, from asthma and CF lungs. Orai1 also aggregates as part of its activation process. Using super resolution microscopy, we also found significantly increased Orai1 aggregation in immune cells from asthmatic and CF lungs. CONCLUSION We found evidence that Orai1 was more active in asthma and CF than normal lungs. These data suggest that Orai1 is a relevant target for reducing pulmonary inflammation.
Collapse
Affiliation(s)
| | | | - Joe A Wrennall
- Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, NC, 27599, USA
| | - Robert Tarran
- Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, NC, 27599, USA.
| |
Collapse
|
10
|
Tiffner A, Hopl V, Derler I. CRAC and SK Channels: Their Molecular Mechanisms Associated with Cancer Cell Development. Cancers (Basel) 2022; 15:101. [PMID: 36612099 PMCID: PMC9817886 DOI: 10.3390/cancers15010101] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 12/16/2022] [Accepted: 12/19/2022] [Indexed: 12/28/2022] Open
Abstract
Cancer represents a major health burden worldwide. Several molecular targets have been discovered alongside treatments with positive clinical outcomes. However, the reoccurrence of cancer due to therapy resistance remains the primary cause of mortality. Endeavors in pinpointing new markers as molecular targets in cancer therapy are highly desired. The significance of the co-regulation of Ca2+-permeating and Ca2+-regulated ion channels in cancer cell development, proliferation, and migration make them promising molecular targets in cancer therapy. In particular, the co-regulation of the Orai1 and SK3 channels has been well-studied in breast and colon cancer cells, where it finally leads to an invasion-metastasis cascade. Nevertheless, many questions remain unanswered, such as which key molecular components determine and regulate their interplay. To provide a solid foundation for a better understanding of this ion channel co-regulation in cancer, we first shed light on the physiological role of Ca2+ and how this ion is linked to carcinogenesis. Then, we highlight the structure/function relationship of Orai1 and SK3, both individually and in concert, their role in the development of different types of cancer, and aspects that are not yet known in this context.
Collapse
Affiliation(s)
- Adéla Tiffner
- 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
|
11
|
Orai1 overexpression improves sepsis-induced T-lymphocyte immunosuppression and acute organ dysfunction in mice. Heliyon 2022; 8:e12082. [PMID: 36568656 PMCID: PMC9768300 DOI: 10.1016/j.heliyon.2022.e12082] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 08/20/2022] [Accepted: 11/25/2022] [Indexed: 12/12/2022] Open
Abstract
Immune paralysis induced by sepsis, especially dysfunction of CD4+ T cells, leads to an increased risk of infection. In sepsis, abnormal differentiation of T lymphocytes is associated with multiorgan dysfunction syndrome. In T lymphocytes, the Orai1/nuclear factor of activated T Cells (NFAT) pathway is a critical mediator of infection, inflammation, and autoimmunity. In this study, we confirmed immunosuppression of splenic CD4+ T cells and abnormal differentiation of T lymphocytes in septic mice. Furthermore, we found that the Orai1/NFAT signaling pathway was inhibited in septic mice; however, the overexpression of Orai1 not only improved immune function of T cells in sepsis but also reduced the mortality and organ damage in septic mice. Moreover, the overexpression of Orai1 could reverse the increases in the numbers of T regulatory and T helper 17 cells in septic mice. These data suggest that the Orai1-mediated NFAT signaling pathway can improve sepsis-induced T-lymphocyte immunosuppression and acute organ dysfunction.
Collapse
|
12
|
Kozak JA. Suppression of Store-operated Calcium Entry Channels and Cytokine Release by Cannabinoids. FUNCTION 2022; 3:zqac044. [PMID: 36168590 PMCID: PMC9508850 DOI: 10.1093/function/zqac044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Revised: 08/05/2022] [Accepted: 08/29/2022] [Indexed: 01/07/2023] Open
Affiliation(s)
- J Ashot Kozak
- Department of Neuroscience, Cell Biology and Physiology, Boonshoft School of Medicine and College of Science and Mathematics, Wright State University, Dayton, OH 45435, USA
| |
Collapse
|
13
|
Nehme A, Ghahramanpouri M, Ahmed I, Golsorkhi M, Thomas N, Munoz K, Abdipour A, Tang X, Wilson SM, Wasnik S, Baylink DJ. Combination therapy of insulin-like growth factor I and BTP-2 markedly improves lipopolysaccharide-induced liver injury in mice. FASEB J 2022; 36:e22444. [PMID: 35839071 DOI: 10.1096/fj.202200227rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 06/14/2022] [Accepted: 06/27/2022] [Indexed: 01/06/2023]
Abstract
Acute liver injury is a common disease without effective therapy in humans. We sought to evaluate a combination therapy of insulin-like growth factor 1 (IGF-I) and BTP-2 in a mouse liver injury model induced by lipopolysaccharide (LPS). We chose this model because LPS is known to increase the expression of the transcription factors related to systemic inflammation (i.e., NFκB, CREB, AP1, IRF 3, and NFAT), which depends on calcium signaling. Notably, these transcription factors all have pleiotropic effects and account for the other observed changes in tissue damage parameters. Additionally, LPS is also known to increase the genes associated with a tissue injury (e.g., NGAL, SOD, caspase 3, and type 1 collagen) and systemic expression of pro-inflammatory cytokines. Finally, LPS compromises vascular integrity. Accordingly, IGF-I was selected because its serum levels were shown to decrease during systemic inflammation. BTP-2 was chosen because it was known to decrease cytosolic calcium, which is increased by LPS. This current study showed that IGF-I, BTP-2, or a combination therapy significantly altered and normalized all of the aforementioned LPS-induced gene changes. Additionally, our therapies reduced the vascular leakage caused by LPS, as evidenced by the Evans blue dye technique. Furthermore, histopathologic studies showed that IGF-I decreased the proportion of hepatocytes with ballooning degeneration. Finally, IGF-I also increased the expression of the hepatic growth factor (HGF) and the receptor for the epidermal growth factor (EGFR), markers of liver regeneration. Collectively, our data suggest that a combination of IGF-I and BTP-2 is a promising therapy for acute liver injury.
Collapse
Affiliation(s)
- Antoine Nehme
- Division of Regenerative Medicine, Department of Medicine, Loma Linda University, Loma Linda, California, USA
| | - Mahdis Ghahramanpouri
- Division of Regenerative Medicine, Department of Medicine, Loma Linda University, Loma Linda, California, USA
| | - Iqbal Ahmed
- Pathology and Laboratory Medicine, Loma Linda University, Loma Linda, California, USA
| | - Mohadese Golsorkhi
- Division of Regenerative Medicine, Department of Medicine, Loma Linda University, Loma Linda, California, USA
| | | | - Kevin Munoz
- Division of Regenerative Medicine, Department of Medicine, Loma Linda University, Loma Linda, California, USA
| | - Amir Abdipour
- Division of Nephrology, Loma Linda University Medical Center, Loma Linda, California, USA
| | - Xiaolei Tang
- Division of Regenerative Medicine, Department of Medicine, Loma Linda University, Loma Linda, California, USA.,Department of Veterinary Biomedical Sciences, College of Veterinary Medicine, Long Island University, Brookville, New York, USA
| | - Sean M Wilson
- The Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California, USA
| | - Samiksha Wasnik
- Division of Regenerative Medicine, Department of Medicine, Loma Linda University, Loma Linda, California, USA
| | - David J Baylink
- Division of Regenerative Medicine, Department of Medicine, Loma Linda University, Loma Linda, California, USA
| |
Collapse
|
14
|
Veytia-Bucheli JI, Alvarado-Velázquez DA, Possani LD, González-Amaro R, Rosenstein Y. The Ca 2+ Channel Blocker Verapamil Inhibits the In Vitro Activation and Function of T Lymphocytes: A 2022 Reappraisal. Pharmaceutics 2022; 14:pharmaceutics14071478. [PMID: 35890372 PMCID: PMC9324055 DOI: 10.3390/pharmaceutics14071478] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Revised: 07/12/2022] [Accepted: 07/12/2022] [Indexed: 02/04/2023] Open
Abstract
Ca2+ channel blockers (CCBs) are commonly used to treat different cardiovascular conditions. These drugs disrupt the intracellular Ca2+ signaling network, inhibiting numerous cellular functions in different cells, including T lymphocytes. We explored the effect of the CCB verapamil on normal human peripheral blood T cell activation, proliferation, and cytokine production. Cells were activated by ligating CD3 or CD3/CD28 in the presence or absence of verapamil, and the expression of activation-induced cell surface molecules (CD25, CD40L, CD69, PD-1, and OX40), cell proliferation, and cytokine release were assessed by flow cytometry. Verapamil exerted a dose-dependent inhibitory effect on the expression of all the activation-induced cell surface molecules tested. In addition, verapamil diminished T cell proliferation induced in response to CD3/CD28 stimulation. Likewise, the production of Th1/Th17 and Th2 cytokines was also reduced by verapamil. Our data substantiate a potent in vitro suppressive effect of verapamil on T lymphocytes, a fact that might be relevant in patients receiving CCBs.
Collapse
Affiliation(s)
- José Ignacio Veytia-Bucheli
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Av. Universidad 2001, Cuernavaca 62210, Mexico; (J.I.V.-B.); (D.A.A.-V.); (L.D.P.)
- Laboratoire de Chimie Bio-Organique, Département de Chimie, Faculté des Sciences, Université de Namur, Rue de Bruxelles 615, 5000 Namur, Belgium
| | - Den Alejandro Alvarado-Velázquez
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Av. Universidad 2001, Cuernavaca 62210, Mexico; (J.I.V.-B.); (D.A.A.-V.); (L.D.P.)
- Posgrado en Ciencias, Instituto de Investigación en Ciencias Básicas y Aplicadas, Universidad Autónoma del Estado de Morelos, Av. Universidad 1001, Cuernavaca 62209, Mexico
| | - Lourival Domingos Possani
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Av. Universidad 2001, Cuernavaca 62210, Mexico; (J.I.V.-B.); (D.A.A.-V.); (L.D.P.)
| | - Roberto González-Amaro
- Centro de Investigación en Ciencias de la Salud y Biomedicina, Universidad Autónoma de San Luis Potosí, Av. Sierra Leona 550, San Luis Potosí 78210, Mexico;
| | - Yvonne Rosenstein
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Av. Universidad 2001, Cuernavaca 62210, Mexico; (J.I.V.-B.); (D.A.A.-V.); (L.D.P.)
- Correspondence: ; Tel.: +52-(777)-329-1606
| |
Collapse
|
15
|
Bieber T, Paller AS, Kabashima K, Feely M, Rueda MJ, Ross Terres JA, Wollenberg A. Atopic dermatitis: pathomechanisms and lessons learned from novel systemic therapeutic options. J Eur Acad Dermatol Venereol 2022; 36:1432-1449. [PMID: 35575442 DOI: 10.1111/jdv.18225] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 04/13/2022] [Indexed: 12/01/2022]
Abstract
Atopic dermatitis (AD) is a chronic, heterogenous, inflammatory skin disorder associated with a high skin-related health burden, typically starting in childhood and often persisting into adulthood. AD is characterized by a wide range of clinical phenotypes, reflecting multiple underlying pathophysiological mechanisms and interactions between genetics, immune system dysregulation, and environmental factors. In this review, we describe the diverse cellular and molecular mechanisms involved in AD, including the critical role of T cell-driven inflammation, primarily via T helper (Th) 2- and Th17-derived cytokines, many of which are mediated by the Janus kinase (JAK) signaling pathway. These local inflammatory processes interact with sensory neuronal pathways, contributing to the clinical manifestations of AD, including itch, pain, and sleep disturbance. The recent elucidation of the molecular pathways involved in AD has allowed treatment strategies to evolve from broad-acting systemic immunosuppressive therapies to more targeted agents, including JAK inhibitors and cytokine-specific biologic agents. Evidence from the clinical development of these targeted therapies has reinforced and expanded our understanding of the pathophysiological mechanisms underlying AD and holds promise for individualized treatment strategies tailored to specific AD subtypes.
Collapse
Affiliation(s)
- T Bieber
- Department of Dermatology and Allergy, University Medical Center, Bonn, Germany.,Christine Kühne-Center for Allergy Research and Education, Davos, Switzerland
| | - A S Paller
- Department of Dermatology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - K Kabashima
- Department Dermatology, Kyoto University School of Medicine, Kyoto, Japan
| | - M Feely
- Eli Lilly and Company, Indianapolis, IN, USA.,Department of Dermatology, Mount Sinai Hospital, New York, NY, USA
| | - M J Rueda
- Eli Lilly and Company, Indianapolis, IN, USA
| | | | - A Wollenberg
- Department of Dermatology and Allergy, University Hospital, Ludwig Maximillian University, Munich, Germany.,Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel (UZ Brussel), Department of Dermatology, Brussels, Belgium
| |
Collapse
|
16
|
Wu B, Woo JS, Sun Z, Srikanth S, Gwack Y. Ca 2+ Signaling Augmented by ORAI1 Trafficking Regulates the Pathogenic State of Effector T Cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 208:1329-1340. [PMID: 35217583 PMCID: PMC8916982 DOI: 10.4049/jimmunol.2100871] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 01/07/2022] [Indexed: 02/02/2023]
Abstract
Activation of the Ca2+ release-activated Ca2+ (CRAC) channel is crucial for T cell functions. It was recently shown that naked cuticle homolog 2 (NKD2), a signaling adaptor molecule, orchestrates trafficking of ORAI1, a pore subunit of the CRAC channels, to the plasma membrane for sustained activation of the CRAC channels. However, the physiological role of sustained Ca2+ entry via ORAI1 trafficking remains poorly understood. Using NKD2 as a molecular handle, we show that ORAI1 trafficking is crucial for sustained Ca2+ entry and cytokine production, especially in inflammatory Th1 and Th17 cells. We find that murine T cells cultured under pathogenic Th17-polarizing conditions have higher Ca2+ levels that are NKD2-dependent than those under nonpathogenic conditions. In vivo, deletion of Nkd2 alleviated clinical symptoms of experimental autoimmune encephalomyelitis in mice by selectively decreasing effector T cell responses in the CNS. Furthermore, we observed a strong correlation between NKD2 expression and proinflammatory cytokine production in effector T cells. Taken together, our findings suggest that the pathogenic effector T cell response demands sustained Ca2+ entry supported by ORAI1 trafficking.
Collapse
Affiliation(s)
- Beibei Wu
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, CA; and
| | - Jin Seok Woo
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, CA; and
| | - Zuoming Sun
- Department of Molecular Imaging and Therapy, Beckman Research Institute of City of Hope, Duarte, CA
| | - Sonal Srikanth
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, CA; and
| | - Yousang Gwack
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, CA; and
| |
Collapse
|
17
|
Shaik FA, Lewuillon C, Guillemette A, Ahmadian B, Brinster C, Quesnel B, Collard D, Touil Y, Lemonnier L, Tarhan MC. Pairing cells of different sizes in a microfluidic device for immunological synapse monitoring. LAB ON A CHIP 2022; 22:908-920. [PMID: 35098952 DOI: 10.1039/d1lc01156a] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Analyzing cell-cell interaction is essential to investigate how immune cells function. Elegant designs have been demonstrated to study lymphocytes and their interaction partners. However, these devices have been targeting cells of similar dimensions. T lymphocytes are smaller, more deformable, and more sensitive to pressure than many cells. This work aims to fill the gap of a method for pairing cells with different dimensions. The developed method uses hydrodynamic flow focusing in the z-direction for on-site modulation of effective channel height to capture smaller cells as single cells. Due to immune cells' sensitivity to pressure, the proposed method provides a stable system without any change in flow conditions at the analysis area throughout experiments. Paired live cells have their activities analyzed with calcium imaging at the immunological synapse formed under a controlled environment. The method is demonstrated with primary human T lymphocytes, acute myeloid leukemia (AML) cell lines, and primary AML blasts.
Collapse
Affiliation(s)
- Faruk Azam Shaik
- University of Lille, Lille, France
- CNRS, IIS, COL, Univ. Lille SMMiL-E project, Lille, France
| | - Clara Lewuillon
- University of Lille, Lille, France
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277 - Canther - Cancer Heterogeneity, Plasticity and Resistance to Therapies, F-59000 Lille, France.
| | - Aurélie Guillemette
- University of Lille, Lille, France
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277 - Canther - Cancer Heterogeneity, Plasticity and Resistance to Therapies, F-59000 Lille, France.
| | - Bahram Ahmadian
- CNRS, IIS, COL, Univ. Lille SMMiL-E project, Lille, France
- Univ. Lille, CNRS, Centrale Lille, Junia, Univ. Polytechnique Hauts-de-France, UMR 8520 -IEMN -Institut d'Electronique de Microélectronique et de Nanotechnologie, F-59000 Lille, France.
| | - Carine Brinster
- University of Lille, Lille, France
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277 - Canther - Cancer Heterogeneity, Plasticity and Resistance to Therapies, F-59000 Lille, France.
| | - Bruno Quesnel
- University of Lille, Lille, France
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277 - Canther - Cancer Heterogeneity, Plasticity and Resistance to Therapies, F-59000 Lille, France.
| | - Dominique Collard
- CNRS, IIS, COL, Univ. Lille SMMiL-E project, Lille, France
- LIMMS/CNRS-IIS IRL2820, The University of Tokyo, Tokyo, Japan
| | - Yasmine Touil
- University of Lille, Lille, France
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277 - Canther - Cancer Heterogeneity, Plasticity and Resistance to Therapies, F-59000 Lille, France.
| | - Loïc Lemonnier
- Univ. Lille, Inserm, U1003 - PHYCEL - Physiologie Cellulaire, F-59000 Lille, France.
- Laboratory of Excellence, Ion Channels Science and Therapeutics, Villeneuve d'Ascq, France
| | - Mehmet Cagatay Tarhan
- CNRS, IIS, COL, Univ. Lille SMMiL-E project, Lille, France
- Univ. Lille, CNRS, Centrale Lille, Junia, Univ. Polytechnique Hauts-de-France, UMR 8520 -IEMN -Institut d'Electronique de Microélectronique et de Nanotechnologie, F-59000 Lille, France.
- LIMMS/CNRS-IIS IRL2820, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
18
|
Wang YY, Wang WC, Su CW, Hsu CW, Yuan SS, Chen YK. Expression of Orai1 and STIM1 in human oral squamous cell carcinogenesis. J Dent Sci 2022; 17:78-88. [PMID: 35028023 PMCID: PMC8739746 DOI: 10.1016/j.jds.2021.07.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 07/07/2021] [Indexed: 12/21/2022] Open
Abstract
Background/purpose Return of Ca2+ to endoplasmic reticulum is mediated by Orai/STIM-mediated store-operated Ca2+ entry (SOCE) channel. We aimed to investigate Orai1 and STIM1 expressions in human oral carcinogenesis. Materials and methods Sixty-six oral squamous cell carcinomas (OSCCs), 14 oral potentially malignant disorders (OPMD) with moderate-severe oral epithelial dysplasia (OED), 19 OPMD with mild OED, and 14 normal oral mucosa (NOM) samples were subjected to immunohistochemical staining. Two human oral cancer cell lines (OCCLs), an oral premalignant cell line (DOK), and a normal oral keratinocyte culture (HOK) were used for Western blot and real-time quantitative reverse transcription-polymerase chain reaction. OCCLs were evaluated for proliferation, migration, and invasion assays. Results Orai1 and STIM1 protein and mRNA expressions in OSCC were significantly enhanced as compared with normal samples. Protein expressions of Orai1 and STIM1 in OCCLs were significantly enhanced as compared with HOK. Significant decreases in degrees of proliferation, migration and invasion were noted in OCCLs with Orai1 and STIM1 siRNA transfection as compared with those without transfection. Significantly increased Orai1 and STIM1 protein levels were noted in OPMD with moderate-severe OED as compared with those with mild OED. Conclusion Our results indicate that Orai1 and STIM1 overexpression is associated with human oral carcinogenesis.
Collapse
Affiliation(s)
- Yen-Yun Wang
- School of Dentistry, College of Dental Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Wen-Chen Wang
- School of Dentistry, College of Dental Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,Division of Oral Pathology & Maxillofacial Radiology, Department of Dentistry, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan.,Oral & Maxillofacial Imaging Center, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chiang-Wei Su
- Division of Oral & Maxillofacial Surgery, Department of Dentistry, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Ching-Wei Hsu
- Division of Oral & Maxillofacial Surgery, Department of Dentistry, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Shyng-Shiou Yuan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan.,Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,Translational Research Center, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Obstetrics and Gynecology and Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yuk-Kwan Chen
- School of Dentistry, College of Dental Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,Division of Oral Pathology & Maxillofacial Radiology, Department of Dentistry, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan.,Oral & Maxillofacial Imaging Center, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
19
|
Wu B, Ramaiah A, Garcia G, Hasiakos S, Arumugaswami V, Srikanth S. ORAI1 Limits SARS-CoV-2 Infection by Regulating Tonic Type I IFN Signaling. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 208:74-84. [PMID: 34819389 PMCID: PMC8702473 DOI: 10.4049/jimmunol.2100742] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 10/27/2021] [Indexed: 01/03/2023]
Abstract
ORAI1 and stromal interaction molecule 1 (STIM1) are the critical mediators of store-operated Ca2+ entry by acting as the pore subunit and an endoplasmic reticulum-resident signaling molecule, respectively. In addition to Ca2+ signaling, STIM1 is also involved in regulation of the type I IFN (IFN-I) response. To examine their potential role in severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, we generated ORAI1 and STIM1 knockout human HEK293-angiotensin-converting enzyme 2 cells and checked their responses. STIM1 knockout cells showed strong resistance to SARS-CoV-2 infection as a result of enhanced IFN-I response. On the contrary, ORAI1 deletion induced high susceptibility to SARS-CoV-2 infection. Mechanistically, ORAI1 knockout cells showed reduced homeostatic cytoplasmic Ca2+ concentration and severe impairment in tonic IFN-I signaling. Transcriptome analysis showed downregulation of multiple antiviral signaling pathways in ORAI1 knockout cells, likely because of reduced expression of the Ca2+-dependent transcription factors of the AP-1 family and MEF2C Accordingly, modulation of homeostatic Ca2+ concentration by pretreatment with ORAI1 blocker or agonist could influence baseline IFNB expression and resistance to SARS-CoV-2 infection in a human lung epithelial cell line. Our results identify a novel role of ORAI1-mediated Ca2+ signaling in regulating the tonic IFN-I levels, which determine host resistance to SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Beibei Wu
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Arunachalam Ramaiah
- Department of Ecology and Evolutionary Biology, University of California, Irvine, CA 92697, USA,Tata Institute for Genetics and Society, Center at inStem, Bangalore, KA, 560065, India
| | - Gustavo Garcia
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Spyridon Hasiakos
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA,Division of Oral Biology and Medicine, UCLA School of Dentistry, Los Angeles, CA, USA
| | - Vaithilingaraja Arumugaswami
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA,Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, CA 90095, USA,California NanoSystems Institute, University of California, Los Angeles, CA 90095, USA,Senior authors,Address correspondence to: Dr. Sonal Srikanth, Department of Physiology, David Geffen School of Medicine, 53-266 CHS, 10833 Le Conte Avenue, Los Angeles, CA 90095, Tel: 310-794-2003; FAX: 310-206-5661, , or, Dr. Vaithilingaraja Arumugaswami, B2-049A CHS, Box 956948, University of California, Los Angeles, CA 90095, Tel: 310 – 794-9568,
| | - Sonal Srikanth
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA,Senior authors,Address correspondence to: Dr. Sonal Srikanth, Department of Physiology, David Geffen School of Medicine, 53-266 CHS, 10833 Le Conte Avenue, Los Angeles, CA 90095, Tel: 310-794-2003; FAX: 310-206-5661, , or, Dr. Vaithilingaraja Arumugaswami, B2-049A CHS, Box 956948, University of California, Los Angeles, CA 90095, Tel: 310 – 794-9568,
| |
Collapse
|
20
|
Plasma Membrane and Organellar Targets of STIM1 for Intracellular Calcium Handling in Health and Neurodegenerative Diseases. Cells 2021; 10:cells10102518. [PMID: 34685498 PMCID: PMC8533710 DOI: 10.3390/cells10102518] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 09/14/2021] [Accepted: 09/21/2021] [Indexed: 01/08/2023] Open
Abstract
Located at the level of the endoplasmic reticulum (ER) membrane, stromal interacting molecule 1 (STIM1) undergoes a complex conformational rearrangement after depletion of ER luminal Ca2+. Then, STIM1 translocates into discrete ER-plasma membrane (PM) junctions where it directly interacts with and activates plasma membrane Orai1 channels to refill ER with Ca2+. Furthermore, Ca2+ entry due to Orai1/STIM1 interaction may induce canonical transient receptor potential channel 1 (TRPC1) translocation to the plasma membrane, where it is activated by STIM1. All these events give rise to store-operated calcium entry (SOCE). Besides the main pathway underlying SOCE, which mainly involves Orai1 and TRPC1 activation, STIM1 modulates many other plasma membrane proteins in order to potentiate the influxof Ca2+. Furthermore, it is now clear that STIM1 may inhibit Ca2+ currents mediated by L-type Ca2+ channels. Interestingly, STIM1 also interacts with some intracellular channels and transporters, including nuclear and lysosomal ionic proteins, thus orchestrating organellar Ca2+ homeostasis. STIM1 and its partners/effectors are significantly modulated in diverse acute and chronic neurodegenerative conditions. This highlights the importance of further disclosing their cellular functions as they might represent promising molecular targets for neuroprotection.
Collapse
|
21
|
Wu B, Woo JS, Vila P, Jew M, Leung J, Sun Z, Srikanth S, Gwack Y. NKD2 mediates stimulation-dependent ORAI1 trafficking to augment Ca 2+ entry in T cells. Cell Rep 2021; 36:109603. [PMID: 34433025 PMCID: PMC8435239 DOI: 10.1016/j.celrep.2021.109603] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 06/21/2021] [Accepted: 08/03/2021] [Indexed: 01/19/2023] Open
Abstract
Sustained activation of the Ca2+-release-activated Ca2+ (CRAC) channel is pivotal for effector T cell responses. The mechanisms underlying this sustainability remain poorly understood. We find that plasma membrane localization of ORAI1, the pore subunit of CRAC channels, is limited in effector T cells, with a significant fraction trapped in intracellular vesicles. From a targeted screen, we identify an essential component of ORAI1+ vesicles, naked cuticle homolog 2 (NKD2). Mechanistically, NKD2, an adaptor molecule activated by signaling pathways downstream of T cell receptors, orchestrates trafficking and insertion of ORAI1+ vesicles to the plasma membrane. Together, our findings suggest that T cell receptor (TCR)-stimulation-dependent insertion of ORAI1 into the plasma membrane is essential for sustained Ca2+ signaling and cytokine production in T cells.
Collapse
Affiliation(s)
- Beibei Wu
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles CA 90095, USA
| | - Jin Seok Woo
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles CA 90095, USA
| | - Pamela Vila
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles CA 90095, USA,Present address: Olive View-UCLA Medical Center, 14445 Olive View Drive, Sylmar, CA 91342, USA
| | - Marcus Jew
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles CA 90095, USA,Present address: Ronald Reagan UCLA Medical Center, 757 Westwood Plaza, Los Angeles, CA 90095, USA
| | - Jennifer Leung
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles CA 90095, USA,Present address: Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Zuoming Sun
- Department of Molecular Imaging & Therapy, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - Sonal Srikanth
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles CA 90095, USA.
| | - Yousang Gwack
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles CA 90095, USA.
| |
Collapse
|
22
|
Li Z, Trakooljul N, Hadlich F, Ponsuksili S, Wimmers K, Murani E. Transcriptome analysis of porcine PBMCs reveals lipopolysaccharide-induced immunomodulatory responses and crosstalk of immune and glucocorticoid receptor signaling. Virulence 2021; 12:1808-1824. [PMID: 34288827 PMCID: PMC8296968 DOI: 10.1080/21505594.2021.1948276] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
Abstract
The current level of knowledge on transcriptome responses triggered by endotoxins and glucocorticoids in immune cells in pigs is limited. Therefore, in the present study, we treated porcine peripheral blood mononuclear cells (PBMCs) with lipopolysaccharide (LPS) and dexamethasone (DEX) separately or combined for 2 hours. The resultant transcriptional responses were examined by mRNA sequencing. We found that the LPS treatment triggered pronounced inflammatory responses as evidenced by upregulation of pro-inflammatory cytokines, chemokines, and related signaling pathways like NF-κB. Concurrently, a series of downregulated pro-inflammatory and upregulated anti-inflammatory molecules were identified. These are involved in the inhibition of TLR, NF-κB, and MAPK cascades and activation of signaling mediated by Tregs and STAT3, respectively. These findings suggested that LPS initiated also an anti-inflammatory process to prevent an overwhelming inflammatory response. The transcriptome responses further revealed substantial crosstalk of immune responses and glucocorticoid receptor (GR) signaling. This was apparent in four aspects: constitutive inhibition of T cell signaling by DEX through a subset of genes showing no response to LPS; inhibition of LPS-induced inflammatory genes by DEX; attenuation of DEX action by LPS paralleled by the regulation of genes implicated in cytokine and calcium signaling; and DEX-induced changes in genes associated with the activation of pro-inflammatory TLR, NF-κB, iNOS, and IL-1 signaling. Consequently, our study provides novel insights into inflammatory and GR signaling in pigs, as well as an understanding of the application of glucocorticoid drugs for the treatment of inflammatory disorders.
Collapse
Affiliation(s)
- Zhiwei Li
- Institute of Genome Biology, Leibniz Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
| | - Nares Trakooljul
- Institute of Genome Biology, Leibniz Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
| | - Frieder Hadlich
- Institute of Genome Biology, Leibniz Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
| | - Siriluck Ponsuksili
- Institute of Genome Biology, Leibniz Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
| | - Klaus Wimmers
- Institute of Genome Biology, Leibniz Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
| | - Eduard Murani
- Institute of Genome Biology, Leibniz Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
| |
Collapse
|
23
|
Wu B, Ramaiah A, Garcia G, Gwack Y, Arumugaswami V, Srikanth S. ORAI1 establishes resistance to SARS-CoV-2 infection by regulating tonic type I interferon signaling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2021. [PMID: 33972946 PMCID: PMC8109209 DOI: 10.1101/2021.05.04.442548] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
ORAI1 and STIM1 are the critical mediators of store-operated Ca2+ entry by acting as the pore subunit and an endoplasmic reticulum-resident signaling molecule, respectively. In addition to Ca2+ signaling, STIM1 is also involved in regulation of a cytosolic nucleic acid sensing pathway. Using ORAI1 and STIM1 knockout cells, we examined their contribution to the host response to SARS-CoV-2 infection. STIM1 knockout cells showed strong resistance to SARS-CoV-2 infection due to enhanced type I interferon response. On the contrary, ORAI1 knockout cells showed high susceptibility to SARS-CoV-2 infection as judged by increased expression of viral proteins and a high viral load. Mechanistically, ORAI1 knockout cells showed reduced homeostatic cytoplasmic Ca2+ concentration and severe impairment in tonic interferon signaling. Transcriptome analysis showed downregulation of multiple cellular defense mechanisms, including antiviral signaling pathways in ORAI1 knockout cells, which are likely due to reduced expression of the Ca2+-dependent transcription factors of the activator protein 1 (AP-1) family and MEF2C. Our results identify a novel role of ORAI1-mediated Ca2+ signaling in regulating the baseline type I interferon level, which is a determinant of host resistance to SARS-CoV-2 infection.
Collapse
|
24
|
Marongiu L, Mingozzi F, Cigni C, Marzi R, Di Gioia M, Garrè M, Parazzoli D, Sironi L, Collini M, Sakaguchi R, Morii T, Crosti M, Moro M, Schurmans S, Catelani T, Rotem R, Colombo M, Shears S, Prosperi D, Zanoni I, Granucci F. Inositol 1,4,5-trisphosphate 3-kinase B promotes Ca 2+ mobilization and the inflammatory activity of dendritic cells. Sci Signal 2021; 14:14/676/eaaz2120. [PMID: 33785611 DOI: 10.1126/scisignal.aaz2120] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Innate immune responses to Gram-negative bacteria depend on the recognition of lipopolysaccharide (LPS) by a receptor complex that includes CD14 and TLR4. In dendritic cells (DCs), CD14 enhances the activation not only of TLR4 but also that of the NFAT family of transcription factors, which suppresses cell survival and promotes the production of inflammatory mediators. NFAT activation requires Ca2+ mobilization. In DCs, Ca2+ mobilization in response to LPS depends on phospholipase C γ2 (PLCγ2), which produces inositol 1,4,5-trisphosphate (IP3). Here, we showed that the IP3 receptor 3 (IP3R3) and ITPKB, a kinase that converts IP3 to inositol 1,3,4,5-tetrakisphosphate (IP4), were both necessary for Ca2+ mobilization and NFAT activation in mouse and human DCs. A pool of IP3R3 was located on the plasma membrane of DCs, where it colocalized with CD14 and ITPKB. Upon LPS binding to CD14, ITPKB was required for Ca2+ mobilization through plasma membrane-localized IP3R3 and for NFAT nuclear translocation. Pharmacological inhibition of ITPKB in mice reduced both LPS-induced tissue swelling and the severity of inflammatory arthritis to a similar extent as that induced by the inhibition of NFAT using nanoparticles that delivered an NFAT-inhibiting peptide specifically to phagocytic cells. Our results suggest that ITPKB may represent a promising target for anti-inflammatory therapies that aim to inhibit specific DC functions.
Collapse
Affiliation(s)
- Laura Marongiu
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126 Milan, Italy
| | - Francesca Mingozzi
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126 Milan, Italy
| | - Clara Cigni
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126 Milan, Italy
| | - Roberta Marzi
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126 Milan, Italy
| | - Marco Di Gioia
- Harvard Medical School and Division of Immunology, Division of Gastroenterology, Boston Children's Hospital, Boston, MA 02115, USA
| | | | | | - Laura Sironi
- Department of Physics, University of Milano-Bicocca, Piazza della Scienza 3, 20126 Milan, Italy
| | - Maddalena Collini
- Department of Physics, University of Milano-Bicocca, Piazza della Scienza 3, 20126 Milan, Italy
| | - Reiko Sakaguchi
- Institute for Integrated Cell-Material Sciences, Kyoto University Katsura, Nishikyo-ku, Kyoto 615-8510, Japan
| | - Takashi Morii
- Institute of Advanced Energy, Kyoto University, Uji, Kyoto 611-0011, Japan
| | - Mariacristina Crosti
- INGM, Istituto Nazionale di Genetica Molecolare "Romeo ed Enrica Invernizzi", 20122 Milan, Italy
| | - Monica Moro
- INGM, Istituto Nazionale di Genetica Molecolare "Romeo ed Enrica Invernizzi", 20122 Milan, Italy
| | - Stéphane Schurmans
- Laboratory of Functional Genetics, GIGA-B34, University of Liège, 4000 Liège, Belgium
| | - Tiziano Catelani
- Piattaforma Interdipartimentale di Microscopia, University of Milano-Bicocca, Piazza della Scienza 3, 20126 Milan, Italy
| | - Rany Rotem
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126 Milan, Italy
| | - Miriam Colombo
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126 Milan, Italy
| | - Stephen Shears
- Signal Transduction Laboratory, NIEHS/NIH, 111 TW Alexander Drive, Research Triangle Park, NC 27709, USA
| | - Davide Prosperi
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126 Milan, Italy
| | - Ivan Zanoni
- Harvard Medical School and Division of Immunology, Division of Gastroenterology, Boston Children's Hospital, Boston, MA 02115, USA.,Division of Immunology, Harvard Medical School, Boston Children's Hospital, Boston, MA 02115, USA
| | - Francesca Granucci
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126 Milan, Italy. .,INGM, Istituto Nazionale di Genetica Molecolare "Romeo ed Enrica Invernizzi", 20122 Milan, Italy
| |
Collapse
|
25
|
Srikanth S, Gwack Y. Blue light opens the ORAI1 LOC(K). Cell Calcium 2021; 95:102371. [PMID: 33626404 DOI: 10.1016/j.ceca.2021.102371] [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: 02/09/2021] [Accepted: 02/10/2021] [Indexed: 11/19/2022]
Abstract
The current optogenetic tools to elevate cytoplasmic Ca2+ concentrations either do not have high ion selectivity or activate undesirable signaling pathways. He et al. (2021) generated a light-operated Ca2+ channel by integrating a photosensitive module directly into ORAI1 channels to selectively raise cytoplasmic Ca2+ levels.
Collapse
Affiliation(s)
- Sonal Srikanth
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA.
| | - Yousang Gwack
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA.
| |
Collapse
|
26
|
Hasiakos S, Gwack Y, Kang M, Nishimura I. Calcium Signaling in T Cells and Chronic Inflammatory Disorders of the Oral Cavity. J Dent Res 2021; 100:693-699. [PMID: 33541200 DOI: 10.1177/0022034521990652] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Acute immune responses to microbial insults in the oral cavity often progress to chronic inflammatory diseases such as periodontitis and apical periodontitis. Chronic oral inflammation causes destruction of the periodontium, potentially leading to loss of the dentition. Previous investigations have demonstrated that the composition of oral immune cells, rather than the overall extent of cellular infiltration, determines the pathological development of chronic inflammation. The role of T lymphocyte populations, including Th1, Th2, Th17, and Treg cells, has been extensively described. Studies now propose pathogenic Th17 cells as a distinct subset, uniquely classifiable from traditional Th17 populations. In situ differentiation of pathogenic Th17 cells has been verified as a source of destructive inflammation, which critically drives pathogenesis in chronic inflammatory diseases such as diabetes, rheumatoid arthritis, and inflammatory bowel disease. Pathogenic Th17 cells resemble a Th1 penotype and produce not only interleukin 17 (IL-17) but also γ-interferon (IFN-γ) and granulocyte-macrophage colony-stimulating factor (GM-CSF). The proinflammatory cytokine-specific mechanisms known to induce IL-17 expression in Th17 cells are well characterized; however, differentiation mechanisms that lead to pathogenic Th17 cells are less understood. Recently, Ca2+ signaling through Ca2+ release-activated Ca2+ channels (CRAC) in T cells has been uncovered as a major signaling axis involved in the regulation of T-cell-mediated chronic inflammation. In particular, pathogenic Th17 cell-mediated immunological diseases appear to be effectively targeted via such Ca2+ signaling pathways. Pathogenic plasticity of Th17 cells has been extensively illustrated in autoimmune and chronic inflammatory diseases. Although their specific causal relationship to oral infection-induced chronic inflammatory diseases is not fully established, pathogenic Th17 cells may be involved in the underlining mechanism. This review highlights the current understanding of T-cell phenotype regulation, calcium signaling pathways in this event, and the potential role of pathogenic Th17 cells in chronic inflammatory disorders of the oral cavity.
Collapse
Affiliation(s)
- S Hasiakos
- Division of Oral Biology and Medicine, UCLA School of Dentistry, Los Angeles, CA, USA.,Department of Physiology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Y Gwack
- Department of Physiology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - M Kang
- Section of Endodontics, UCLA School of Dentistry, Los Angeles, CA, USA
| | - I Nishimura
- Division of Oral Biology and Medicine, UCLA School of Dentistry, Los Angeles, CA, USA.,Weintraub Center for Reconstructive Biotechnology, UCLA School of Dentistry, Los Angeles, CA, USA
| |
Collapse
|
27
|
Tiffner A, Derler I. Molecular Choreography and Structure of Ca 2+ Release-Activated Ca 2+ (CRAC) and K Ca2+ Channels and Their Relevance in Disease with Special Focus on Cancer. MEMBRANES 2020; 10:E425. [PMID: 33333945 PMCID: PMC7765462 DOI: 10.3390/membranes10120425] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 12/04/2020] [Accepted: 12/07/2020] [Indexed: 12/16/2022]
Abstract
Ca2+ ions play a variety of roles in the human body as well as within a single cell. Cellular Ca2+ signal transduction processes are governed by Ca2+ sensing and Ca2+ transporting proteins. In this review, we discuss the Ca2+ and the Ca2+-sensing ion channels with particular focus on the structure-function relationship of the Ca2+ release-activated Ca2+ (CRAC) ion channel, the Ca2+-activated K+ (KCa2+) ion channels, and their modulation via other cellular components. Moreover, we highlight their roles in healthy signaling processes as well as in disease with a special focus on cancer. As KCa2+ channels are activated via elevations of intracellular Ca2+ levels, we summarize the current knowledge on the action mechanisms of the interplay of CRAC and KCa2+ ion channels and their role in cancer cell development.
Collapse
Affiliation(s)
| | - Isabella Derler
- Institute of Biophysics, JKU Life Science Center, Johannes Kepler University Linz, A-4020 Linz, Austria;
| |
Collapse
|
28
|
Shrestha S, Cho W, Stump B, Imani J, Lamattina AM, Louis PH, Pazzanese J, Rosas IO, Visner G, Perrella MA, El-Chemaly S. FK506 induces lung lymphatic endothelial cell senescence and downregulates LYVE-1 expression, with associated decreased hyaluronan uptake. Mol Med 2020; 26:75. [PMID: 32736525 PMCID: PMC7395348 DOI: 10.1186/s10020-020-00204-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 07/24/2020] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Therapeutic lymphangiogenesis in an orthotopic lung transplant model has been shown to improve acute allograft rejection that is mediated at least in part through hyaluronan drainage. Lymphatic vessel endothelial hyaluronan receptor (LYVE-1) expressed on the surface of lymphatic endothelial cells plays important roles in hyaluronan uptake. The impact of current immunosuppressive therapies on lung lymphatic endothelial cells is largely unknown. We tested the hypothesis that FK506, the most commonly used immunosuppressant after lung transplantation, induces lung lymphatic endothelial cell dysfunction. METHODS Lung lymphatic endothelial cells were cultured in vitro and treated with FK506. Telomerase activity was measured using the TRAP assay. Protein expression of LYVE-1 and senescence markers p21 and β-galactosidase was assessed with western blotting. Matrigel tubulation assay were used to investigate the effects of FK506 on TNF-α-induced lymphangiogenesis. Dual luciferase reporter assay was used to confirm NFAT-dependent transcriptional regulation of LYVE-1. Flow cytometry was used to examine the effects of FK506 on LYVE-1 in precision-cut-lung-slices ex vivo and on hyaluronan uptake in vitro. RESULTS In vitro, FK506 downregulated telomerase reverse transcriptase expression, resulting in decreased telomerase activity and subsequent induction of p21 expression and cell senescence. Treatment with FK506 decreased LYVE-1 mRNA and protein levels and resulted in decreased LEC HA uptake. Similar result showing reduction of LYVE-1 expression when treated with FK506 was observed ex vivo. We identified a putative NFAT binding site on the LYVE-1 promoter and cloned this region of the promoter in a luciferase-based reporter construct. We showed that this NFAT binding site regulates LYVE-1 transcription, and mutation of this binding site blunted FK506-dependent downregulation of LYVE-1 promoter-dependent transcription. Finally, FK506-treated lymphatic endothelial cells show a blunted response to TNF-α-mediated lymphangiogenesis. CONCLUSION FK506 alters lymphatic endothelial cell molecular characteristics and causes lymphatic endothelial cell dysfunction in vitro and ex vivo. These effects of FK506 on lymphatic endothelial cell may impair the ability of the transplanted lung to drain hyaluronan macromolecules in vivo. The implications of our findings on the long-term health of lung allografts merit more investigation.
Collapse
Affiliation(s)
- Shikshya Shrestha
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Woohyun Cho
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
- Present Address: Division of Pulmonology, Allergy and Critical Care Medicine, Department of Internal Medicine, Pusan National University Yangsan Hospital, Yangsan, Republic of Korea
| | - Benjamin Stump
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Jewel Imani
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Anthony M Lamattina
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Pierce H Louis
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - James Pazzanese
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Ivan O Rosas
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Gary Visner
- Deparmtent of Pediatrics, Boston Children Hospital, Harvard Medical School, Boston, MA, USA
| | - Mark A Perrella
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Souheil El-Chemaly
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
29
|
Cancer stem cells and oral cancer: insights into molecular mechanisms and therapeutic approaches. Cancer Cell Int 2020; 20:113. [PMID: 32280305 PMCID: PMC7137421 DOI: 10.1186/s12935-020-01192-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Accepted: 03/27/2020] [Indexed: 02/06/2023] Open
Abstract
Cancer stem cells (CSCs) have been identified as a little population of cancer cells, which have features as the same as the cells normal stem cells. There is enough knowledge of the CSCs responsibility for metastasis, medicine resistance, and cancer outbreak. Therefore, CSCs control possibly provides an efficient treatment intervention inhibiting tumor growth and invasion. In spite of the significance of targeting CSCs in treating cancer, few study comprehensively explored the nature of oral CSCs. It has been showed that oral CSCs are able to contribute to oral cancer progression though activation/inhibition a sequences of cellular and molecular pathways (microRNA network, histone modifications and calcium regulation). Hence, more understanding about the properties of oral cancers and their behaviors will help us to develop new therapeutic platforms. Head and neck CSCs remain a viable and intriguing option for targeted therapy. Multiple investigations suggested the major contribution of the CSCs to the metastasis, tumorigenesis, and resistance to the new therapeutic regimes. Therefore, experts in the field are examining the encouraging targeted therapeutic choices. In spite of the advancements, there are not enough information in this area and thus a magic bullet for targeting and eliminating the CSCs deviated us. Hence, additional investigations on the combined therapies against the head and neck CSCs could offer considerable achievements. The present research is a review of the recent information on oral CSCs, and focused on current advancements in new signaling pathways contributed to their stemness regulation. Moreover, we highlighted various therapeutic approaches against oral CSCs.
Collapse
|
30
|
Fenninger F, Jefferies WA. What's Bred in the Bone: Calcium Channels in Lymphocytes. THE JOURNAL OF IMMUNOLOGY 2019; 202:1021-1030. [PMID: 30718290 DOI: 10.4049/jimmunol.1800837] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Accepted: 08/22/2018] [Indexed: 12/30/2022]
Abstract
Calcium (Ca2+) is an important second messenger in lymphocytes and is essential in regulating various intracellular pathways that control critical cell functions. Ca2+ channels are located in the plasma membrane and intracellular membranes, facilitating Ca2+ entry into the cytoplasm. Upon Ag receptor stimulation, Ca2+ can enter the lymphocyte via the Ca2+ release-activated Ca2+ channel found in the plasma membrane. The increase of cytosolic Ca2+ modulates signaling pathways, resulting in the transcription of target genes implicated in differentiation, activation, proliferation, survival, and apoptosis of lymphocytes. Along with Ca2+ release-activated Ca2+ channels, several other channels have been found in the membranes of T and B lymphocytes contributing to key cellular events. Among them are the transient receptor potential channels, the P2X receptors, voltage-dependent Ca2+ channels, and the inositol 1,4,5-trisphosphate receptor as well as the N-methyl-d-aspartate receptors. In this article, we review the contributions of these channels to mediating Ca2+ currents that drive specific lymphocyte functions.
Collapse
Affiliation(s)
- Franz Fenninger
- Michael Smith Laboratories, University of British Columbia, Vancouver V6T 1Z4, British Columbia, Canada.,Department of Microbiology and Immunology, University of British Columbia, Vancouver V6T 1Z3, British Columbia, Canada
| | - Wilfred A Jefferies
- Michael Smith Laboratories, University of British Columbia, Vancouver V6T 1Z4, British Columbia, Canada; .,Department of Microbiology and Immunology, University of British Columbia, Vancouver V6T 1Z3, British Columbia, Canada.,Vancouver Prostate Centre, University of British Columbia, Vancouver V6H 3Z6, British Columbia, Canada.,Centre for Blood Research, University of British Columbia, Vancouver V6T 1Z4, British Columbia, Canada.,The Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver V6T 1Z3, British Columbia, Canada.,Department of Medical Genetics, University of British Columbia, Vancouver V6T 1Z4, British Columbia, Canada; and.,Department of Zoology, University of British Columbia, Vancouver V6T 1Z4, British Columbia, Canada
| |
Collapse
|
31
|
Shin KH, Kim RH. An Updated Review of Oral Cancer Stem Cells and Their Stemness Regulation. Crit Rev Oncog 2019; 23:189-200. [PMID: 30311574 DOI: 10.1615/critrevoncog.2018027501] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Cancer stem cells (CSCs; also known as tumor-initiating cells) are a small population of cancer cells that retain characteristics similar to those of normal stem cells. CSCs are known to be responsible for metastasis, drug resistance, and cancer recurrence. Thus, controlling CSCs may provide an effective therapeutic intervention that inhibits tumor growth and aggressiveness. Despite the importance of targeting CSCs in cancer therapy, the detailed nature of oral CSCs remains underexplored. This article reviews the current understanding of oral CSCs, with emphasis on recent advances in novel signaling pathways involved in their stemness regulation.
Collapse
Affiliation(s)
- Ki-Hyuk Shin
- The Shapiro Family Laboratory of Viral Oncology and Aging Research, UCLA School of Dentistry, Los Angeles, CA 90095; UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA 90095
| | - Reuben H Kim
- The Shapiro Family Laboratory of Viral Oncology and Aging Research, UCLA School of Dentistry, Los Angeles, CA 90095; UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA 90095
| |
Collapse
|
32
|
A calcium/cAMP signaling loop at the ORAI1 mouth drives channel inactivation to shape NFAT induction. Nat Commun 2019; 10:1971. [PMID: 31036819 PMCID: PMC6488650 DOI: 10.1038/s41467-019-09593-0] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Accepted: 03/20/2019] [Indexed: 02/06/2023] Open
Abstract
ORAI1 constitutes the store-operated Ca2+ release-activated Ca2+ (CRAC) channel crucial for life. Whereas ORAI1 activation by Ca2+-sensing STIM proteins is known, still obscure is how ORAI1 is turned off through Ca2+-dependent inactivation (CDI), protecting against Ca2+ toxicity. Here we identify a spatially-restricted Ca2+/cAMP signaling crosstalk critical for mediating CDI. Binding of Ca2+-activated adenylyl cyclase 8 (AC8) to the N-terminus of ORAI1 positions AC8 near the mouth of ORAI1 for sensing Ca2+. Ca2+ permeating ORAI1 activates AC8 to generate cAMP and activate PKA. PKA, positioned by AKAP79 near ORAI1, phosphorylates serine-34 in ORAI1 pore extension to induce CDI whereas recruitment of the phosphatase calcineurin antagonizes the effect of PKA. Notably, CDI shapes ORAI1 cytosolic Ca2+ signature to determine the isoform and degree of NFAT activation. Thus, we uncover a mechanism of ORAI1 inactivation, and reveal a hitherto unappreciated role for inactivation in shaping cellular Ca2+ signals and NFAT activation. ORAI1 constitutes the store-operated Ca2+ release-activated Ca2+ (CRAC) channel, but how this channel is turned off through Ca2+-dependent inactivation (CDI) remained unclear. Here the authors identify a spatially-restricted Ca2+/cAMP signaling crosstalk critical for mediating CDI which in turn regulates cellular Ca2+ signals and NFAT activation.
Collapse
|
33
|
TRPC-mediated Ca 2+ signaling and control of cellular functions. Semin Cell Dev Biol 2019; 94:28-39. [PMID: 30738858 DOI: 10.1016/j.semcdb.2019.02.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 01/30/2019] [Accepted: 02/06/2019] [Indexed: 12/15/2022]
Abstract
Canonical members of the TRP superfamily of ion channels have long been recognized as key elements of Ca2+ handling in a plethora of cell types. The emerging role of TRPC channels in human physiopathology has generated considerable interest in their pharmacological targeting, which requires detailed understanding of their molecular function. Although consent has been reached that receptor-phospholipase C (PLC) pathways and generation of lipid mediators constitute the prominent upstream signaling process that governs channel activity, multimodal sensing features of TRPC complexes have been demonstrated repeatedly. Downstream signaling by TRPC channels is similarly complex and involves the generation of local and global cellular Ca2+ rises, which are well-defined in space and time to govern specific cellular functions. These TRPC-mediated Ca2+ signals rely in part on Ca2+ permeation through the channels, but are essentially complemented by secondary mechanisms such as Ca2+ mobilization from storage sites and Na+/Ca2+ exchange, which involve coordinated interaction with signaling partners. Consequently, the control of cell functions by TRPC molecules is critically determined by dynamic assembly and subcellular targeting of the TRPC complexes. The very recent availability of high-resolution structure information on TRPC channel complexes has paved the way towards a comprehensive understanding of signal transduction by TRPC channels. Here, we summarize current concepts of cation permeation in TRPC complexes, TRPC-mediated shaping of cellular Ca2+ signals and the associated control of specific cell functions.
Collapse
|
34
|
Jiang J, Tang M, Huang Z, Chen L. Junctophilins emerge as novel therapeutic targets. J Cell Physiol 2019; 234:16933-16943. [DOI: 10.1002/jcp.28405] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 01/25/2019] [Accepted: 01/30/2019] [Indexed: 12/24/2022]
Affiliation(s)
- Jinyong Jiang
- Institute of Pharmacy and Pharmacology, Learning Key Laboratory for Pharmacoproteomics, Hunan Province Cooperative Innovation Center for Molecular Target New Drugs Study University of South China Hengyang China
| | - Mingzhu Tang
- Institute of Pharmacy and Pharmacology, Learning Key Laboratory for Pharmacoproteomics, Hunan Province Cooperative Innovation Center for Molecular Target New Drugs Study University of South China Hengyang China
| | - Zhen Huang
- Institute of Pharmacy and Pharmacology, Learning Key Laboratory for Pharmacoproteomics, Hunan Province Cooperative Innovation Center for Molecular Target New Drugs Study University of South China Hengyang China
| | - Linxi Chen
- Institute of Pharmacy and Pharmacology, Learning Key Laboratory for Pharmacoproteomics, Hunan Province Cooperative Innovation Center for Molecular Target New Drugs Study University of South China Hengyang China
| |
Collapse
|
35
|
Bhuvaneshwari S, Sankaranarayanan K. Identification of potential CRAC channel inhibitors: Pharmacophore mapping, 3D-QSAR modelling, and molecular docking approach. SAR AND QSAR IN ENVIRONMENTAL RESEARCH 2019; 30:81-108. [PMID: 30773908 DOI: 10.1080/1062936x.2019.1566172] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Indexed: 06/09/2023]
Abstract
Upregulation of store-operated Ca2+ influx via ORAI1, an integral component of the CRAC channel, is responsible for abnormal cytokine release in active rheumatoid arthritis, and therefore ORAI1 has been proposed as an attractive molecular target. In this study, we attempted to predict the mechanical insights of ORAI1 inhibitors through pharmacophore modelling, 3D-QSAR, molecular docking and free energy analysis. Various hypotheses of pharmacophores were generated and from that, a pharmacophore hypothesis with two hydrogen bond acceptors, one hydrogen bond donor and two aromatic rings (AADRR) resulted in a statistically significant 3D-QSAR model (r2 = 0.84 and q2 = 0.74). We believe that the obtained statistical model is a reliable QSAR model for the diverse dataset of inhibitors against the IL-2 production assay. The visualization of contours in active and inactive compounds generated from the 3D-QSAR models and molecular docking studies revealed major interaction with GLN108, HIS113 and ASP114, and interestingly, these residues are located near the Ca2+ selectivity filter region. Free energy binding analysis revealed that Coulomb energy, van der Waals energy and non-polar solvation terms are more favourable for ligand binding. Thus, the present study provides the physical and chemical requirements for the development of novel ORAI1 inhibitors with improved biological activity.
Collapse
Affiliation(s)
- S Bhuvaneshwari
- a Ion Channel Biology Laboratory, AU-KBC Research Centre, Madras Institute of Technology, Anna University , Chennai , India
| | - K Sankaranarayanan
- a Ion Channel Biology Laboratory, AU-KBC Research Centre, Madras Institute of Technology, Anna University , Chennai , India
| |
Collapse
|
36
|
Nakamura A, Ikeda K, Hamaoka K. Aetiological Significance of Infectious Stimuli in Kawasaki Disease. Front Pediatr 2019; 7:244. [PMID: 31316950 PMCID: PMC6611380 DOI: 10.3389/fped.2019.00244] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Accepted: 05/29/2019] [Indexed: 01/23/2023] Open
Abstract
Kawasaki disease (KD) is a pediatric vasculitis syndrome that is often involves coronary artery lesions (e. g., coronary artery aneurysms). Although its causal factors and entire pathogenesis remain elusive, the available evidence indicates that the pathogenesis of KD is closely associated with dysregulation of immune responses to various viruses or microbes. In this short review, we address several essential aspects of the etiology of KD with respect to the immune response to infectious stimuli: 1) the role of viral infections, 2) the role of bacterial infections and the superantigen hypothesis, 3) involvement of innate immune response including pathogens/microbe-associated molecular patterns and complement pathways, and 4) the influence of genetic background on the response to infectious stimuli. Based on the clinical and experimental evidence, we discuss the possibility that a wide range of microbes and viruses could cause KD through common and distinct immune processes.
Collapse
Affiliation(s)
- Akihiro Nakamura
- Central Research Laboratory, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Kazuyuki Ikeda
- Department of Pediatrics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Kenji Hamaoka
- Pediatric Cardiology and Kawasaki Disease Center, Uji-Tokushukai Medical Center, Kyoto, Japan.,Faculty of Life and Medical Sciences, Doshisha University, Kyoto, Japan
| |
Collapse
|
37
|
Schober R, Waldherr L, Schmidt T, Graziani A, Stilianu C, Legat L, Groschner K, Schindl R. STIM1 and Orai1 regulate Ca 2+ microdomains for activation of transcription. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2018; 1866:1079-1091. [PMID: 30408546 DOI: 10.1016/j.bbamcr.2018.11.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 10/31/2018] [Accepted: 11/01/2018] [Indexed: 02/07/2023]
Abstract
Since calcium (Ca2+) regulates a large variety of cellular signaling processes in a cell's life, precise control of Ca2+ concentrations within the cell is essential. This enables the transduction of information via Ca2+ changes in a time-dependent and spatially defined manner. Here, we review molecular and functional aspects of how the store-operated Ca2+ channel Orai1 creates spatiotemporal Ca2+ microdomains. The architecture of this channel is unique, with a long helical pore and a six-fold symmetry. Energetic barriers within the Ca2+ channel pathway limit permeation to allow an extensive local Ca2+ increase in close proximity to the channel. The precise timing of the Orai1 channel function is controlled by direct binding to STIM proteins upon Ca2+ depletion in the endoplasmic reticulum. These induced Ca2+ microdomains are tailored to, and sufficient for, triggering long-term activation processes, such as transcription factor activation and subsequent gene regulation. We describe the principles of spatiotemporal activation of the transcription factor NFAT and compare its signaling characteristics to those of the autophagy regulating transcription factors, MITF and TFEB.
Collapse
Affiliation(s)
- Romana Schober
- Institute for Biophysics, Johannes Kepler University Linz, A-4040 Linz, Austria.
| | - Linda Waldherr
- Gottfried Schatz Research Center, Medical University of Graz, A-8010 Graz, Austria
| | - Tony Schmidt
- Gottfried Schatz Research Center, Medical University of Graz, A-8010 Graz, Austria
| | - Annarita Graziani
- Gottfried Schatz Research Center, Medical University of Graz, A-8010 Graz, Austria
| | - Clemens Stilianu
- Gottfried Schatz Research Center, Medical University of Graz, A-8010 Graz, Austria
| | - Lorenz Legat
- Gottfried Schatz Research Center, Medical University of Graz, A-8010 Graz, Austria
| | - Klaus Groschner
- Gottfried Schatz Research Center, Medical University of Graz, A-8010 Graz, Austria
| | - Rainer Schindl
- Gottfried Schatz Research Center, Medical University of Graz, A-8010 Graz, Austria.
| |
Collapse
|
38
|
Diener C, Hart M, Alansary D, Poth V, Walch-Rückheim B, Menegatti J, Grässer F, Fehlmann T, Rheinheimer S, Niemeyer BA, Lenhof HP, Keller A, Meese E. Modulation of intracellular calcium signaling by microRNA-34a-5p. Cell Death Dis 2018; 9:1008. [PMID: 30262862 PMCID: PMC6160487 DOI: 10.1038/s41419-018-1050-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 09/10/2018] [Accepted: 09/10/2018] [Indexed: 12/21/2022]
Abstract
Adjusting intracellular calcium signaling is an important feature in the regulation of immune cell function and survival. Here we show that miR-34a-5p, a small non-coding RNA that is deregulated in many common diseases, is a regulator of store-operated Ca2+ entry (SOCE) and calcineurin signaling. Upon miR-34a-5p overexpression, we observed both a decreased depletion of ER calcium content and a decreased Ca2+ influx through Ca2+ release-activated Ca2+ channels. Based on an in silico target prediction we identified multiple miR-34a-5p target genes within both pathways that are implicated in the balance between T-cell activation and apoptosis including ITPR2, CAMLG, STIM1, ORAI3, RCAN1, PPP3R1, and NFATC4. Functional analysis revealed a decrease in Ca2+ activated calcineurin pathway activity measured by a reduced IL-2 secretion due to miR-34a-5p overexpression. Impacting SOCE and/or downstream calcineurin/NFAT signaling by miR-34a-5p offers a possible future approach to manipulate immune cells for clinical interventions.
Collapse
Affiliation(s)
- Caroline Diener
- Institute of Human Genetics, Saarland University, 66421, Homburg, Germany.
| | - Martin Hart
- Institute of Human Genetics, Saarland University, 66421, Homburg, Germany
| | - Dalia Alansary
- Molecular Biophysics, Center for Integrative Physiology and Molecular Medicine, School of Medicine, Saarland University, 66421, Homburg, Germany
| | - Vanessa Poth
- Molecular Biophysics, Center for Integrative Physiology and Molecular Medicine, School of Medicine, Saarland University, 66421, Homburg, Germany
| | - Barbara Walch-Rückheim
- Institute of Virology and Center of Human and Molecular Biology, Saarland University, 66421, Homburg, Germany
| | - Jennifer Menegatti
- Institute of Virology and Center of Human and Molecular Biology, Medical School, Saarland University, 66421, Homburg, Germany
| | - Friedrich Grässer
- Institute of Virology and Center of Human and Molecular Biology, Medical School, Saarland University, 66421, Homburg, Germany
| | - Tobias Fehlmann
- Chair for Clinical Bioinformatics, Saarland University, 66123, Saarbrücken, Germany
| | | | - Barbara A Niemeyer
- Molecular Biophysics, Center for Integrative Physiology and Molecular Medicine, School of Medicine, Saarland University, 66421, Homburg, Germany
| | - Hans-Peter Lenhof
- Center for Bioinformatics, Saarland Informatics Campus, Saarland University, 66123, Saarbrücken, Germany
| | - Andreas Keller
- Chair for Clinical Bioinformatics, Saarland University, 66123, Saarbrücken, Germany
| | - Eckart Meese
- Institute of Human Genetics, Saarland University, 66421, Homburg, Germany
| |
Collapse
|
39
|
CRAC Channel Components Quantitative Expression (In Tissues and Cell Lines) Using qPCR. Methods Mol Biol 2018. [PMID: 30203280 DOI: 10.1007/978-1-4939-8704-7_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
Since last decade real-time qPCR has become a routine and robust approach for measuring the expression of genes of interest. Indeed, using qPCR, expression profile analyses are now possible and participate to the understanding of physiological or pathological role of channels such as calcium release-activated channels (CRAC). Initially discovered in lymphocyte T and immunity perturbations, recent studies have highlighted the role of CRAC channels in other pathologies such as cancer. Here we describe a protocol to quantify CRAC components expression, in tissue sample and cell lines, to validate knockdown strategies or identify their roles in physiological and pathological conditions (Hoth and Penner, J Physiol 465:359-386, 1993; Hoth and Penner, Nature 355:353-356, 1992; and Zweifach and Lewis, Proc Natl Acad Sci U S A 90:6295-6299, 1993).
Collapse
|
40
|
Duong-Ly KC, Kuo YM, Johnson MC, Cote JM, Kollman JM, Soboloff J, Rall GF, Andrews AJ, Peterson JR. T cell activation triggers reversible inosine-5'-monophosphate dehydrogenase assembly. J Cell Sci 2018; 131:jcs.223289. [PMID: 30154209 DOI: 10.1242/jcs.223289] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 08/06/2018] [Indexed: 12/17/2022] Open
Abstract
T cell-mediated adaptive immunity requires naïve, unstimulated T cells to transition from a quiescent metabolic state into a highly proliferative state upon T cell receptor engagement. This complex process depends on transcriptional changes mediated by Ca2+-dependent NFAT signaling, mTOR-mediated signaling and increased activity of the guanine nucleotide biosynthetic inosine-5'-monophosphate (IMP) dehydrogenase 1 and 2 enzymes (IMPDH1 and IMPDH2, hereafter IMPDH). Inhibitors of these pathways serve as potent immunosuppressants. Unexpectedly, we discovered that all three pathways converge to promote the assembly of IMPDH protein into micron-scale macromolecular filamentous structures in response to T cell activation. Assembly is post-transcriptionally controlled by mTOR and the Ca2+ influx regulator STIM1. Furthermore, IMPDH assembly and catalytic activity were negatively regulated by guanine nucleotide levels, suggesting a negative feedback loop that limits biosynthesis of guanine nucleotides. Filamentous IMPDH may be more resistant to this inhibition, facilitating accumulation of the higher GTP levels required for T cell proliferation.
Collapse
Affiliation(s)
- Krisna C Duong-Ly
- Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Yin-Ming Kuo
- Cancer Epigenetics Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Matthew C Johnson
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Joy M Cote
- Cancer Epigenetics Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Justin M Kollman
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Jonathan Soboloff
- Fels Institute for Cancer Research and Molecular Biology, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | - Glenn F Rall
- Blood Cell Development and Function Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Andrew J Andrews
- Cancer Epigenetics Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Jeffrey R Peterson
- Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| |
Collapse
|
41
|
Mei Y, Barrett JE, Hu H. Calcium release-activated calcium channels and pain. Cell Calcium 2018; 74:180-185. [PMID: 30096536 DOI: 10.1016/j.ceca.2018.07.009] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 07/10/2018] [Accepted: 07/27/2018] [Indexed: 12/30/2022]
Abstract
Calcium release-activated calcium (CRAC) channels are unique among ion channels that are activated in response to depletion of intracellular calcium stores and are highly permeable to Ca2+ compared to other cations. CRAC channels mediate an important calcium signal for a wide variety of cell types and are well studied in the immune system. They have been implicated in a number of disorders such as immunodeficiency, musculosketal disorders and cancer. There is growing evidence showing that CRAC channels are expressed in the nervous system and are involved in pathological conditions including pain. This review summarizes the expression, distribution, and function of the CRAC channel family in the dorsal root ganglion, spinal cord and some brain regions, and discusses their functional significance in neurons and glial cells and involvement in nociception and chronic pain. Although further studies are needed to understand how these channels are activated under physiological conditions, the recent findings indicate that the CRAC channel Orai1 is an important player in pain modulation and could represent a new target for pathological pain.
Collapse
Affiliation(s)
- Yixiao Mei
- Department of Anesthesiology, Rutgers New Jersey Medical School, Newark, NJ 07103, United States
| | - James E Barrett
- Department of Neurology, Drexel University College of Medicine Philadelphia, PA 19102, United States
| | - Huijuan Hu
- Department of Anesthesiology, Rutgers New Jersey Medical School, Newark, NJ 07103, United States.
| |
Collapse
|
42
|
Huang TY, Lin YH, Chang HA, Yeh TY, Chang YH, Chen YF, Chen YC, Li CC, Chiu WT. STIM1 Knockout Enhances PDGF-Mediated Ca 2+ Signaling through Upregulation of the PDGFR⁻PLCγ⁻STIM2 Cascade. Int J Mol Sci 2018; 19:ijms19061799. [PMID: 29912163 PMCID: PMC6032054 DOI: 10.3390/ijms19061799] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2018] [Revised: 06/13/2018] [Accepted: 06/14/2018] [Indexed: 01/24/2023] Open
Abstract
Platelet-derived growth factor (PDGF) has mitogenic and chemotactic effects on fibroblasts. An increase in intracellular Ca2+ is one of the first events that occurs following the stimulation of PDGF receptors (PDGFRs). PDGF activates Ca2+ elevation by activating the phospholipase C gamma (PLCγ)-signaling pathway, resulting in ER Ca2+ release. Store-operated Ca2+ entry (SOCE) is the major form of extracellular Ca2+ influx following depletion of ER Ca2+ stores and stromal interaction molecule 1 (STIM1) is a key molecule in the regulation of SOCE. In this study, wild-type and STIM1 knockout mouse embryonic fibroblasts (MEF) cells were used to investigate the role of STIM1 in PDGF-induced Ca2+ oscillation and its functions in MEF cells. The unexpected findings suggest that STIM1 knockout enhances PDGFR–PLCγ–STIM2 signaling, which in turn increases PDGF-BB-induced Ca2+ elevation. Enhanced expressions of PDGFRs and PLCγ in STIM1 knockout cells induce Ca2+ release from the ER store through PLCγ–IP3 signaling. Moreover, STIM2 replaces STIM1 to act as the major ER Ca2+ sensor in activating SOCE. However, activation of PDGFRs also activate Akt, ERK, and JNK to regulate cellular functions, such as cell migration. These results suggest that alternative switchable pathways can be observed in cells, which act downstream of the growth factors that regulate Ca2+ signaling.
Collapse
Affiliation(s)
- Tzu-Yu Huang
- Department of Biomedical Engineering, National Cheng Kung University, Tainan 701, Taiwan.
| | - Yi-Hsin Lin
- Department of Biomedical Engineering, National Cheng Kung University, Tainan 701, Taiwan.
| | - Heng-Ai Chang
- Institute of Basic Medical Sciences, National Cheng Kung University, Tainan 701, Taiwan.
| | - Tzu-Ying Yeh
- Department of Biomedical Engineering, National Cheng Kung University, Tainan 701, Taiwan.
| | - Ya-Han Chang
- Department of Biomedical Engineering, National Cheng Kung University, Tainan 701, Taiwan.
| | - Yi-Fan Chen
- Institute of Basic Medical Sciences, National Cheng Kung University, Tainan 701, Taiwan.
| | - Ying-Chi Chen
- Department of Biomedical Engineering, National Cheng Kung University, Tainan 701, Taiwan.
| | - Chun-Chun Li
- Department of Life Sciences, National Cheng Kung University, Tainan 701, Taiwan.
| | - Wen-Tai Chiu
- Department of Biomedical Engineering, National Cheng Kung University, Tainan 701, Taiwan.
- Institute of Basic Medical Sciences, National Cheng Kung University, Tainan 701, Taiwan.
| |
Collapse
|
43
|
McIvor E, Coombes S, Thul R. Three-dimensional spatio-temporal modelling of store operated Ca 2+ entry: Insights into ER refilling and the spatial signature of Ca 2+ signals. Cell Calcium 2018; 73:11-24. [PMID: 29880194 DOI: 10.1016/j.ceca.2018.03.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Revised: 03/29/2018] [Accepted: 03/30/2018] [Indexed: 11/19/2022]
Abstract
The spatial organisation of Orai channels and SERCA pumps within ER-PM junctions is important for enhancing the versatility and specificity of sub-cellular Ca2+ signals generated during store operated Ca2+ entry (SOCE). In this paper, we present a novel three dimensional spatio-temporal model describing Ca2+ dynamics in the ER-PM junction and sub-PM ER during SOCE. We investigate the role of Orai channel and SERCA pump location to provide insights into how these components shape the Ca2+ signals generated and affect ER refilling. We find that the organisation of Orai channels within the ER-PM junction controls the amplitude and shape of the Ca2+ profile but does not enhance ER refilling. The model shows that ER refilling is only weakly affected by the location of SERCA2b pumps within the ER-PM junction and that the placement of SERCA2a pumps within the ER-PM junction has much greater control over ER refilling.
Collapse
Affiliation(s)
- Emma McIvor
- Centre for Mathematical Medicine and Biology, School of Mathematical Sciences, University of Nottingham, University Park, Nottingham NG7 2RD, UK.
| | - Stephen Coombes
- Centre for Mathematical Medicine and Biology, School of Mathematical Sciences, University of Nottingham, University Park, Nottingham NG7 2RD, UK
| | - Rüdiger Thul
- Centre for Mathematical Medicine and Biology, School of Mathematical Sciences, University of Nottingham, University Park, Nottingham NG7 2RD, UK
| |
Collapse
|
44
|
Lee SH, Rigas NK, Lee CR, Bang A, Srikanth S, Gwack Y, Kang MK, Kim RH, Park NH, Shin KH. Orai1 promotes tumor progression by enhancing cancer stemness via NFAT signaling in oral/oropharyngeal squamous cell carcinoma. Oncotarget 2017; 7:43239-43255. [PMID: 27259269 PMCID: PMC5190020 DOI: 10.18632/oncotarget.9755] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Accepted: 05/26/2016] [Indexed: 01/03/2023] Open
Abstract
Emerging evidence indicates that Orai1, a key calcium channel for store-operated Ca2+ entry, is associated with human cancer. However, the underlying mechanism by which Orai1 regulates cancer progression remains unknown. Here we report that intracellular level of Orai1 is increased in a stepwise manner during oral/oropharyngeal carcinogenesis and highly expressed in cancer stem-like cell (CSC)-enriched populations of human oral/oropharyngeal squamous cell carcinoma (OSCC). Ectopic Orai1 expression converted non-tumorigenic immortalized oral epithelial cells to malignant cells that showed CSC properties, e.g., self-renewal capacity, increased ALDH1HIGH cell population, increased key stemness transcription factors, and enhanced mobility. Conversely, inhibition of Orai1 suppressed tumorigenicity and CSC phenotype of OSCC, indicating that Orai1 could be an important element for tumorigenicity and stemness of OSCC. Mechanistically, Orai1 activates its major downstream effector molecule, NFATc3. Knockdown of NFATc3 in the Orai1-overexpressing oral epithelial cells abrogates the effect of Orai1 on CSC phenotype. Moreover, antagonist of NFAT signaling also decreases CSC phenotype, implying the functional importance of Orai1/NFAT axis in OSCC CSC regulation. Our study identifies Orai1 as a novel molecular determinant for OSCC progression by enhancing cancer stemness, suggesting that inhibition of Orai1 signaling may offer an effective therapeutic modality against OSCC.
Collapse
Affiliation(s)
- Sung Hee Lee
- The Shapiro Family Laboratory of Viral Oncology and Aging Research, UCLA School of Dentistry, Los Angeles, CA, USA
| | - Nicole Kristina Rigas
- The Shapiro Family Laboratory of Viral Oncology and Aging Research, UCLA School of Dentistry, Los Angeles, CA, USA
| | - Chang-Ryul Lee
- The Shapiro Family Laboratory of Viral Oncology and Aging Research, UCLA School of Dentistry, Los Angeles, CA, USA
| | - April Bang
- The Shapiro Family Laboratory of Viral Oncology and Aging Research, UCLA School of Dentistry, Los Angeles, CA, USA
| | - Sonal Srikanth
- Department of Physiology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Yousang Gwack
- Department of Physiology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Mo K Kang
- The Shapiro Family Laboratory of Viral Oncology and Aging Research, UCLA School of Dentistry, Los Angeles, CA, USA.,UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA, USA
| | - Reuben H Kim
- The Shapiro Family Laboratory of Viral Oncology and Aging Research, UCLA School of Dentistry, Los Angeles, CA, USA.,UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA, USA
| | - No-Hee Park
- The Shapiro Family Laboratory of Viral Oncology and Aging Research, UCLA School of Dentistry, Los Angeles, CA, USA.,UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA, USA.,Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Ki-Hyuk Shin
- The Shapiro Family Laboratory of Viral Oncology and Aging Research, UCLA School of Dentistry, Los Angeles, CA, USA.,UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA, USA
| |
Collapse
|
45
|
Nam JH, Jung HW, Chin YW, Yang WM, Bae HS, Kim WK. Spirodela polyrhiza extract modulates the activation of atopic dermatitis-related ion channels, Orai1 and TRPV3, and inhibits mast cell degranulation. PHARMACEUTICAL BIOLOGY 2017; 55:1324-1329. [PMID: 28290212 PMCID: PMC6130684 DOI: 10.1080/13880209.2017.1300819] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Revised: 11/25/2016] [Accepted: 01/25/2017] [Indexed: 06/06/2023]
Abstract
CONTEXT Spirodela polyrhiza (L.) Schleid. (Lemnaceae), Spirodelae Herba (SH), has been known to relieve inflammation, urticaria and skin symptoms including pruritus, eczema and rash. OBJECTIVE The effects of SH extract on two calcium ion channels, Orai1 and TRPV3, and their potential as novel therapeutics for atopic dermatitis (AD) were investigated. The regulatory role of Orai1 on mast cell degranulation was evaluated. MATERIALS AND METHODS The dried leaves of SH were extracted by 70% methanol. Effects of SH extract (100 μg/mL) in an HEK293T cell line overexpressing human Orai1 or TRPV3 were assessed. Ion channel modulation in transfected HEK293T cells was measured using a conventional whole-cell patch-clamp technique. IgE-antigen complex-stimulated mast cell degranulation was measured by β-hexosaminidase assay with morphological observation after treatment with 20, 50 and 100 μg/mL SH extract. RESULTS SH extract (100 μg/mL) significantly inhibited Orai1 activity (63.8 ± 0.97%) in Orai1-STIM1 co-overexpressed HEK293T cells. SH extract significantly increased TRPV3 activity (81.29 ± 0.05% at -100 mV) compared with the positive control 2-APB (100 μM), which induced full activation. SH extract inhibited degranulation in IgE-antigen complex-stimulated RBL-2H3 mast cells by decreasing β-hexosaminidase activity (3.14 ± 0.03, 2.56 ± 0.12 and 2.29 ± 0.08 mU/mg, respectively). CONCLUSION Our results suggested that SH extract could treat abnormal skin barrier pathologies in AD through modulation of the activities of the calcium ion channels Orai1 and TRPV3 and inhibition of mast cell degranulation. This is the first report of an herbal effect on the modulation of ion channels associated with skin barrier disruption in AD pathogenesis.
Collapse
Affiliation(s)
- Joo Hyun Nam
- Department of Physiology, Dongguk University College of Medicine, Gyeongju, Republic of Korea
- Channelopathy Research Center (CRC), Dongguk University College of Medicine, Goyang, Gyeonggi-do, Republic of Korea
| | - Hyo Won Jung
- College of Korean Medicine, Dongguk University, Gyeongju, Republic of Korea
| | - Young-Won Chin
- College of Pharmacy, Dongguk University, Goyang, Gyeonggi-do, Republic of Korea
| | - Won-Mo Yang
- Department of Biochemistry, Dongguk University College of Medicine, Gyeongju, Republic of Korea
| | - Hyo Sang Bae
- Department of Sasang Constitutional Medicine, College of Korean Medicine, Dongguk University, Goyang, Gyeonggi-do, Republic of Korea
| | - Woo Kyung Kim
- Channelopathy Research Center (CRC), Dongguk University College of Medicine, Goyang, Gyeonggi-do, Republic of Korea
- Department of Internal Medicine, Graduate School of Medicine, Dongguk University, Goyang, Gyeonggi-do, Republic of Korea
| |
Collapse
|
46
|
Uzhachenko R, Shanker A, Dupont G. Computational properties of mitochondria in T cell activation and fate. Open Biol 2017; 6:rsob.160192. [PMID: 27852805 PMCID: PMC5133440 DOI: 10.1098/rsob.160192] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Accepted: 10/12/2016] [Indexed: 01/09/2023] Open
Abstract
In this article, we review how mitochondrial Ca2+ transport (mitochondrial Ca2+ uptake and Na+/Ca2+ exchange) is involved in T cell biology, including activation and differentiation through shaping cellular Ca2+ signals. Based on recent observations, we propose that the Ca2+ crosstalk between mitochondria, endoplasmic reticulum and cytoplasm may form a proportional–integral–derivative (PID) controller. This PID mechanism (which is well known in engineering) could be responsible for computing cellular decisions. In addition, we point out the importance of analogue and digital signal processing in T cell life and implication of mitochondrial Ca2+ transport in this process.
Collapse
Affiliation(s)
- Roman Uzhachenko
- Department of Biochemistry and Cancer Biology, School of Medicine, Meharry Medical College, Nashville, TN, USA
| | - Anil Shanker
- Department of Biochemistry and Cancer Biology, School of Medicine, Meharry Medical College, Nashville, TN, USA .,Host-Tumor Interactions Research Program, Vanderbilt-Ingram Cancer Center, and the Center for Immunobiology, Vanderbilt University, Nashville, TN, USA
| | - Geneviève Dupont
- Unité de Chronobiologie Théorique, Université Libre de Bruxelles, CP231, Boulevard du Triomphe, 1050 Brussels, Belgium
| |
Collapse
|
47
|
Joshi RN, Binai NA, Marabita F, Sui Z, Altman A, Heck AJR, Tegnér J, Schmidt A. Phosphoproteomics Reveals Regulatory T Cell-Mediated DEF6 Dephosphorylation That Affects Cytokine Expression in Human Conventional T Cells. Front Immunol 2017; 8:1163. [PMID: 28993769 PMCID: PMC5622166 DOI: 10.3389/fimmu.2017.01163] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Accepted: 09/01/2017] [Indexed: 12/25/2022] Open
Abstract
Regulatory T cells (Tregs) control key events of immune tolerance, primarily by suppression of effector T cells. We previously revealed that Tregs rapidly suppress T cell receptor (TCR)-induced calcium store depletion in conventional CD4+CD25− T cells (Tcons) independently of IP3 levels, consequently inhibiting NFAT signaling and effector cytokine expression. Here, we study Treg suppression mechanisms through unbiased phosphoproteomics of primary human Tcons upon TCR stimulation and Treg-mediated suppression, respectively. Tregs induced a state of overall decreased phosphorylation as opposed to TCR stimulation. We discovered novel phosphosites (T595_S597) in the DEF6 (SLAT) protein that were phosphorylated upon TCR stimulation and conversely dephosphorylated upon coculture with Tregs. Mutation of these DEF6 phosphosites abrogated interaction of DEF6 with the IP3 receptor and affected NFAT activation and cytokine transcription in primary Tcons. This novel mechanism and phosphoproteomics data resource may aid in modifying sensitivity of Tcons to Treg-mediated suppression in autoimmune disease or cancer.
Collapse
Affiliation(s)
- Rubin N Joshi
- Unit of Computational Medicine, Center for Molecular Medicine, Department of Medicine Solna, Karolinska University Hospital, Science for Life Laboratory, Karolinska Institutet, Stockholm, Sweden
| | - Nadine A Binai
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands.,Netherlands Proteomics Centre, Utrecht, Netherlands
| | - Francesco Marabita
- Unit of Computational Medicine, Center for Molecular Medicine, Department of Medicine Solna, Karolinska University Hospital, Science for Life Laboratory, Karolinska Institutet, Stockholm, Sweden
| | - Zhenhua Sui
- Division of Cell Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA, United States
| | - Amnon Altman
- Division of Cell Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA, United States
| | - Albert J R Heck
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands.,Netherlands Proteomics Centre, Utrecht, Netherlands
| | - Jesper Tegnér
- Unit of Computational Medicine, Center for Molecular Medicine, Department of Medicine Solna, Karolinska University Hospital, Science for Life Laboratory, Karolinska Institutet, Stockholm, Sweden.,Biological and Environmental Sciences and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia.,Computer, Electrical and Mathematical Sciences and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
| | - Angelika Schmidt
- Unit of Computational Medicine, Center for Molecular Medicine, Department of Medicine Solna, Karolinska University Hospital, Science for Life Laboratory, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
48
|
Drebrin Regulation of Calcium Signaling in Immune Cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017. [PMID: 28865026 DOI: 10.1007/978-4-431-56550-5_16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register]
Abstract
Store-operated Ca2+ channels are plasma membrane channels that are activated by depletion of intracellular Ca2+ stores, resulting in an increase in intracellular Ca2+; however, little is known about their regulation. Our work has shown that the immunosuppressant compound BTP2, which blocks Ca2+ influx into cells, interacts with the actin-reorganizing protein, drebrin. Here we review the role of drebrin in the regulation of calcium signaling, with a focus on immune cells.
Collapse
|
49
|
Calcineurin inhibitors regulate fibroblast growth factor 23 (FGF23) synthesis. Naunyn Schmiedebergs Arch Pharmacol 2017; 390:1117-1123. [DOI: 10.1007/s00210-017-1411-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Accepted: 07/21/2017] [Indexed: 01/06/2023]
|
50
|
Thaxton JE, Wallace C, Riesenberg B, Zhang Y, Paulos CM, Beeson CC, Liu B, Li Z. Modulation of Endoplasmic Reticulum Stress Controls CD4 + T-cell Activation and Antitumor Function. Cancer Immunol Res 2017. [PMID: 28642246 DOI: 10.1158/2326-6066.cir-17-0081] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The endoplasmic reticulum (ER) is an energy-sensing organelle with intimate ties to programming cell activation and metabolic fate. T-cell receptor (TCR) activation represents a form of acute cell stress and induces mobilization of ER Ca2+ stores. The role of the ER in programming T-cell activation and metabolic fate remains largely undefined. Gp96 is an ER protein with functions as a molecular chaperone and Ca2+ buffering protein. We hypothesized that the ER stress response may be important for CD4+ T-cell activation and that gp96 may be integral to this process. To test our hypothesis, we utilized genetic deletion of the gp96 gene Hsp90b1 in a CD4+ T cell-specific manner. We show that gp96-deficient CD4+ T cells cannot undergo activation-induced glycolysis due to defective Ca2+ mobilization upon TCR engagement. We found that activating naïve CD4+ T cells while inhibiting ER Ca2+ exchange, through pharmacological blockade of the ER Ca2+ channel inositol trisphosphate receptor (IP3R), led to a reduction in cytosolic Ca2+ content and generated a pool of CD62Lhigh/CD44low CD4+ T cells compared with wild-type (WT) matched controls. In vivo IP3R-inhibited CD4+ T cells exhibited elevated tumor control above WT T cells. Together, these data show that ER-modulated cytosolic Ca2+ plays a role in defining CD4+ T-cell phenotype and function. Factors associated with the ER stress response are suitable targets for T cell-based immunotherapies. Cancer Immunol Res; 5(8); 666-75. ©2017 AACR.
Collapse
Affiliation(s)
- Jessica E Thaxton
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, South Carolina.,Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina.,Department of Orthopaedic Surgery, Medical University of South Carolina, Charleston, South Carolina
| | - Caroline Wallace
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, South Carolina
| | - Brian Riesenberg
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, South Carolina
| | - Yongliang Zhang
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, South Carolina
| | - Chrystal M Paulos
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, South Carolina.,Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina
| | - Craig C Beeson
- Department of Pharmaceutical and Biomedical Sciences, Medical University of South Carolina, Charleston, South Carolina
| | - Bei Liu
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, South Carolina.,Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina
| | - Zihai Li
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, South Carolina. .,Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina.,First Affiliated Hospital, Zhengzhou University School of Medicine, Zhengzhou, China
| |
Collapse
|