1
|
Hoffmann da Silva J, Hoefle CA, Cupper Vieira C, Kreutz LC, Cabrera Dalto AG, Dias Gonçalves PB, Tomazele Rovani M, Ferreira R. How can blood calcium concentration and oral supplementation affect uterine health in dairy cows? Theriogenology 2025; 244:117496. [PMID: 40408801 DOI: 10.1016/j.theriogenology.2025.117496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Revised: 05/13/2025] [Accepted: 05/16/2025] [Indexed: 05/25/2025]
Abstract
A series of experiments was carried out to study the effects of blood calcium (Ca) concentration on the occurrence of subclinical endometritis (SE) in dairy cattle. In Experiment 1, serum Ca level of 26 cows in the first 24 h after calving was evaluated and correlated with the incidence of SE between 34 and 40 days postpartum. The polymorphonuclear leukocytes (PMN) count was affected by Ca blood concentration in the first 24 h postpartum (P ≤ 0.01). In Experiment 2, a total of 47 cows retrospectively classified as normocalcemic (Ca ≥8.5 mg/dL) or hypocalcemic (Ca <8.5 mg/dL) were randomly assigned to receive or not oral Ca formate at 6 and 30 h postpartum, and the effects of supplementation on SE incidence were evaluated. The treatment with Ca formate, regardless of Ca concentration, reduced PMN count and incidence of SE between 34 and 40 days postpartum (P < 0.05). For each increase of 1 mg/dL in blood Ca, SE incidence decreased by 22 %. In Experiment 3, five multiparous Holstein cows were enrolled in a 2x2 crossover model and assigned to 1 of 2 treatments: normocalcemic (Control) or induced subclinical hypocalcemia (iSCH) group. The sequence of treatments for a given cow was either Control-iSCH or iSCH-Control, followed by a 3-week washout period. The iSCH group received an intravenous infusion of 5 % disodium tetraacetic ethylenediamine Na2EDTA at 500 mL/h for 45 min, while the Control group received 0.9 % NaCl. Levels of ionized Ca (iCa), total Ca (tCa) and Mg were determined immediately before the treatments (M0), at 15 (M1), 45 min (M2, end of infusions) and 3 h after the end of treatments (M3). Furthermore, glucose, insulin, NEFA and BHB levels were determined at M3. Immediately after the end of the infusions (M2), all animals received an intrauterine challenge with 300 μg of LPS and, after 3 h (M3), an endometrial biopsy was performed to determine the expression of IL-6, CXCL8 and TNF and gene expression of glucose-insulin receptors and insulin-related genes. Subclinical hypocalcemia was effectively induced; however, the expression of IL-6, CXCL8 and TNF in the endometrium did not differ between groups within 3 h after intrauterine LPS challenge. The iSCH group presented a significant reduction in serum BHB levels compared to the Control group, but there were no differences in serum glucose, insulin, NEFA and in the endometrial expression of the INSR, IRS1, IGF1, SLC2A1, and SLC2A3 genes. In conclusion, PMN infiltration and the incidence of SE from 34 to 40 days postpartum is related to blood Ca immediately after calving. Ca supplementation with Ca formate decreases the number of PMNs in the endometrium, reducing SE incidence. In addition, transient iSCH does not cause immediate significant changes in the endometrial expression of key inflammatory cytokines and does not adversely affect the insulin signaling pathway in the uterus within 3 h following an intrauterine challenge with LPS. This study provides valuable insights on the role of blood calcium and oral calcium supplementation during the immediate postpartum period on uterine health and the metabolic and immunological effects of transient induced subclinical hypocalcemia in dairy cows.
Collapse
Affiliation(s)
| | | | - Camila Cupper Vieira
- Universidade Federal de Santa Maria (UFSM), Santa Maria, Rio Grande do Sul, Brazil
| | - Luiz Carlos Kreutz
- Universidade de Passo Fundo (UPF), Passo Fundo, Rio Grande do Sul, Brazil
| | | | | | | | - Rogério Ferreira
- Universidade do Estado de Santa Catarina (UDESC), Chapecó, Santa Catarina, Brazil.
| |
Collapse
|
2
|
Taha S, Aljishi M, Sultan A, Al-Nashmi ME, Bakhiet M, Spicuglia S, Belhocine M. Progressive Elevation of Store-Operated Calcium Entry-Associated Regulatory Factor (SARAF) and Calcium Pathway Dysregulation in Multiple Sclerosis. Int J Mol Sci 2025; 26:4520. [PMID: 40429665 DOI: 10.3390/ijms26104520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2025] [Revised: 04/15/2025] [Accepted: 04/16/2025] [Indexed: 05/29/2025] Open
Abstract
Multiple Sclerosis (MS) is a chronic autoimmune disorder characterized by demyelination and neuronal damage in the central nervous system. Dysregulation of calcium homeostasis, particularly through the Store-Operated Calcium Entry-Associated Regulatory Factor (SARAF), has been implicated in MS pathogenesis. This study investigated SARAF, STIM1, and Orai1 expression patterns and their relationship to calcium homeostasis in 45 Bahraini MS patients and 45 matched healthy controls using ELISA and real-time PCR analyses. MS patients showed significantly elevated serum SARAF levels in both early (192.26 ± 47.00 pg/mL) and late MS stages (341.47 ± 96.19 pg/mL) compared to controls (129.82 ± 30.82 pg/mL; p < 0.001. SARAF expressions were markedly increased in MS patients (3.829 ± 0.04422 vs. 1 ± 0; p < 0.0001), while STIM1 (0.4324 ± 0.01471) and ORAI1 (0.2963 ± 0.02156) expressions were significantly reduced compared to the controls (p < 0.0001). Intracellular calcium levels were notably elevated in both early and late MS stages. These findings suggest that the progressive elevation of SARAF, coupled with altered STIM1 and ORAI1 expression, may serve as potential biomarkers for MS progression and represent promising therapeutic targets.
Collapse
Affiliation(s)
- Safa Taha
- Princess Al Jawhara Center for Molecular Medicine, Genetics and Inherited Diseases, Department of Molecular Medicine, College of Medicine and Health Sciences, Arabian Gulf University, Manama P.O. Box 26671, Bahrain
| | - Muna Aljishi
- Princess Al Jawhara Center for Molecular Medicine, Genetics and Inherited Diseases, Department of Molecular Medicine, College of Medicine and Health Sciences, Arabian Gulf University, Manama P.O. Box 26671, Bahrain
| | - Ameera Sultan
- Princess Al Jawhara Center for Molecular Medicine, Genetics and Inherited Diseases, Department of Molecular Medicine, College of Medicine and Health Sciences, Arabian Gulf University, Manama P.O. Box 26671, Bahrain
| | - Moudi E Al-Nashmi
- Princess Al Jawhara Center for Molecular Medicine, Genetics and Inherited Diseases, Department of Molecular Medicine, College of Medicine and Health Sciences, Arabian Gulf University, Manama P.O. Box 26671, Bahrain
| | - Moiz Bakhiet
- Princess Al Jawhara Center for Molecular Medicine, Genetics and Inherited Diseases, Department of Molecular Medicine, College of Medicine and Health Sciences, Arabian Gulf University, Manama P.O. Box 26671, Bahrain
| | - Salvatore Spicuglia
- Aix-Marseille University, INSERM, TAGC, UMR1090, Equipe Labélisée Ligue Contre le Cancer, 13288 Marseille, France
| | - Mohamed Belhocine
- Princess Al Jawhara Center for Molecular Medicine, Genetics and Inherited Diseases, Department of Molecular Medicine, College of Medicine and Health Sciences, Arabian Gulf University, Manama P.O. Box 26671, Bahrain
| |
Collapse
|
3
|
Uddin MN, Thomas DW. T Lymphocyte Integrated Endoplasmic Reticulum Ca 2+ Store Signaling Functions Are Linked to Sarco/Endoplasmic Reticulum Ca 2+-ATPase Isoform-Specific Levels of Regulation. Int J Mol Sci 2025; 26:4147. [PMID: 40362384 PMCID: PMC12071366 DOI: 10.3390/ijms26094147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2025] [Revised: 04/24/2025] [Accepted: 04/25/2025] [Indexed: 05/15/2025] Open
Abstract
We explored the effects of altering expression levels of the sarco/endoplasmic reticulum Ca2+-ATPase (SERCA) ion-transporting enzymes on key T lymphocyte signaling functions. In these studies, we have taken advantage of the Jurkat T cell line which provides a T lymphocyte model cell phenotype with a well-characterized T cell receptor (TCR)-activated signaling pathway, as well as offering a cellular system with a good understanding of the SERCA expression profile. These studies have been prompted by a strong imperative to gain a better understanding of the complex roles SERCA Ca2+ pumps play in the integrated TCR-activated signaling network, particularly given the central role of SERCA functions in regulating essential endoplasmic reticulum (ER) integrity. We find in this study that altering SERCA expression can significantly reconfigure ER Ca2+ stores, increasing or decreasing Ca2+ storage capacity depending on upregulation or downregulation of SERCA expression, and these effects are also associated with substantial changes in agonist-induced Ca2+ release and influx patterns. Not surprisingly, these fundamental changes in TCR-regulated Ca2+ signaling properties are associated with major alterations in T lymphocyte functions including regulation of growth patterns, cytokine secretion and energy utilization. Our study also describes additional evidence revealing intriguing functional distinctions between the major SERCA isoform-regulated Ca2+ stores in T lymphocytes. Our work thus serves to reinforce increasing efforts to target the SERCA pumps as a potential profitable strategy to produce novel engineered T lymphocytes in the rapidly growing field of T-cell immunotherapy.
Collapse
Affiliation(s)
| | - David W. Thomas
- Department of Pharmaceutical Sciences, Thomas J. Long School of Pharmacy, University of the Pacific, Stockton, CA 95211, USA;
| |
Collapse
|
4
|
Areesanan A, Wolf L, Nicolay S, Zimmermann-Klemd AM, Gründemann C. Immunomodulatory Effects of Copper Bis-Glycinate In Vitro. Molecules 2025; 30:1282. [PMID: 40142058 PMCID: PMC11944375 DOI: 10.3390/molecules30061282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Revised: 03/04/2025] [Accepted: 03/07/2025] [Indexed: 03/28/2025] Open
Abstract
Copper functions as a cofactor and antioxidants in a large number of enzymes that are important for cellular respiration and the nervous system. In the last century scholars have explored copper's relationship with the immune system, with copper deficiency drastically upsetting the overall function of the immune system, as seen in symptoms such as increased susceptibility to pathogens, decreased proliferation of lymphocytes, and impaired function of both cytotoxic T lymphocytes and helper T cells. Among copper's various forms, copper bis-glycinate (Cbg) has been used as an official EU-approved oral supplement to promote health. In this study, we observed the influence of Cbg on human epithelial cells (HCE-T cells) to determine its cytotoxicity, anti-reactive oxygen (ROS), and wound healing capabilities. We also evaluated Cbg's anti-inflammatory immune cells like primary human mononuclear cells (PBMCs), monocytic THP-1, and Jurkat cells in the context of anti-inflammation. At all the investigated concentrations of Cbg (0.05-100 μg/mL), ther was no considerable impact detected on the epithelial cells. However, the proliferation rate of stimulated PBMCs was affected progressively (3-50 μg/mL). In CD4+ helper T cells, interleukin (IL)-17 and IL-2 cytokine levels were decreased in a dose-dependent manner, while interferon (IFN)-γ and IL-2 levels were slightly decreased with no noticeable changes between each treated concentration. Furthermore, stimulated monocytic THP-1 cells treated with Cbg reduced IL-6 and significantly reduced tumor necrosis factor (TNF)-α cytokines secretion. Lastly, stimulated Jurkat intracellular Ca2+ influx was significantly inhibited in a dose-dependent manner. Taken together, this study demonstrated that copper possesses modulatory effects on immune cells but not on epithelial cells, but further studies are needed to underline this hypothesis.
Collapse
Affiliation(s)
| | | | | | | | - Carsten Gründemann
- Translational Complementary Medicine, Department of Pharmaceutical Sciences, University of Basel, Campus Rosental—Mattenstrasse 22, CH-4058 Basel, Switzerland; (A.A.); (L.W.); (S.N.); (A.M.Z.-K.)
| |
Collapse
|
5
|
Opgenorth J, Goetz BM, Rodriguez-Jimenez S, Freestone AD, Combs GJ, Flemming TA, McGill JL, Gorden PJ, Tikofsky L, Baumgard LH. Comparing oral versus intravenous calcium administration on alleviating markers of production, metabolism, and inflammation during an intravenous lipopolysaccharide challenge in mid-lactation dairy cows. J Dairy Sci 2025; 108:2883-2896. [PMID: 39603495 DOI: 10.3168/jds.2024-24831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 10/28/2024] [Indexed: 11/29/2024]
Abstract
Animals, including dairy cows, develop hypocalcemia during infection. Prior independent research suggests supplementing oral Ca, but not i.v. Ca, improves multiple health metrics after immune activation. Therefore, study objectives were to directly compare the effects of administering an oral Ca bolus versus i.v. Ca on mineral and energetic metabolism variables and inflammatory parameters following an i.v. LPS challenge. Mid-lactation cows (124 ± 43 DIM) were assigned to 1 of 4 treatments: (1) saline control (CON; 4 mL of saline; n = 4), (2) LPS control (CON-LPS; 0.375 µg/kg BW; n = 6), (3) LPS with oral Ca bolus (OCa-LPS; 0.375 µg/kg BW and a 192-g bolus of Bovikalc [Boehringer Ingelheim Animal Health USA Inc., Duluth, GA] containing 43 g of Ca [71% CaCl2 and 29% CaSO4] supplemented at -0.5 and 6 h relative to LPS administration; n = 8), and (4) LPS with i.v. Ca (IVCa-LPS; 0.375 µg/kg BW and 500 mL of Ca-gluconate, 23% [VetOne, Boise, ID]) supplemented at -0.5 and 6 h relative to LPS infusion; n = 8). During period (P) 1 (4 d), baseline data were obtained. At the initiation of P2 (5 d), LPS and Ca supplements were administered. As anticipated, CON-LPS became hypocalcemic, but OCa-LPS and IVCa-LPS had increased ionized Ca compared with CON-LPS cows (1.11 and 1.28 vs. 0.95 ± 0.02 mmol/L, respectively). Rectal temperature increased after LPS and was additionally elevated in IVCa-LPS from 3 to 4 h (38.9 and 39.8 ± 0.1°C in CON-LPS and IVCa-LPS, respectively). Administering LPS decreased DMI and milk yield relative to CON. Circulating glucose was decreased in OCa-LPS compared with CON-LPS and IVCa-LPS during the initial hyperglycemic phase at 1 h (75.1 vs. 94.9 and 95.7 ± 3.4 mg/dL, respectively, but all LPS infused cows regardless of treatment had similar glucose concentrations thereafter, which were decreased relative to baseline during the first 12 h. Blood urea nitrogen increased after LPS but this was attenuated in OCa-LPS compared with CON-LPS and IVCa-LPS cows (8.7 vs. 10.0 and 10.4 ± 0.3 mg/dL). Glucagon increased in OCa-LPS and IVCa-LPS compared with CON-LPS cows (459 and 472 vs. 335 ± 28 pg/mL, respectively), and insulin markedly increased over time regardless of LPS treatment. Lipopolysaccharide substantially increased serum amyloid A, LPS-binding protein (LBP), and haptoglobin in all treatments, but OCa-LPS tended to have increased LBP concentrations relative to IVCa-LPS (10.7 vs. 8.6 ± 0.7 µg/mL, respectively). Several cytokines increased after LPS administration, but most temporal cytokine profiles did not differ by treatment. In summary, LPS administration intensely activated the immune system and both Ca delivery routes successfully ameliorated the hypocalcemia. The i.v. and oral Ca treatments had differential effects on multiple metabolism variables and appeared to mildly influence production responses to LPS.
Collapse
Affiliation(s)
- J Opgenorth
- Department of Animal Science, Iowa State University, Ames, IA 50011
| | - B M Goetz
- Department of Animal Science, Iowa State University, Ames, IA 50011
| | | | - A D Freestone
- Department of Animal Science, Iowa State University, Ames, IA 50011
| | - G J Combs
- Department of Animal Science, Iowa State University, Ames, IA 50011
| | - T A Flemming
- Department of Animal Science, Iowa State University, Ames, IA 50011
| | - J L McGill
- Department of Veterinary Microbiology and Preventative Medicine, Iowa State University, Ames, IA 50011
| | - P J Gorden
- Department of Veterinary Diagnostic and Production Animal Medicine, Iowa State University, Ames, IA 50011
| | - L Tikofsky
- Boehringer Ingelheim Animal Health USA Inc., Duluth, GA 30096
| | - L H Baumgard
- Department of Animal Science, Iowa State University, Ames, IA 50011.
| |
Collapse
|
6
|
Rakovskaya A, Volkova E, Bezprozvanny I, Pchitskaya E. Hippocampal dendritic spines store-operated calcium entry and endoplasmic reticulum content is dynamic microtubule dependent. Sci Rep 2025; 15:1314. [PMID: 39779788 PMCID: PMC11711194 DOI: 10.1038/s41598-024-85024-5] [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: 07/12/2024] [Accepted: 12/30/2024] [Indexed: 01/11/2025] Open
Abstract
One of the mechanisms of calcium signalling in neurons is store-operated calcium entry (SOCE), which is activated when the calcium concentration in the smooth endoplasmic reticulum (ER) decreases and its protein-calcium sensor STIM (stromal interacting molecule) relocate to the endoplasmic reticulum and plasma membrane junctions, forms clusters and induces calcium entry. In electrically non-excitable cells, STIM1 is coupled with the positive end of a tubulin microtubule through interaction with EB1 (end-binding) protein, which controls its oligomerization, SOCE and participates in ER movement. STIM2 homologue, which is specific for mature hippocampal dendritic spines, is known to interact with EB3 protein, however, not much is known about the role of this interaction in STIM2 clustering or ER trafficking in neurons. Intriguingly, in neurons, reducing the expression of EB3 protein or disrupting the interaction of STIM2 protein with EB proteins results in decreased SOCE, in contrast to experiments with STIM1 in non-excitable cells. In this study, these two homologues are compared side-by-side in HEK-293T, and it is shown for the first time that their clustering and SOCE is oppositely regulated by dynamic tubulin microtubules. In particular, for STIM2, the interaction with dynamic microtubule cytoskeleton is required for clustering and is shown to potentiate SOCE, while for STIM1 this interaction restricts clustering, resulting in SOCE decrease. After store depletion in primary hippocampal neurons, the wild type STIM2 is redistributed from the necks to the heads of dendritic spines, while the STIM2 variant with a mutation that disrupts the interaction with EB proteins is excluded from dendritic spines. In addition, overexpression of the mutant variant leads to ER reorganization in neuronal soma and reduction of ER presence in spines. It also leads to a reduction in the number of spines containing the spine apparatus formed by ER cisternae, as well as a reduction in dendritic spines SOCE. These effects are opposite of those detected during overexpression of the wild type STIM2. Considered together, these findings underline the important role of dynamic microtubules in regulation of neuronal SOCE and ER morphology.
Collapse
Affiliation(s)
- Anastasiya Rakovskaya
- Laboratory of Biomedical Imaging and Data Analysis, Institute of Biomedical Systems and Biotechnology, Peter the Great St. Petersburg Polytechnic University, Khlopina St. 11, St. Petersburg, Russia, 194021
- Laboratory of Molecular Neurodegeneration, Institute of Biomedical Systems and Biotechnology, Peter the Great St. Petersburg Polytechnic University, Khlopina St. 11, St. Petersburg, Russia, 194021
| | - Ekaterina Volkova
- Laboratory of Biomedical Imaging and Data Analysis, Institute of Biomedical Systems and Biotechnology, Peter the Great St. Petersburg Polytechnic University, Khlopina St. 11, St. Petersburg, Russia, 194021
| | - Ilya Bezprozvanny
- Laboratory of Molecular Neurodegeneration, Institute of Biomedical Systems and Biotechnology, Peter the Great St. Petersburg Polytechnic University, Khlopina St. 11, St. Petersburg, Russia, 194021
| | - Ekaterina Pchitskaya
- Laboratory of Biomedical Imaging and Data Analysis, Institute of Biomedical Systems and Biotechnology, Peter the Great St. Petersburg Polytechnic University, Khlopina St. 11, St. Petersburg, Russia, 194021.
- Laboratory of Molecular Neurodegeneration, Institute of Biomedical Systems and Biotechnology, Peter the Great St. Petersburg Polytechnic University, Khlopina St. 11, St. Petersburg, Russia, 194021.
| |
Collapse
|
7
|
Wang K, Liu Y, Li S, Zhao N, Qin F, Tao Y, Song Z. Unveiling the therapeutic potential and mechanisms of stanniocalcin-1 in retinal degeneration. Surv Ophthalmol 2025; 70:106-120. [PMID: 39270826 DOI: 10.1016/j.survophthal.2024.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 07/30/2024] [Accepted: 08/16/2024] [Indexed: 09/15/2024]
Abstract
Retinal degeneration (RD) is a group of ocular diseases characterized by progressive photoreceptor apoptosis and visual impairment. Mitochondrial malfunction, excessive oxidative stress, and chronic activation of neuroglia collectively contribute to the development of RD. Currently, there is a lack of efficacious therapeutic interventions for RD. Stanniocalcin-1 (STC-1) is a promising candidate molecule to decelerate photoreceptor cell death. STC-1 is a secreted calcium/phosphorus regulatory protein that exerts diverse protective effects. Accumulating evidence suggests that STC-1 protects retinal cells from ischemic injury, oxidative stress, and excessive apoptosis through enhancing the expression of uncoupling protein-2 (UCP-2). Furthermore, STC-1 exerts its antiinflammatory effects by inhibiting the activation of microglia and macrophages, as well as the synthesis and secretion of proinflammatory cytokines, such as TNF-α, IL-1, and IL-6. By employing these mechanisms, STC-1 effectively shields the retinal photoreceptors and optic nerve, thereby slowing down the progression of RD. We summarize the STC-1-mediated therapeutic effects on the degenerating retina, with a particular focus on its underlying mechanisms. These findings highlight that STC-1 may act as a versatile molecule to treat degenerative retinopathy. Further research on STC-1 is imperative to establish optimal protocols for its clinical use.
Collapse
Affiliation(s)
- Kexin Wang
- Department of Ophthalmology, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Henan Eye Institute, Henan Eye Hospital, Zhengzhou 450003, China
| | - Yashuang Liu
- Department of Ophthalmology, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Henan Eye Institute, Henan Eye Hospital, Zhengzhou 450003, China
| | - Siyu Li
- College of Medicine, Zhengzhou University, Zhengzhou 450001, China
| | - Na Zhao
- College of Medicine, Zhengzhou University, Zhengzhou 450001, China
| | - Fangyuan Qin
- Department of Ophthalmology, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Henan Eye Institute, Henan Eye Hospital, Zhengzhou 450003, China
| | - Ye Tao
- Department of Ophthalmology, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Henan Eye Institute, Henan Eye Hospital, Zhengzhou 450003, China.
| | - Zongming Song
- Department of Ophthalmology, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Henan Eye Institute, Henan Eye Hospital, Zhengzhou 450003, China.
| |
Collapse
|
8
|
Zhang B, Wen J, Li M, Wang J, Ji Z, Lv X, Usman M, Mauck J, Loor JJ, Yang W, Wang G, Ma J, Xu C. Fatty acids promote migration of CD4 + T cells through calcium release-activated calcium modulator ORAI1 sensitive glycolysis in dairy cows. J Dairy Sci 2025; 108:856-867. [PMID: 39477060 DOI: 10.3168/jds.2024-24845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 09/17/2024] [Indexed: 12/28/2024]
Abstract
Nutritional and metabolic state in dairy cows are important determinants of the immune response. During the periparturient period, a state of negative energy balance in the cow increases plasma concentrations of fatty acids (FA), which are associated with inflammation. Among immune cells, CD4+ T are able to function under high-FA conditions, but the underlying mechanisms regulating these events remain unclear. The objective of this study was to clarify the functional mechanisms of CD4+ T cells under high-FA conditions. The effects of glycolysis and calcium release-activated calcium modulator 1 (ORAI1) on migration of CD4+ T cells exposed to high FA were investigated in vivo and in vitro. The CD4+ T cells were isolated from peripheral blood of healthy (n = 9) and high-FA (n = 9) Holstein cows (average 2.5 ± 0.2 lactations [SE], 12.3 ± 0.8 DIM). In the first experiment, real-time quantitative PCR was used to assess chemokine receptors in isolated CD4+ T cells and migration capacity. The relative mRNA measurements results revealed downregulation of CCR1 and CXCR2, and upregulation of CCR2, CCR4, CCR5, CCR7, CCR8, CCR10, CXCR1, CXCR3, CXCR4, and CX3CR1. Among them, the expression of CXCR4 was relatively high. Therefore, CXCL12, a ligand chemokine of CXCR4, was an inducer of CD4+ T cell migration. The CD4+ T cells were inoculated in the upper chamber and CXCL12 (100 ng/mL, Peprotech) in RPMI1640 was added to the lower chamber and transmigrated for 3 h at 37°C and 5% CO2. The cell migration assay revealed that the migration capacity of CD4+ T cells from high-FA cows was greater. Real-time-qPCR indicated greater abundance of the glycolysis-related targets HIF1A, HK2, PKM2, Glut1, GAPDH, and LDHA and Western blotting indicated greater abundance of the glycolysis-related targets HIF1A, HK2, PKM2, Glut1, GAPDH, and LDHA in CD4+ T cells of high-FA cows. To characterize specific mechanisms of CD4+ T cell migration in vitro, cells from the spleens of 3 newborn (1 d old, 40-50 kg) healthy female Holstein calves were isolated after euthanasia. Inhibition of glycolysis attenuated the migration ability of cells, but had no effect on the protein and mRNA abundance of store-operated Ca2+ entry (SOCE)-associated calcium release-activated calcium modulator 1 (ORAI1) and stromal interaction molecule 1 (STIM1). In contrast, ORAI1 was upregulated in CD4+ T cells of cows exposed to high FA. To explore the potential mechanisms whereby an active glycolytic metabolism affects CD4+ T cells under high-FA conditions, we knocked down ORAI1 using small interfering RNA (siORAI1). Isolated CD4+ T cells from high-FA cows with the siORAI1 had an attenuated glycolytic metabolism and migration capacity. Taken together, these data suggested that calcium ions in CD4+ T cells from cows with high FA regulate glycolytic metabolism and influence cell migration at least in part by modulating ORAI1. Thus, these studies identified a novel mechanism of Ca2+ regulation of CD4+ T cell glycolytic metabolism affecting their migration through the SOCE pathway.
Collapse
Affiliation(s)
- Bingbing Zhang
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing 163319, China
| | - Jianan Wen
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing 163319, China; College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong 032699, China
| | - Ming Li
- College of Veterinary Medicine, China Agricultural University, Beijing 100000, China
| | - Jingjing Wang
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing 163319, China
| | - Ziwei Ji
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing 163319, China
| | - Xinquan Lv
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, China
| | - Muhammad Usman
- Mammalian Nutri Physio Genomics, Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, Urbana, IL 61801
| | - John Mauck
- Mammalian Nutri Physio Genomics, Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, Urbana, IL 61801
| | - Juan J Loor
- Mammalian Nutri Physio Genomics, Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, Urbana, IL 61801
| | - Wei Yang
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, China
| | - Guihua Wang
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing 163319, China
| | - Jinzhu Ma
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing 163319, China
| | - Chuang Xu
- College of Veterinary Medicine, China Agricultural University, Beijing 100000, China.
| |
Collapse
|
9
|
Granados ST, Yanushkevich S, Lok J, Concepcion AR. Analysis of Store-Operated Ca 2+ Entry in Primary T Cells. Methods Mol Biol 2025; 2904:91-113. [PMID: 40220228 DOI: 10.1007/978-1-0716-4414-0_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/14/2025]
Abstract
Calcium ions (Ca2+) are key second messengers for signal transduction in virtually all cells. In T cells, Ca2+ signals are generated upon T cell receptor (TCR) stimulation in a two-step integrated process known as Store-Operated Ca2+ Entry (SOCE), which involves the depletion of endoplasmic reticulum (ER) Ca2+ stores, followed by the influx of extracellular Ca2+ via Ca2+ release-activated Ca2+ (CRAC) channels. The Ca2+ influx generated by the opening of CRAC channels in T cells is essential for their metabolic reprogramming, proliferation, cytokine production, and adaptive immune response.In this book chapter, we review general concepts, discuss the rationale for using ratiometric Ca2+-sensitive chemical dyes to monitor SOCE in primary murine T cells, and weigh the advantages and disadvantages of the different methods that are currently available to detect cytosolic Ca2+ dynamics. We provide detailed protocols to measure SOCE in mouse T cells including flow cytometry, fluorescent microplate reader and single-cell imaging, and offer a general guideline on how to quantify SOCE in these cells. These protocols are easily adaptable to monitor cytosolic Ca2+ dynamics in human T cells and other cell types of interest.
Collapse
Affiliation(s)
- Sara T Granados
- Department of Biochemistry & Molecular Biology, University of Chicago, Chicago, IL, USA
| | - Sergei Yanushkevich
- Department of Biochemistry & Molecular Biology, University of Chicago, Chicago, IL, USA
| | - Jessica Lok
- Committee on Immunology, University of Chicago, Chicago, IL, USA
| | - Axel R Concepcion
- Department of Biochemistry & Molecular Biology, University of Chicago, Chicago, IL, USA.
- Committee on Immunology, University of Chicago, Chicago, IL, USA.
- Institute for Biophysical Dynamics, University of Chicago, Chicago, IL, USA.
- University of Chicago Medicine Comprehensive Cancer Center, Chicago, IL, USA.
| |
Collapse
|
10
|
Yang W, Feng Z, Lai X, Li J, Cao Z, Jiang F, Chen F, Zhan S, Kong F, Yang L, Teng Y, Watford WT, Zhou G, Xie J. Calcium nanoparticles target and activate T cells to enhance anti-tumor function. Nat Commun 2024; 15:10095. [PMID: 39572569 PMCID: PMC11582315 DOI: 10.1038/s41467-024-54402-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 11/08/2024] [Indexed: 11/24/2024] Open
Abstract
Calcium signaling plays a crucial role in the activation of T lymphocytes. However, modulating calcium levels to control T cell activation in vivo remains a challenge. In this study, we investigate T cell activation using 12-myristate 13-acetate (PMA)-encapsulated CaCO3 nanoparticles. We find that anti-PD-1 antibody-conjugated CaCO3 nanoparticles can be internalized by T cells via receptor-mediated endocytosis and then gradually release calcium. This results in an increase in cytosolic calcium, which triggers the activation of NFAT and NF-κB pathways, especially when the surface of the CaCO3 nanoparticles is loaded with PMA. Animal studies demonstrate that the PMA-loaded calcium nanoparticles enhance the activation and proliferation of cytotoxic T cells, leading to improved tumor suppression without additional toxicity. When tested in metastatic tumor models, T cells loaded with the calcium nanoparticles prior to adoptive cell transfer control tumor growth better, resulting in prolonged animal survival. Our approach offers an alternative T cell activation strategy to potentiate immunotherapy by targeting a fundamental signaling pathway.
Collapse
Affiliation(s)
- Wei Yang
- Department of Chemistry, University of Georgia, Athens, GA, USA
| | - Zhizi Feng
- Department of Chemistry, University of Georgia, Athens, GA, USA
| | - Xinning Lai
- Department of Chemistry, University of Georgia, Athens, GA, USA
| | - Jianwen Li
- Department of Chemistry, University of Georgia, Athens, GA, USA
| | - Zhengwei Cao
- Department of Chemistry, University of Georgia, Athens, GA, USA
| | - Fangchao Jiang
- Department of Chemistry, University of Georgia, Athens, GA, USA
| | - Fanghui Chen
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
| | - Shuyue Zhan
- Department of Chemistry, University of Georgia, Athens, GA, USA
| | - Feng Kong
- Department of Plant Pathology, University of Georgia, Athens, GA, USA
| | - Li Yang
- Department of Plant Pathology, University of Georgia, Athens, GA, USA
| | - Yong Teng
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
| | - Wendy T Watford
- Department of Infectious Diseases, University of Georgia, Athens, GA, USA
| | - Gang Zhou
- Georgia Cancer Center, Department of Medicine, Medical College of Georgia, Augusta, GA, USA.
| | - Jin Xie
- Department of Chemistry, University of Georgia, Athens, GA, USA.
| |
Collapse
|
11
|
Uddin MN, Thomas DW. SERCA Modulators Reveal Distinct Signaling and Functional Roles of T Lymphocyte Ca 2+ Stores. Int J Mol Sci 2024; 25:12095. [PMID: 39596161 PMCID: PMC11593871 DOI: 10.3390/ijms252212095] [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: 10/19/2024] [Revised: 11/04/2024] [Accepted: 11/09/2024] [Indexed: 11/28/2024] Open
Abstract
The allosteric SERCA (Sarcoplasmic/Endoplasmic Reticulum Ca2+-ATPase) activator CDN1163 has been recently added to the group of pharmacological tools for probing SERCA function. We chose to investigate the effects of the compound on T lymphocyte Ca2+ stores, using the well-described Jurkat T lymphocyte as a reliable cell system for Ca2+ signaling pathways. Our study identified the lowest concentrations of the SERCA inhibitors thapsigargin (TG) and 2,5-di-(tert butyl)-1,4-benzohydroquinone (tBHQ) capable of releasing Ca2+, permitting the differentiation of the TG-sensitive SERCA 2b Ca2+ store from the tBHQ-sensitive SERCA 3 Ca2+ store. We proceeded to test the effects of CDN1163 on Ca2+ stores, examining specific actions on the SERCA 2b and SERCA 3 Ca2+ pools using our low-dose SERCA blocker regimen. In contrast to previous work, we find CDN1163 exerts complex time-sensitive and SERCA isoform-specific actions on Ca2+ stores. Surprisingly, short-term exposure (0-30 min) to CDN1163 perturbs T cell Ca2+ stores by suppressing Ca2+ uptake with diminished Ca2+ release from the SERCA 2b-controlled store. Concomitantly, we find evidence for a SERCA-activating effect of CDN1163 on the SERCA-3 regulated store, given the observation of increased Ca2+ release inducible by low-dose tBHQ. Intriguingly, longer-term (>12 h) CDN1163 exposure reversed this pattern, with increased Ca2+ release from SERCA 2b-regulated pools yet decreased Ca2+ release responses from the tBHQ-sensitive SERCA 3 pool. Indeed, this remodeling of SERCA 2b Ca2+ stores with longer-term CDN1163 exposure also translated into the compound's ability to protect Jurkat T lymphocytes from TG but not tBHQ-induced growth suppression.
Collapse
Affiliation(s)
| | - David W. Thomas
- Department of Pharmaceutical Sciences, Thomas J. Long School of Pharmacy University of the Pacific, Stockton, CA 95211, USA;
| |
Collapse
|
12
|
Wang Z, Zeng Y, Ahmed Z, Qin H, Bhatti IA, Cao H. Calcium‐dependent antimicrobials: Nature‐inspired materials and designs. EXPLORATION (BEIJING, CHINA) 2024; 4:20230099. [PMID: 39439493 PMCID: PMC11491315 DOI: 10.1002/exp.20230099] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 02/02/2024] [Indexed: 10/25/2024]
Abstract
Bacterial infection remains a major complication answering for the failures of various implantable medical devices. Tremendous extraordinary advances have been published in the design and synthesis of antimicrobial materials addressing this issue; however, the clinical translation has largely been blocked due to the challenge of balancing the efficacy and safety of these materials. Here, calcium's biochemical features, natural roles in pathogens and the immune systems, and advanced uses in infection medications are illuminated, showing calcium is a promising target for developing implantable devices with less infection tendency. The paper gives a historical overview of biomedical uses of calcium and summarizes calcium's merits in coordination, hydration, ionization, and stereochemistry for acting as a structural former or trigger in biological systems. It focuses on the involvement of calcium in pathogens' integrity, motility, and metabolism maintenance, outlining the potential antimicrobial targets for calcium. It addresses calcium's uses in the immune systems that the authors can learn from for antimicrobial synthesis. Additionally, the advances in calcium's uses in infection medications are highlighted to sketch the future directions for developing implantable antimicrobial materials. In conclusion, calcium is at the nexus of antimicrobial defense, and future works on taking advantage of calcium in antimicrobial developments are promising in clinical translation.
Collapse
Affiliation(s)
- Zhong Wang
- Interfacial Electrochemistry and BiomaterialsSchool of Materials Science and EngineeringEast China University of Science and TechnologyShanghaiChina
| | - Yongjie Zeng
- Interfacial Electrochemistry and BiomaterialsSchool of Materials Science and EngineeringEast China University of Science and TechnologyShanghaiChina
| | - Zubair Ahmed
- Interfacial Electrochemistry and BiomaterialsSchool of Materials Science and EngineeringEast China University of Science and TechnologyShanghaiChina
| | - Hui Qin
- Department of OrthopaedicsShanghai Jiaotong University Affiliated Sixth People's HospitalShanghaiChina
| | | | - Huiliang Cao
- Interfacial Electrochemistry and BiomaterialsSchool of Materials Science and EngineeringEast China University of Science and TechnologyShanghaiChina
- Engineering Research Center for Biomedical Materials of Ministry of EducationEast China University of Science and TechnologyShanghaiChina
- Key Laboratory for Ultrafine Materials of Ministry of EducationEast China University of Science & TechnologyShanghaiChina
| |
Collapse
|
13
|
Jakobs J, Bertram J, Rink L. Ca 2+ signals are essential for T-cell proliferation, while Zn 2+ signals are necessary for T helper cell 1 differentiation. Cell Death Discov 2024; 10:336. [PMID: 39043646 PMCID: PMC11266428 DOI: 10.1038/s41420-024-02104-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 07/11/2024] [Accepted: 07/16/2024] [Indexed: 07/25/2024] Open
Abstract
The regulation of T-cell fate is crucial for the balance between infection control and tolerance. Calcium (Ca2+) and zinc (Zn2+) signals are both induced after T-cell stimulation, but their specific roles in the fate of activation and differentiation remain to be elucidated. Are Zn2+- and Ca2+ signals responsible for different aspects in T-cell activation and differentiation and do they act in concert or in opposition? It is crucial to understand the interplay of the intracellular signals to influence the fate of T cells in diseases with undesirable T-cell activities or in Zn2+-deficient patients. Human peripheral blood mononuclear cells were stimulated with the Zn2+ ionophore pyrithione and thapsigargin, an inhibitor of the sarcoplasmic/endoplasmic reticulum Ca2+ ATPase (SERCA). Intracellular Zn2+ and Ca2+ signals were monitored by flow cytometry and ELISA, quantitative PCR and western blot were used to evaluate T-cell differentiation and the underlying molecular mechanism. We found that Zn2+ signals upregulated the early T-cell activation marker CD69, interferon regulatory factor 1 (IRF-1), and Krüppel-like factor 10 (KLF-10) expression, which are important for T helper cell (Th) 1 differentiation. Ca2+ signals, on the other hand, increased T-bet and Forkhead box P3 (FoxP3) expression and interleukin (IL)-2 release. Most interestingly, the combination of Zn2+ and Ca2+ signals was indispensable to induce interferon (IFN)-γ expression and increased the surface expression of CD69 by several-fold. These results highlight the importance of the parallel occurrence of Ca2+ and Zn2+ signals. Both signals act in concert and are required for the differentiation into Th1 cells, for the stabilization of regulatory T cells, and induces T-cell activation by several-fold. This provides further insight into the impaired immune functions of patients with zinc deficiency.
Collapse
Affiliation(s)
- Jana Jakobs
- Institute of Immunology, Faculty of Medicine, RWTH Aachen University Hospital, Aachen, Germany
| | - Jens Bertram
- Institute for Occupational, Social and Environmental Medicine, Faculty of Medicine, RWTH Aachen University, Aachen, Germany
| | - Lothar Rink
- Institute of Immunology, Faculty of Medicine, RWTH Aachen University Hospital, Aachen, Germany.
| |
Collapse
|
14
|
Shi J, Yin W, Chen W. Mathematical models of TCR initial triggering. Front Immunol 2024; 15:1411614. [PMID: 39091495 PMCID: PMC11291225 DOI: 10.3389/fimmu.2024.1411614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 07/05/2024] [Indexed: 08/04/2024] Open
Abstract
T cell receptors (TCRs) play crucial roles in regulating T cell response by rapidly and accurately recognizing foreign and non-self antigens. The process involves multiple molecules and regulatory mechanisms, forming a complex network to achieve effective antigen recognition. Mathematical modeling techniques can help unravel the intricate network of TCR signaling and identify key regulators that govern it. In this review, we introduce and briefly discuss relevant mathematical models of TCR initial triggering, with a focus on kinetic proofreading (KPR) models with different modified structures. We compare the topology structures, biological hypotheses, parameter choices, and simulation performance of each model, and summarize the advantages and limitations of them. Further studies on TCR modeling design, aiming for an optimized balance of specificity and sensitivity, are expected to contribute to the development of new therapeutic strategies.
Collapse
Affiliation(s)
- Jiawei Shi
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory for Biomedical Engineering of the Ministry of Education, College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, China
| | - Weiwei Yin
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Zhejiang Provincial Key Laboratory of Cardio-Cerebral Vascular Detection Technology and Medicinal Effectiveness Appraisal, Zhejiang University, Hangzhou, China
| | - Wei Chen
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory for Biomedical Engineering of the Ministry of Education, College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, China
- Department of Cell Biology, School of Medicine, Zhejiang University, Hangzhou, China
- Liangzhu Laboratory, Zhejiang University, Hangzhou, China
| |
Collapse
|
15
|
Haghmorad D, Soltanmohammadi A, Jadid Tavaf M, Zargarani S, Yazdanpanah E, Shadab A, Yousefi B. The protective role of interaction between vitamin D, sex hormones and calcium in multiple sclerosis. Int J Neurosci 2024; 134:735-753. [PMID: 36369838 DOI: 10.1080/00207454.2022.2147431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Revised: 11/01/2022] [Accepted: 11/07/2022] [Indexed: 11/14/2022]
Abstract
Multiple sclerosis (MS) is a neurological disorder that causes disability and paralysis, especially among young adults. Although interactions of several factors, such as viral infections, autoimmunity, genetic and environmental factors, performance a role in the beginning and progression of the disease, the exact cause of MS is unknown to date. Different immune cells such as Th1 and Th17 play an impressive role in the immunopathogenesis of MS, while, regulatory cells such as Th2 and Treg diminish the severity of the illness. Sex hormones have a vital role in many autoimmune disorders, including multiple sclerosis. Testosterone, estrogen and progesterone have various roles in the progress of MS, which higher prevalence of disease in women and more severe in men reveals the importance of sex hormones' role in this disease. Vitamin D after chemical changes in the body, as an active hormone called calcitriol, plays an important role in regulating immune responses and improves MS by modulating the immune system. The optimum level of calcium in the body with vitamin D modulates immune responses and calcium as an essential ion in the body plays a key role in the treatment of autoimmune diseases. The interaction between vitamin D and sex hormones has protective and therapeutic effects against MS and functional synergy between estrogen and calcitriol occurs in disease recovery. Moreover, vitamin D and calcium interact with each other to regulate the immune system and shift them to anti-inflammatory responses.
Collapse
Affiliation(s)
- Dariush Haghmorad
- Cancer Research Center, Semnan University of Medical Sciences, Semnan, Iran
- Department of Immunology, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Azita Soltanmohammadi
- Department of Immunology, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Maryam Jadid Tavaf
- Department of Immunology, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Simin Zargarani
- Department of Immunology, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Esmaeil Yazdanpanah
- Immunology Research Center, Department of Immunology and Allergy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Alireza Shadab
- Department of Immunology, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Bahman Yousefi
- Cancer Research Center, Semnan University of Medical Sciences, Semnan, Iran
- Department of Immunology, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| |
Collapse
|
16
|
Jakobs J, Rink L. Zinc Ionophore Pyrithione Mimics CD28 Costimulatory Signal in CD3 Activated T Cells. Int J Mol Sci 2024; 25:4302. [PMID: 38673887 PMCID: PMC11050009 DOI: 10.3390/ijms25084302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 04/10/2024] [Accepted: 04/11/2024] [Indexed: 04/28/2024] Open
Abstract
Zinc is an essential trace element that plays a crucial role in T cell immunity. During T cell activation, zinc is not only structurally important, but zinc signals can also act as a second messenger. This research investigates zinc signals in T cell activation and their function in T helper cell 1 differentiation. For this purpose, peripheral blood mononuclear cells were activated via the T cell receptor-CD3 complex, and via CD28 as a costimulatory signal. Fast and long-term changes in intracellular zinc and calcium were monitored by flow cytometry. Further, interferon (IFN)-γ was analyzed to investigate the differentiation into T helper 1 cells. We show that fast zinc fluxes are induced via CD3. Also, the intracellular zinc concentration dramatically increases 72 h after anti-CD3 and anti-CD28 stimulation, which goes along with the high release of IFN-γ. Interestingly, we found that zinc signals can function as a costimulatory signal for T helper cell 1 differentiation when T cells are activated only via CD3. These results demonstrate the importance of zinc signaling alongside calcium signaling in T cell differentiation.
Collapse
Affiliation(s)
| | - Lothar Rink
- Institute of Immunology, Faculty of Medicine, RWTH Aachen University, Pauwelsstraße 30, 52074 Aachen, Germany;
| |
Collapse
|
17
|
Chan D, Cromar GL, Taj B, Parkinson J. Cell4D: a general purpose spatial stochastic simulator for cellular pathways. BMC Bioinformatics 2024; 25:121. [PMID: 38515063 PMCID: PMC10956314 DOI: 10.1186/s12859-024-05739-0] [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: 09/13/2023] [Accepted: 03/11/2024] [Indexed: 03/23/2024] Open
Abstract
BACKGROUND With the generation of vast compendia of biological datasets, the challenge is how best to interpret 'omics data alongside biochemical and other small-scale experiments to gain meaningful biological insights. Key to this challenge are computational methods that enable domain-users to generate novel hypotheses that can be used to guide future experiments. Of particular interest are flexible modeling platforms, capable of simulating a diverse range of biological systems with low barriers of adoption to those with limited computational expertise. RESULTS We introduce Cell4D, a spatial-temporal modeling platform combining a robust simulation engine with integrated graphics visualization, a model design editor, and an underlying XML data model capable of capturing a variety of cellular functions. Cell4D provides an interactive visualization mode, allowing intuitive feedback on model behavior and exploration of novel hypotheses, together with a non-graphics mode, compatible with high performance cloud compute solutions, to facilitate generation of statistical data. To demonstrate the flexibility and effectiveness of Cell4D, we investigate the dynamics of CEACAM1 localization in T-cell activation. We confirm the importance of Ca2+ microdomains in activating calmodulin and highlight a key role of activated calmodulin on the surface expression of CEACAM1. We further show how lymphocyte-specific protein tyrosine kinase can help regulate this cell surface expression and exploit spatial modeling features of Cell4D to test the hypothesis that lipid rafts regulate clustering of CEACAM1 to promote trans-binding to neighbouring cells. CONCLUSIONS Through demonstrating its ability to test and generate hypotheses, Cell4D represents an effective tool to help integrate knowledge across diverse, large and small-scale datasets.
Collapse
Affiliation(s)
- Donny Chan
- Program in Molecular Medicine, Hospital for Sick Children, Toronto, M5G 0A4, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, M5S 1A8, Canada
| | - Graham L Cromar
- Program in Molecular Medicine, Hospital for Sick Children, Toronto, M5G 0A4, Canada
| | - Billy Taj
- Program in Molecular Medicine, Hospital for Sick Children, Toronto, M5G 0A4, Canada
| | - John Parkinson
- Program in Molecular Medicine, Hospital for Sick Children, Toronto, M5G 0A4, Canada.
- Department of Molecular Genetics, University of Toronto, Toronto, M5S 1A8, Canada.
- Department of Biochemistry, University of Toronto, Toronto, M5S 1A8, Canada.
| |
Collapse
|
18
|
Kar A, Ghosh P, Gautam A, Chowdhury S, Basak D, Sarkar I, Bhoumik A, Barman S, Chakraborty P, Mukhopadhyay A, Mehrotra S, Ganesan SK, Paul S, Chatterjee S. CD38-RyR2 axis-mediated signaling impedes CD8 + T cell response to anti-PD1 therapy in cancer. Proc Natl Acad Sci U S A 2024; 121:e2315989121. [PMID: 38451948 PMCID: PMC10945783 DOI: 10.1073/pnas.2315989121] [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: 09/14/2023] [Accepted: 01/08/2024] [Indexed: 03/09/2024] Open
Abstract
PD1 blockade therapy, harnessing the cytotoxic potential of CD8+ T cells, has yielded clinical success in treating malignancies. However, its efficacy is often limited due to the progressive differentiation of intratumoral CD8+ T cells into a hypofunctional state known as terminal exhaustion. Despite identifying CD8+ T cell subsets associated with immunotherapy resistance, the molecular pathway triggering the resistance remains elusive. Given the clear association of CD38 with CD8+ T cell subsets resistant to anti-PD1 therapy, we investigated its role in inducing resistance. Phenotypic and functional characterization, along with single-cell RNA sequencing analysis of both in vitro chronically stimulated and intratumoral CD8+ T cells, revealed that CD38-expressing CD8+ T cells are terminally exhausted. Exploring the molecular mechanism, we found that CD38 expression was crucial in promoting terminal differentiation of CD8+ T cells by suppressing TCF1 expression, thereby rendering them unresponsive to anti-PD1 therapy. Genetic ablation of CD38 in tumor-reactive CD8+ T cells restored TCF1 levels and improved the responsiveness to anti-PD1 therapy in mice. Mechanistically, CD38 expression on exhausted CD8+ T cells elevated intracellular Ca2+ levels through RyR2 calcium channel activation. This, in turn, promoted chronic AKT activation, leading to TCF1 loss. Knockdown of RyR2 or inhibition of AKT in CD8+ T cells maintained TCF1 levels, induced a sustained anti-tumor response, and enhanced responsiveness to anti-PD1 therapy. Thus, targeting CD38 represents a potential strategy to improve the efficacy of anti-PD1 treatment in cancer.
Collapse
Affiliation(s)
- Anwesha Kar
- Division of Cancer Biology and Inflammatory Disorder, Translational Research Unit of Excellence, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology, Kolkata700032, West Bengal, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad201002, Uttar Pradesh, India
| | - Puspendu Ghosh
- Division of Cancer Biology and Inflammatory Disorder, Translational Research Unit of Excellence, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology, Kolkata700032, West Bengal, India
| | - Anupam Gautam
- Algorithms in Bioinformatics, Institute for Bioinformatics and Medical Informatics, University of Tübingen, Sand 1472076, Tübingen, Baden-Württemberg, Germany
- International Max Planck Research School “From Molecules to Organisms”, Max Planck Institute for Biology Tübingen72076, Tübingen, Baden-Württemberg, Germany
| | - Snehanshu Chowdhury
- Division of Cancer Biology and Inflammatory Disorder, Translational Research Unit of Excellence, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology, Kolkata700032, West Bengal, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad201002, Uttar Pradesh, India
| | - Debashree Basak
- Division of Cancer Biology and Inflammatory Disorder, Translational Research Unit of Excellence, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology, Kolkata700032, West Bengal, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad201002, Uttar Pradesh, India
| | - Ishita Sarkar
- Division of Cancer Biology and Inflammatory Disorder, Translational Research Unit of Excellence, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology, Kolkata700032, West Bengal, India
| | - Arpita Bhoumik
- Division of Cancer Biology and Inflammatory Disorder, Translational Research Unit of Excellence, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology, Kolkata700032, West Bengal, India
| | - Shubhrajit Barman
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad201002, Uttar Pradesh, India
- Division of Structural Biology & Bioinformatics, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology, Kolkata700032, West Bengal, India
| | - Paramita Chakraborty
- Department of Surgery, Medical University of South Carolina, Charleston, South CarolinaSC- 29425
| | - Asima Mukhopadhyay
- Kolkata Gynaecology Oncology Trials and Translational Research Group, Kolkata700156, West Bengal, India
| | - Shikhar Mehrotra
- Department of Surgery, Medical University of South Carolina, Charleston, South CarolinaSC- 29425
| | - Senthil Kumar Ganesan
- Division of Structural Biology & Bioinformatics, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology, Kolkata700032, West Bengal, India
| | - Sandip Paul
- System Biology Informatics Lab, Center for Health Science and Technology, JIS Institute of Advanced Studies and Research, JIS University, Kolkata700091, West Bengal, India
| | - Shilpak Chatterjee
- Division of Cancer Biology and Inflammatory Disorder, Translational Research Unit of Excellence, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology, Kolkata700032, West Bengal, India
| |
Collapse
|
19
|
Petersen SH, Al Badawy K, Hopkins R, Vu DL, Rahmani M, Maia SM, Connolly JE. A novel GPI-anchored dominant-negative TGF-β receptor II renders T cells unresponsive to TGF-β signaling. Mol Ther Oncolytics 2023; 31:100730. [PMID: 37829123 PMCID: PMC10565558 DOI: 10.1016/j.omto.2023.100730] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 09/21/2023] [Indexed: 10/14/2023] Open
Abstract
Transforming growth factor β (TGF-β) is a pleiotropic cytokine expressed by a wide range of cell types and is known for hampering the effectiveness of cancer immune cell therapeutic approaches. We have designed a novel construct containing the extracellular domain of the TGF-β receptor II linked to a glycosylphosphatidylinositol (GPI) anchor (GPI-ecto-TβRII) lacking the transmembrane and cytoplasmic signaling domain of TGF-β receptor II (TβRII). T cells transduced with lentivirus expressing the GPI-ecto-TβRII construct show 5 to 15 times higher membrane expression compared with a previously established dominant-negative receptor carrying a truncated signaling domain. GPI-ecto-TβRII expression renders T cells unresponsive to TGF-β-induced signaling seen by a lack of SMAD phosphorylation upon exogeneous TGF-β treatment. Transduced T cells continue to express high levels of IFNγ and granulocyte-macrophage colony-stimulating factor (GM-CSF), among other cytokines, in the presence of TGF-β while cytokine expression in untransduced T cells is being markedly suppressed. Furthermore, T cells expressing GPI-ecto-TβRII constructs have been shown to efficiently capture and inactivate TGF-β from their environment. These results indicate the potential benefits of GPI-ecto-TβRII expressing cytotoxic T cells (CTLs) in future cell therapies.
Collapse
Affiliation(s)
| | | | | | - Dang L. Vu
- Tessa Therapeutics, Singapore, Singapore
- Program in Translational Immunology, Institute of Molecular and Cell Biology, A∗STAR, Singapore, Singapore
| | | | - Sonia M.P. Maia
- Tessa Therapeutics, Singapore, Singapore
- Program in Translational Immunology, Institute of Molecular and Cell Biology, A∗STAR, Singapore, Singapore
| | - John E. Connolly
- Tessa Therapeutics, Singapore, Singapore
- Program in Translational Immunology, Institute of Molecular and Cell Biology, A∗STAR, Singapore, Singapore
- Department of Microbiology and Immunity, National University of Singapore, Singapore, Singapore
- Institute of Biomedical Studies, Baylor University Medical Center, Waco, TX, USA
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
| |
Collapse
|
20
|
Mackiewicz J, Lisek M, Boczek T. Targeting CaN/NFAT in Alzheimer's brain degeneration. Front Immunol 2023; 14:1281882. [PMID: 38077352 PMCID: PMC10701682 DOI: 10.3389/fimmu.2023.1281882] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 11/06/2023] [Indexed: 12/18/2023] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder characterized by a progressive loss of cognitive functions. While the exact causes of this debilitating disorder remain elusive, numerous investigations have characterized its two core pathologies: the presence of β-amyloid plaques and tau tangles. Additionally, multiple studies of postmortem brain tissue, as well as results from AD preclinical models, have consistently demonstrated the presence of a sustained inflammatory response. As the persistent immune response is associated with neurodegeneration, it became clear that it may also exacerbate other AD pathologies, providing a link between the initial deposition of β-amyloid plaques and the later development of neurofibrillary tangles. Initially discovered in T cells, the nuclear factor of activated T-cells (NFAT) is one of the main transcription factors driving the expression of inflammatory genes and thus regulating immune responses. NFAT-dependent production of inflammatory mediators is controlled by Ca2+-dependent protein phosphatase calcineurin (CaN), which dephosphorylates NFAT and promotes its transcriptional activity. A substantial body of evidence has demonstrated that aberrant CaN/NFAT signaling is linked to several pathologies observed in AD, including neuronal apoptosis, synaptic deficits, and glia activation. In view of this, the role of NFAT isoforms in AD has been linked to disease progression at different stages, some of which are paralleled to diminished cognitive status. The use of classical inhibitors of CaN/NFAT signaling, such as tacrolimus or cyclosporine, or adeno-associated viruses to specifically inhibit astrocytic NFAT activation, has alleviated some symptoms of AD by diminishing β-amyloid neurotoxicity and neuroinflammation. In this article, we discuss the recent findings related to the contribution of CaN/NFAT signaling to the progression of AD and highlight the possible benefits of targeting this pathway in AD treatment.
Collapse
Affiliation(s)
| | | | - Tomasz Boczek
- Department of Molecular Neurochemistry, Medical University of Lodz, Lodz, Poland
| |
Collapse
|
21
|
Ren R, Li Y. STIM1 in tumor cell death: angel or devil? Cell Death Discov 2023; 9:408. [PMID: 37932320 PMCID: PMC10628139 DOI: 10.1038/s41420-023-01703-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 10/21/2023] [Accepted: 10/27/2023] [Indexed: 11/08/2023] Open
Abstract
Stromal interaction molecule 1 (STIM1) is involved in mediating the store-operated Ca2+ entry (SOCE), driving the influx of the intracellular second messenger calcium ion (Ca2+), which is closely associated with tumor cell proliferation, metastasis, apoptosis, autophagy, metabolism and immune processes. STIM1 is not only regulated at the transcriptional level by NF-κB and HIF-1, but also post-transcriptionally modified by miRNAs and degraded by ubiquitination. Recent studies have shown that STIM1 or Ca2+ signaling can regulate apoptosis, autophagy, pyroptosis, and ferroptosis in tumor cells and act discrepantly in different cancers. Furthermore, STIM1 contributes to resistance against antitumor therapy by influencing tumor cell death. Further investigation into the mechanisms through which STIM1 controls other forms of tumor cell death could aid in the discovery of novel therapeutic targets. Moreover, STIM1 has the ability to regulate immune cells within the tumor microenvironment. Here, we review the basic structure, function and regulation of STIM1, summarize the signaling pathways through which STIM1 regulates tumor cell death, and propose the prospects of antitumor therapy by targeting STIM1.
Collapse
Affiliation(s)
- Ran Ren
- Chongqing University Cancer Hospital, School of Medicine, Chongqing University, 400044, Chongqing, China
| | - Yongsheng Li
- Chongqing University Cancer Hospital, School of Medicine, Chongqing University, 400044, Chongqing, China.
- Department of Medical Oncology, Chongqing University Cancer Hospital, 400030, Chongqing, China.
| |
Collapse
|
22
|
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
|
23
|
Ye Y, Morita S, Chang JJ, Buckley PM, Wilhelm KB, DiMaio D, Groves JT, Barrera FN. Allosteric inhibition of the T cell receptor by a designed membrane ligand. eLife 2023; 12:e82861. [PMID: 37796108 PMCID: PMC10554751 DOI: 10.7554/elife.82861] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 09/20/2023] [Indexed: 10/06/2023] Open
Abstract
The T cell receptor (TCR) is a complex molecular machine that directs the activation of T cells, allowing the immune system to fight pathogens and cancer cells. Despite decades of investigation, the molecular mechanism of TCR activation is still controversial. One of the leading activation hypotheses is the allosteric model. This model posits that binding of pMHC at the extracellular domain triggers a dynamic change in the transmembrane (TM) domain of the TCR subunits, which leads to signaling at the cytoplasmic side. We sought to test this hypothesis by creating a TM ligand for TCR. Previously we described a method to create a soluble peptide capable of inserting into membranes and binding to the TM domain of the receptor tyrosine kinase EphA2 (Alves et al., eLife, 2018). Here, we show that the approach is generalizable to complex membrane receptors, by designing a TM ligand for TCR. We observed that the designed peptide caused a reduction of Lck phosphorylation of TCR at the CD3ζ subunit in T cells. As a result, in the presence of this peptide inhibitor of TCR (PITCR), the proximal signaling cascade downstream of TCR activation was significantly dampened. Co-localization and co-immunoprecipitation in diisobutylene maleic acid (DIBMA) native nanodiscs confirmed that PITCR was able to bind to the TCR. AlphaFold-Multimer predicted that PITCR binds to the TM region of TCR, where it interacts with the two CD3ζ subunits. Our results additionally indicate that PITCR disrupts the allosteric changes in the compactness of the TM bundle that occur upon TCR activation, lending support to the allosteric TCR activation model. The TCR inhibition achieved by PITCR might be useful to treat inflammatory and autoimmune diseases and to prevent organ transplant rejection, as in these conditions aberrant activation of TCR contributes to disease.
Collapse
Affiliation(s)
- Yujie Ye
- Department of Biochemistry & Cellular and Molecular Biology, University of Tennessee at KnoxvilleKnoxvilleUnited States
| | - Shumpei Morita
- Department of Chemistry, University of California, BerkeleyBerkeleyUnited States
| | - Justin J Chang
- Department of Genetics, Yale UniversityNew HavenUnited States
| | - Patrick M Buckley
- Department of Microbial Pathogenesis, Yale UniversityNew HavenUnited States
| | - Kiera B Wilhelm
- Department of Chemistry, University of California, BerkeleyBerkeleyUnited States
| | - Daniel DiMaio
- Department of Genetics, Yale UniversityNew HavenUnited States
| | - Jay T Groves
- Department of Chemistry, University of California, BerkeleyBerkeleyUnited States
- Institute for Digital Molecular Analytics and Science, Nanyang Technological UniversitySingaporeSingapore
| | - Francisco N Barrera
- Department of Biochemistry & Cellular and Molecular Biology, University of Tennessee at KnoxvilleKnoxvilleUnited States
| |
Collapse
|
24
|
Zhang S, Zhao L, Guo M, Liu P, Li S, Xie W, Tian AL, Pol JG, Chen H, Pan H, Mao M, Li Y, Zitvogel L, Jin Y, Kepp O, Kroemer G. Anticancer effects of ikarugamycin and astemizole identified in a screen for stimulators of cellular immune responses. J Immunother Cancer 2023; 11:e006785. [PMID: 37419511 PMCID: PMC10347457 DOI: 10.1136/jitc-2023-006785] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/21/2023] [Indexed: 07/09/2023] Open
Abstract
BACKGROUND Most immunotherapies approved for clinical use rely on the use of recombinant proteins and cell-based approaches, rendering their manufacturing expensive and logistics onerous. The identification of novel small molecule immunotherapeutic agents might overcome such limitations. METHOD For immunopharmacological screening campaigns, we built an artificial miniature immune system in which dendritic cells (DCs) derived from immature precursors present MHC (major histocompatibility complex) class I-restricted antigen to a T-cell hybridoma that then secretes interleukin-2 (IL-2). RESULTS The screening of three drug libraries relevant to known signaling pathways, FDA (Food and Drug Administration)-approved drugs and neuroendocrine factors yielded two major hits, astemizole and ikarugamycin. Mechanistically, ikarugamycin turned out to act on DCs to inhibit hexokinase 2, hence stimulating their antigen presenting potential. In contrast, astemizole acts as a histamine H1 receptor (H1R1) antagonist to activate T cells in a non-specific, DC-independent fashion. Astemizole induced the production of IL-2 and interferon-γ (IFN-γ) by CD4+ and CD8+ T cells both in vitro and in vivo. Both ikarugamycin and astemizole improved the anticancer activity of the immunogenic chemotherapeutic agent oxaliplatin in a T cell-dependent fashion. Of note, astemizole enhanced the CD8+/Foxp3+ ratio in the tumor immune infiltrate as well as IFN-γ production by local CD8+ T lymphocytes. In patients with cancer, high H1R1 expression correlated with low infiltration by TH1 cells, as well as with signs of T-cell exhaustion. The combination of astemizole and oxaliplatin was able to cure the majority of mice bearing orthotopic non-small cell lung cancers (NSCLC), then inducing a state of protective long-term immune memory. The NSCLC-eradicating effect of astemizole plus oxaliplatin was lost on depletion of either CD4+ or CD8+ T cells, as well as on neutralization of IFN-γ. CONCLUSIONS These findings underscore the potential utility of this screening system for the identification of immunostimulatory drugs with anticancer effects.
Collapse
Affiliation(s)
- Shuai Zhang
- Metabolomics and Cell Biology Platforms, Gustave Roussy, Villejuif, France
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Faculté de Médecine, Université de Paris Saclay, Kremlin Bicêtre, France
| | - Liwei Zhao
- Metabolomics and Cell Biology Platforms, Gustave Roussy, Villejuif, France
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
| | - Mengfei Guo
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Peng Liu
- Metabolomics and Cell Biology Platforms, Gustave Roussy, Villejuif, France
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
| | - Sijing Li
- Metabolomics and Cell Biology Platforms, Gustave Roussy, Villejuif, France
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Faculté de Médecine, Université de Paris Saclay, Kremlin Bicêtre, France
| | - Wei Xie
- Cell death and Inflammation Unit, VIB-UGent Center for Inflammation Research, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Ai-Ling Tian
- Metabolomics and Cell Biology Platforms, Gustave Roussy, Villejuif, France
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
| | - Jonathan G Pol
- Metabolomics and Cell Biology Platforms, Gustave Roussy, Villejuif, France
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
| | - Hui Chen
- Metabolomics and Cell Biology Platforms, Gustave Roussy, Villejuif, France
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Faculté de Médecine, Université de Paris Saclay, Kremlin Bicêtre, France
| | - Hui Pan
- Metabolomics and Cell Biology Platforms, Gustave Roussy, Villejuif, France
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Faculté de Médecine, Université de Paris Saclay, Kremlin Bicêtre, France
| | - Misha Mao
- Metabolomics and Cell Biology Platforms, Gustave Roussy, Villejuif, France
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Faculté de Médecine, Université de Paris Saclay, Kremlin Bicêtre, France
- Surgical Oncology Department, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yumei Li
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Laurence Zitvogel
- INSERM U1015, Equipe labellisée par la Ligue contre le cancer, Gustave Roussy, Villjuif, France
- ClinicObiome, Gustave-Roussy, Villejuif, France
| | - Yang Jin
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Oliver Kepp
- Metabolomics and Cell Biology Platforms, Gustave Roussy, Villejuif, France
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
| | - Guido Kroemer
- Metabolomics and Cell Biology Platforms, Gustave Roussy, Villejuif, France
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Institut du Cancer Paris CARPEM, Department of Biology, Hôpital Européen Georges Pompidou, AP-HP, Paris, France
| |
Collapse
|
25
|
Angot L, Schneider P, Vannier JP, Abdoul-Azize S. Beyond Corticoresistance, A Paradoxical Corticosensitivity Induced by Corticosteroid Therapy in Pediatric Acute Lymphoblastic Leukemias. Cancers (Basel) 2023; 15:2812. [PMID: 37345151 DOI: 10.3390/cancers15102812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 05/10/2023] [Accepted: 05/16/2023] [Indexed: 06/23/2023] Open
Abstract
Known as a key effector in relapse of acute lymphoblastic leukemia (ALL), resistance to drug-induced apoptosis, is tightly considered one of the main prognostic factors for the disease. ALL cells are constantly developing cellular strategies to survive and resist therapeutic drugs. Glucocorticoids (GCs) are one of the most important agents used in the treatment of ALL due to their ability to induce cell death. The mechanisms of GC resistance of ALL cells are largely unknown and intense research is currently focused on this topic. Such resistance can involve different cellular and molecular mechanisms, including the modulation of signaling pathways involved in the regulation of proliferation, apoptosis, autophagy, metabolism, epigenetic modifications and tumor suppressors. Recently, several studies point to the paradoxical role of GCs in many survival processes that may lead to therapy-induced resistance in ALL cells, which we called "paradoxical corticosensitivity". In this review, we aim to summarize all findings on cell survival pathways paradoxically activated by GCs with an emphasis on previous and current knowledge on gene expression and signaling pathways.
Collapse
Affiliation(s)
- Laure Angot
- Normandie University, UNIROUEN, IRIB, Inserm, U1234, 76183 Rouen, France
| | - Pascale Schneider
- Normandie University, UNIROUEN, IRIB, Inserm, U1234, 76183 Rouen, France
- Department of Pediatric Immuno-Hemato-Oncology, Rouen University Hospital, 76038 Rouen, France
| | | | | |
Collapse
|
26
|
Zöphel S, Schäfer G, Nazarieh M, Konetzki V, Hoxha C, Meese E, Hoth M, Helms V, Hamed M, Schwarz EC. Identification of molecular candidates which regulate calcium-dependent CD8 + T-cell cytotoxicity. Mol Immunol 2023; 157:202-213. [PMID: 37075611 DOI: 10.1016/j.molimm.2023.04.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 01/10/2023] [Accepted: 04/02/2023] [Indexed: 04/21/2023]
Abstract
Cytotoxic CD8+ T lymphocytes (CTL) eliminate infected cells or transformed tumor cells by releasing perforin-containing cytotoxic granules at the immunological synapse. The secretion of such granules depends on Ca2+-influx through store operated Ca2+ channels, formed by STIM (stromal interaction molecule)-activated Orai proteins. Whereas molecular mechanisms of the secretion machinery are well understood, much less is known about the molecular machinery that regulates the efficiency of Ca2+-dependent target cell killing. CTL killing efficiency is of high interest considering the number of studies on CD8+ T lymphocytes modified for clinical use. Here, we isolated total RNA from primary human cells: natural killer (NK) cells, non-stimulated CD8+ T-cells, and from Staphylococcus aureus enterotoxin A (SEA) stimulated CD8+ T-cells (SEA-CTL) and conducted whole genome expression profiling by microarray experiments. Based on differential expression analysis of the transcriptome data and analysis of master regulator genes, we identified 31 candidates which potentially regulate Ca2+-homeostasis in CTL. To investigate a putative function of these candidates in CTL cytotoxicity, we transfected either SEA-stimulated CTL (SEA-CTL) or antigen specific CD8+ T-cell clones (CTL-MART-1) with siRNAs specific against the identified candidates and analyzed the killing capacity using a real-time killing assay. In addition, we complemented the analysis by studying the effect of inhibitory substances acting on the candidate proteins if available. Finally, to unmask their involvement in Ca2+ dependent cytotoxicity, candidates were also analyzed under Ca2+-limiting conditions. Overall, we identified four hits, CCR5 (C-C chemokine receptor type five), KCNN4 (potassium calcium-activated channel subfamily N), RCAN3 (regulator of calcineurin) and BCL (B-cell lymphoma) 2 which clearly affect the efficiency of Ca2+ dependent cytotoxicity in CTL-MART-1 cells, CCR5, BCL2, and KCNN4 in a positive manner, and RCAN3 in a negative way.
Collapse
Affiliation(s)
- Sylvia Zöphel
- Biophysics, Center for Integrative Physiology and Molecular Medicine, School of Medicine, Saarland University, Building 48, 66421 Homburg, Germany
| | - Gertrud Schäfer
- Biophysics, Center for Integrative Physiology and Molecular Medicine, School of Medicine, Saarland University, Building 48, 66421 Homburg, Germany
| | - Maryam Nazarieh
- Center for Bioinformatics, Saarland Informatics Campus, Saarland University, 66041 Saarbrücken, Germany
| | - Verena Konetzki
- Biophysics, Center for Integrative Physiology and Molecular Medicine, School of Medicine, Saarland University, Building 48, 66421 Homburg, Germany
| | - Cora Hoxha
- Biophysics, Center for Integrative Physiology and Molecular Medicine, School of Medicine, Saarland University, Building 48, 66421 Homburg, Germany
| | - Eckart Meese
- Human Genetics, School of Medicine, Saarland University, Building 60, 66421 Homburg, Germany
| | - Markus Hoth
- Biophysics, Center for Integrative Physiology and Molecular Medicine, School of Medicine, Saarland University, Building 48, 66421 Homburg, Germany
| | - Volkhard Helms
- Center for Bioinformatics, Saarland Informatics Campus, Saarland University, 66041 Saarbrücken, Germany
| | - Mohamed Hamed
- Institute for Biostatistics and Informatics in Medicine and Ageing Research, Rostock University Medical Centre, 18057 Rostock, Germany
| | - Eva C Schwarz
- Biophysics, Center for Integrative Physiology and Molecular Medicine, School of Medicine, Saarland University, Building 48, 66421 Homburg, Germany.
| |
Collapse
|
27
|
Zani F, Blagih J, Gruber T, Buck MD, Jones N, Hennequart M, Newell CL, Pilley SE, Soro-Barrio P, Kelly G, Legrave NM, Cheung EC, Gilmore IS, Gould AP, Garcia-Caceres C, Vousden KH. The dietary sweetener sucralose is a negative modulator of T cell-mediated responses. Nature 2023; 615:705-711. [PMID: 36922598 PMCID: PMC10033444 DOI: 10.1038/s41586-023-05801-6] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 02/06/2023] [Indexed: 03/17/2023]
Abstract
Artificial sweeteners are used as calorie-free sugar substitutes in many food products and their consumption has increased substantially over the past years1. Although generally regarded as safe, some concerns have been raised about the long-term safety of the consumption of certain sweeteners2-5. In this study, we show that the intake of high doses of sucralose in mice results in immunomodulatory effects by limiting T cell proliferation and T cell differentiation. Mechanistically, sucralose affects the membrane order of T cells, accompanied by a reduced efficiency of T cell receptor signalling and intracellular calcium mobilization. Mice given sucralose show decreased CD8+ T cell antigen-specific responses in subcutaneous cancer models and bacterial infection models, and reduced T cell function in models of T cell-mediated autoimmunity. Overall, these findings suggest that a high intake of sucralose can dampen T cell-mediated responses, an effect that could be used in therapy to mitigate T cell-dependent autoimmune disorders.
Collapse
Affiliation(s)
- Fabio Zani
- p53 and Metabolism Laboratory, The Francis Crick Institute, London, UK.
| | - Julianna Blagih
- p53 and Metabolism Laboratory, The Francis Crick Institute, London, UK.
- University of Montreal, Maisonneuve-Rosemont Hospital Research Centre, Montreal, Quebec, Canada.
| | - Tim Gruber
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München and German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Michael D Buck
- Immunobiology Laboratory, The Francis Crick Institute, London, UK
| | - Nicholas Jones
- Institute of Life Science, Swansea University Medical School, Swansea University, Swansea, UK
| | - Marc Hennequart
- p53 and Metabolism Laboratory, The Francis Crick Institute, London, UK
| | - Clare L Newell
- National Physical Laboratory, Teddington, UK
- Laboratory of Physiology and Metabolism, The Francis Crick Institute, London, UK
| | - Steven E Pilley
- p53 and Metabolism Laboratory, The Francis Crick Institute, London, UK
| | - Pablo Soro-Barrio
- Bioinformatics and Biostatistics Science Technology Platform, The Francis Crick Institute, London, UK
| | - Gavin Kelly
- Bioinformatics and Biostatistics Science Technology Platform, The Francis Crick Institute, London, UK
| | - Nathalie M Legrave
- Metabolomics Science Technology Platform, The Francis Crick Institute, London, UK
| | - Eric C Cheung
- p53 and Metabolism Laboratory, The Francis Crick Institute, London, UK
| | | | - Alex P Gould
- Laboratory of Physiology and Metabolism, The Francis Crick Institute, London, UK
| | - Cristina Garcia-Caceres
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München and German Center for Diabetes Research (DZD), Neuherberg, Germany
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Karen H Vousden
- p53 and Metabolism Laboratory, The Francis Crick Institute, London, UK.
| |
Collapse
|
28
|
Uzieliene I, Bironaite D, Bagdonas E, Pachaleva J, Sobolev A, Tsai WB, Kvederas G, Bernotiene E. The Effects of Mechanical Load on Chondrogenic Responses of Bone Marrow Mesenchymal Stem Cells and Chondrocytes Encapsulated in Chondroitin Sulfate-Based Hydrogel. Int J Mol Sci 2023; 24:ijms24032915. [PMID: 36769232 PMCID: PMC9918200 DOI: 10.3390/ijms24032915] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/23/2023] [Accepted: 01/27/2023] [Indexed: 02/05/2023] Open
Abstract
Articular cartilage is vulnerable to mechanical overload and has limited ability to restore lesions, which leads to the development of chronic diseases such as osteoarthritis (OA). In this study, the chondrogenic responses of human bone marrow mesenchymal stem cells (BMMSCs) and OA cartilage-derived chondrocytes in 3D chondroitin sulfate-tyramine/gelatin (CS-Tyr)/Gel) hydrogels with or without experimental mechanical load have been investigated. Chondrocytes were smaller in size, had slower proliferation rate and higher level of intracellular calcium (iCa2+) compared to BMMSCs. Under 3D chondrogenic conditions in CS-Tyr/Gel with or without TGF-β3, chondrocytes more intensively secreted cartilage oligomeric matrix protein (COMP) and expressed collagen type II (COL2A1) and aggrecan (ACAN) genes but were more susceptible to mechanical load compared to BMMSCs. ICa2+ was more stably controlled in CS-Tyr/Gel/BMMSCs than in CS-Tyr/Gel/chondrocytes ones, through the expression of L-type channel subunit CaV1.2 (CACNA1C) and Serca2 pump (ATP2A2) genes, and their balance was kept more stable. Due to the lower susceptibility to mechanical load, BMMSCs in CS-Tyr/Gel hydrogel may have an advantage over chondrocytes in application for cartilage regeneration purposes. The mechanical overload related cartilage damage in vivo and the vague regenerative processes of OA chondrocytes might be associated to the inefficient control of iCa2+ regulating channels.
Collapse
Affiliation(s)
- Ilona Uzieliene
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, LT-08406 Vilnius, Lithuania
| | - Daiva Bironaite
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, LT-08406 Vilnius, Lithuania
| | - Edvardas Bagdonas
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, LT-08406 Vilnius, Lithuania
| | - Jolita Pachaleva
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, LT-08406 Vilnius, Lithuania
| | - Arkadij Sobolev
- Latvian Institute of Organic Synthesis, LV-1006 Riga, Latvia
| | - Wei-Bor Tsai
- Department of Chemical Engineering, National Taiwan University, Taipei 104, Taiwan
| | - Giedrius Kvederas
- The Clinic of Rheumatology, Orthopaedics Traumatology and Reconstructive Surgery, Institute of Clinical Medicine, Faculty of Medicine, Vilnius University, LT-03101 Vilnius, Lithuania
| | - Eiva Bernotiene
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, LT-08406 Vilnius, Lithuania
- Correspondence: ; Tel.: +370-6837-7130
| |
Collapse
|
29
|
Corre B, El Janati Elidrissi Y, Duval J, Quilhot M, Lefebvre G, Ecomard S, Lemaître F, Garcia Z, Bohineust A, Russo E, Bousso P. Integration of intermittent calcium signals in T cells revealed by temporally patterned optogenetics. iScience 2023; 26:106068. [PMID: 36824271 PMCID: PMC9942117 DOI: 10.1016/j.isci.2023.106068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 11/28/2022] [Accepted: 01/20/2023] [Indexed: 01/27/2023] Open
Abstract
T cells become activated following one or multiple contacts with antigen-presenting cells. Calcium influx is a key signaling event elicited during these cellular interactions; however, it is unclear whether T cells recall and integrate calcium signals elicited during temporally separated contacts. To study the integration of calcium signals, we designed a programmable, multiplex illumination strategy for temporally patterned optogenetics (TEMPO). We found that a single round of calcium elevation was insufficient to promote nuclear factor of activated T cells (NFAT) activity and cytokine production in a T cell line. However, robust responses were detected after a second identical stimulation even when signals were separated by several hours. Our results suggest the existence of a biochemical memory of calcium signals in T cells that favors signal integration during temporally separated contacts and promote cytokine production. As illustrated here, TEMPO is a versatile approach for dissecting temporal integration in defined signaling pathways.
Collapse
Affiliation(s)
- Béatrice Corre
- Dynamics of Immune Responses Unit, Institut Pasteur, Université Paris Cité, Inserm U1223, 75015 Paris, France
| | - Yassine El Janati Elidrissi
- Dynamics of Immune Responses Unit, Institut Pasteur, Université Paris Cité, Inserm U1223, 75015 Paris, France
| | - Justine Duval
- Dynamics of Immune Responses Unit, Institut Pasteur, Université Paris Cité, Inserm U1223, 75015 Paris, France
| | - Mailys Quilhot
- Dynamics of Immune Responses Unit, Institut Pasteur, Université Paris Cité, Inserm U1223, 75015 Paris, France
| | - Gaëtan Lefebvre
- Dynamics of Immune Responses Unit, Institut Pasteur, Université Paris Cité, Inserm U1223, 75015 Paris, France
| | - Solène Ecomard
- Dynamics of Immune Responses Unit, Institut Pasteur, Université Paris Cité, Inserm U1223, 75015 Paris, France
| | - Fabrice Lemaître
- Dynamics of Immune Responses Unit, Institut Pasteur, Université Paris Cité, Inserm U1223, 75015 Paris, France
| | - Zacarias Garcia
- Dynamics of Immune Responses Unit, Institut Pasteur, Université Paris Cité, Inserm U1223, 75015 Paris, France
| | - Armelle Bohineust
- Dynamics of Immune Responses Unit, Institut Pasteur, Université Paris Cité, Inserm U1223, 75015 Paris, France
| | - Erica Russo
- Dynamics of Immune Responses Unit, Institut Pasteur, Université Paris Cité, Inserm U1223, 75015 Paris, France
| | - Philippe Bousso
- Dynamics of Immune Responses Unit, Institut Pasteur, Université Paris Cité, Inserm U1223, 75015 Paris, France,Corresponding author
| |
Collapse
|
30
|
Feng J, Xie Z, Hu H. Ion channel regulation of gut immunity. J Gen Physiol 2022; 155:213734. [PMID: 36459135 PMCID: PMC9723512 DOI: 10.1085/jgp.202113042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 08/15/2022] [Accepted: 11/17/2022] [Indexed: 12/03/2022] Open
Abstract
Mounting evidence indicates that gastrointestinal (GI) homeostasis hinges on communications among many cellular networks including the intestinal epithelium, the immune system, and both intrinsic and extrinsic nerves innervating the gut. The GI tract, especially the colon, is the home base for gut microbiome which dynamically regulates immune function. The gut's immune system also provides an effective defense against harmful pathogens entering the GI tract while maintaining immune homeostasis to avoid exaggerated immune reaction to innocuous food and commensal antigens which are important causes of inflammatory disorders such as coeliac disease and inflammatory bowel diseases (IBD). Various ion channels have been detected in multiple cell types throughout the GI tract. By regulating membrane properties and intracellular biochemical signaling, ion channels play a critical role in synchronized signaling among diverse cellular components in the gut that orchestrates the GI immune response. This work focuses on the role of ion channels in immune cells, non-immune resident cells, and neuroimmune interactions in the gut at the steady state and pathological conditions. Understanding the cellular and molecular basis of ion channel signaling in these immune-related pathways and initial testing of pharmacological intervention will facilitate the development of ion channel-based therapeutic approaches for the treatment of intestinal inflammation.
Collapse
Affiliation(s)
- Jing Feng
- Department of Anesthesiology, The Center for the Study of Itch and Sensory Disorders, Washington University School of Medicine, St. Louis, MO,Center for Neurological and Psychiatric Research and Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Science, Shanghai, China,Correspondence to Jing Feng:
| | - Zili Xie
- Department of Anesthesiology, The Center for the Study of Itch and Sensory Disorders, Washington University School of Medicine, St. Louis, MO
| | - Hongzhen Hu
- Department of Anesthesiology, The Center for the Study of Itch and Sensory Disorders, Washington University School of Medicine, St. Louis, MO,Hongzhen Hu:
| |
Collapse
|
31
|
Teppert K, Wang X, Anders K, Evaristo C, Lock D, Künkele A. Joining Forces for Cancer Treatment: From "TCR versus CAR" to "TCR and CAR". Int J Mol Sci 2022; 23:14563. [PMID: 36498890 PMCID: PMC9739809 DOI: 10.3390/ijms232314563] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 11/14/2022] [Accepted: 11/19/2022] [Indexed: 11/24/2022] Open
Abstract
T cell-based immunotherapy has demonstrated great therapeutic potential in recent decades, on the one hand, by using tumor-infiltrating lymphocytes (TILs) and, on the other hand, by engineering T cells to obtain anti-tumor specificities through the introduction of either engineered T cell receptors (TCRs) or chimeric antigen receptors (CARs). Given the distinct design of both receptors and the type of antigen that is encountered, the requirements for proper antigen engagement and downstream signal transduction by TCRs and CARs differ. Synapse formation and signal transduction of CAR T cells, despite further refinement of CAR T cell designs, still do not fully recapitulate that of TCR T cells and might limit CAR T cell persistence and functionality. Thus, deep knowledge about the molecular differences in CAR and TCR T cell signaling would greatly advance the further optimization of CAR designs and elucidate under which circumstances a combination of both receptors would improve the functionality of T cells for cancer treatment. Herein, we provide a comprehensive review about similarities and differences by directly comparing the architecture, synapse formation and signaling of TCRs and CARs, highlighting the knowns and unknowns. In the second part of the review, we discuss the current status of combining CAR and TCR technologies, encouraging a change in perspective from "TCR versus CAR" to "TCR and CAR".
Collapse
Affiliation(s)
- Karin Teppert
- Miltenyi Biotec B.V. & Co. KG, 51429 Bergisch Gladbach, Germany
| | - Xueting Wang
- Miltenyi Biotec B.V. & Co. KG, 51429 Bergisch Gladbach, Germany
| | - Kathleen Anders
- German Cancer Consortium (DKTK), 10117 Berlin, Germany
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - César Evaristo
- Miltenyi Biotec B.V. & Co. KG, 51429 Bergisch Gladbach, Germany
| | - Dominik Lock
- Miltenyi Biotec B.V. & Co. KG, 51429 Bergisch Gladbach, Germany
| | - Annette Künkele
- German Cancer Consortium (DKTK), 10117 Berlin, Germany
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Department of Pediatric Oncology and Hematology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 13353 Berlin, Germany
| |
Collapse
|
32
|
Zang H, Siddiqui M, Gummuluru S, Wong WW, Reinhard BM. Ganglioside-Functionalized Nanoparticles for Chimeric Antigen Receptor T-Cell Activation at the Immunological Synapse. ACS NANO 2022; 16:18408-18420. [PMID: 36282488 PMCID: PMC9815837 DOI: 10.1021/acsnano.2c06516] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2023]
Abstract
Chimeric Antigen Receptor (CAR) T cell therapy has proven to be an effective strategy against hematological malignancies but persistence and activity against solid tumors must be further improved. One emerging strategy for enhancing efficacy is based on directing CAR T cells to antigen presenting cells (APCs). Activation of CAR T cells at the immunological synapse (IS) formed between APC and T cell is thought to promote strong, persistent antigen-specific T cell-mediated immune responses but requires integration of CAR ligands into the APC/T-cell interface. Here, we demonstrate that CAR ligand functionalized, lipid-coated, biodegradable polymer nanoparticles (NPs) that contain the ganglioside GM3 (GM3-NPs) bind to CD169 (Siglec-1)-expressing APCs and localize to the cell contact site between APCs and CAR T cells upon initiation of cell conjugates. The CD169+ APC/CAR T-cell interface is characterized by a strong optical colocalization of GM3-NPs and CARs, enrichment of F-actin, and recruitment of ZAP-70, indicative of integration of GM3-NPs into a functional IS. Ligands associated with GM3-NPs localized to the APC/T-cell contact site remain accessible to CARs and result in robust T-cell activation. Overall, this work identifies GM3-NPs as a potential antigen delivery platform for active targeting of CD169 expressing APCs and enhancement of CAR T-cell activation at the NP-containing IS.
Collapse
Affiliation(s)
- Han Zang
- Departments of Chemistry and The Photonics Center, Boston University, Boston, MA, 02215, United States
| | - Menna Siddiqui
- Department of Biomedical Engineering and Biological Design Center, Boston University, Boston, MA, 02215, USA
| | - Suryaram Gummuluru
- Department of Microbiology, Boston University School of Medicine, Boston, MA, 02118, United States
| | - Wilson W. Wong
- Department of Biomedical Engineering and Biological Design Center, Boston University, Boston, MA, 02215, USA
| | - Björn M. Reinhard
- Departments of Chemistry and The Photonics Center, Boston University, Boston, MA, 02215, United States
| |
Collapse
|
33
|
Brochet P, Ianni BM, Laugier L, Frade AF, Silva Nunes JP, Teixeira PC, Mady C, Ferreira LRP, Ferré Q, Santos RHB, Kuramoto A, Cabantous S, Steffen S, Stolf AN, Pomerantzeff P, Fiorelli AI, Bocchi EA, Pissetti CW, Saba B, Cândido DDS, Dias FC, Sampaio MF, Gaiotto FA, Marin-Neto JA, Fragata A, Zaniratto RCF, Siqueira S, Peixoto GDL, Rigaud VOC, Bacal F, Buck P, Almeida RR, Lin-Wang HT, Schmidt A, Martinelli M, Hirata MH, Donadi EA, Costa Pereira A, Rodrigues Junior V, Puthier D, Kalil J, Spinelli L, Cunha-Neto E, Chevillard C. Epigenetic regulation of transcription factor binding motifs promotes Th1 response in Chagas disease cardiomyopathy. Front Immunol 2022; 13:958200. [PMID: 36072583 PMCID: PMC9441916 DOI: 10.3389/fimmu.2022.958200] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 07/26/2022] [Indexed: 01/03/2023] Open
Abstract
Chagas disease, caused by the protozoan Trypanosoma cruzi, is an endemic parasitic disease of Latin America, affecting 7 million people. Although most patients are asymptomatic, 30% develop complications, including the often-fatal Chronic Chagasic Cardiomyopathy (CCC). Although previous studies have demonstrated some genetic deregulations associated with CCCs, the causes of their deregulations remain poorly described. Based on bulk RNA-seq and whole genome DNA methylation data, we investigated the genetic and epigenetic deregulations present in the moderate and severe stages of CCC. Analysis of heart tissue gene expression profile allowed us to identify 1407 differentially expressed transcripts (DEGs) specific from CCC patients. A tissue DNA methylation analysis done on the same tissue has permitted the identification of 92 regulatory Differentially Methylated Regions (DMR) localized in the promoter of DEGs. An in-depth study of the transcription factors binding sites (TFBS) in the DMRs corroborated the importance of TFBS’s DNA methylation for gene expression in CCC myocardium. TBX21, RUNX3 and EBF1 are the transcription factors whose binding motif appears to be affected by DNA methylation in the largest number of genes. By combining both transcriptomic and methylomic analysis on heart tissue, and methylomic analysis on blood, 4 biological processes affected by severe CCC have been identified, including immune response, ion transport, cardiac muscle processes and nervous system. An additional study on blood methylation of moderate CCC samples put forward the importance of ion transport and nervous system in the development of the disease.
Collapse
Affiliation(s)
- Pauline Brochet
- Institut National de la Santé Et de la Recherche Médicale (INSERM), Unité Mixte de Recherche (UMR)_1090, Aix Marseille Université, TAGC Theories and Approaches of Genomic Complexity, Institut MarMaRa, Marseille, France
| | - Barbara Maria Ianni
- Laboratory of Immunology, Heart Institute Instituto do Coração (InCor), University of São Paulo, School of Medicine, São Paulo, Brazil
| | - Laurie Laugier
- Aix Marseille Université, Génétique et Immunologie des Maladies Parasitaires, Inserm, UMR_906, Marseille, France
| | - Amanda Farage Frade
- Laboratory of Immunology, Heart Institute Instituto do Coração (InCor), University of São Paulo, School of Medicine, São Paulo, Brazil
- Division of Clinical Immunology and Allergy, University of São Paulo, School of Medicine, São Paulo, Brazil
- Instituto Nacional de Ciência e Tecnologia, INCT, III- Institute for Investigation in Immunology, São Paulo, Brazil
| | - João Paulo Silva Nunes
- Institut National de la Santé Et de la Recherche Médicale (INSERM), Unité Mixte de Recherche (UMR)_1090, Aix Marseille Université, TAGC Theories and Approaches of Genomic Complexity, Institut MarMaRa, Marseille, France
- Laboratory of Immunology, Heart Institute Instituto do Coração (InCor), University of São Paulo, School of Medicine, São Paulo, Brazil
- Division of Clinical Immunology and Allergy, University of São Paulo, School of Medicine, São Paulo, Brazil
- Instituto Nacional de Ciência e Tecnologia, INCT, III- Institute for Investigation in Immunology, São Paulo, Brazil
| | - Priscila Camillo Teixeira
- Laboratory of Immunology, Heart Institute Instituto do Coração (InCor), University of São Paulo, School of Medicine, São Paulo, Brazil
- Division of Clinical Immunology and Allergy, University of São Paulo, School of Medicine, São Paulo, Brazil
- Instituto Nacional de Ciência e Tecnologia, INCT, III- Institute for Investigation in Immunology, São Paulo, Brazil
| | - Charles Mady
- Myocardiopathies and Aortic Diseases Unit, Heart Institute Instituto do Coração (InCor), School of Medicine, University of São Paulo, São Paulo, Brazil
| | - Ludmila Rodrigues Pinto Ferreira
- RNA Systems Biology Laboratory (RSBL), Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Quentin Ferré
- Institut National de la Santé Et de la Recherche Médicale (INSERM), Unité Mixte de Recherche (UMR)_1090, Aix Marseille Université, TAGC Theories and Approaches of Genomic Complexity, Institut MarMaRa, Marseille, France
| | - Ronaldo Honorato Barros Santos
- Division of Transplantation, Heart Institute Instituto do Coração (InCor), University of São Paulo, School of Medicine, São Paulo, Brazil
| | - Andreia Kuramoto
- Laboratory of Immunology, Heart Institute Instituto do Coração (InCor), University of São Paulo, School of Medicine, São Paulo, Brazil
| | - Sandrine Cabantous
- Aix Marseille Université, Génétique et Immunologie des Maladies Parasitaires, Inserm, UMR_906, Marseille, France
| | - Samuel Steffen
- Division of Transplantation, Heart Institute Instituto do Coração (InCor), University of São Paulo, School of Medicine, São Paulo, Brazil
- Division of Surgery, Heart Institute Instituto do Coração (InCor), University of São Paulo, School of Medicine, São Paulo, Brazil
| | - Antonio Noedir Stolf
- Division of Surgery, Heart Institute Instituto do Coração (InCor), University of São Paulo, School of Medicine, São Paulo, Brazil
| | - Pablo Pomerantzeff
- Heart Institute (InCor), School of Medicine, University of São Paulo, São Paulo, São Paulo, Brazil
| | - Alfredo Inacio Fiorelli
- Division of Surgery, Heart Institute Instituto do Coração (InCor), University of São Paulo, School of Medicine, São Paulo, Brazil
| | - Edimar Alcides Bocchi
- Division of Surgery, Heart Institute Instituto do Coração (InCor), University of São Paulo, School of Medicine, São Paulo, Brazil
| | - Cristina Wide Pissetti
- Laboratory of Immunology, Universidade Federal Do Triângulo Mineiro (UFTM), Uberaba, Brazil
| | - Bruno Saba
- Laboratório de Investigação Molecular em Cardiologia, Instituto de Cardiologia Dante Pazzanese (IDPC), São Paulo, Brazil
| | - Darlan da Silva Cândido
- Laboratory of Immunology, Heart Institute Instituto do Coração (InCor), University of São Paulo, School of Medicine, São Paulo, Brazil
- Division of Clinical Immunology and Allergy, University of São Paulo, School of Medicine, São Paulo, Brazil
- Instituto Nacional de Ciência e Tecnologia, INCT, III- Institute for Investigation in Immunology, São Paulo, Brazil
| | - Fabrício C. Dias
- School of Medicine of Ribeirão Preto Faculdade de Medicina de Ribeirão Preto (FMRP), University of São Paulo, Ribeirão Preto, Brazil
| | - Marcelo Ferraz Sampaio
- Laboratório de Investigação Molecular em Cardiologia, Instituto de Cardiologia Dante Pazzanese (IDPC), São Paulo, Brazil
| | - Fabio Antônio Gaiotto
- Division of Transplantation, Heart Institute Instituto do Coração (InCor), University of São Paulo, School of Medicine, São Paulo, Brazil
- Division of Surgery, Heart Institute Instituto do Coração (InCor), University of São Paulo, School of Medicine, São Paulo, Brazil
| | - José Antonio Marin-Neto
- School of Medicine of Ribeirão Preto Faculdade de Medicina de Ribeirão Preto (FMRP), University of São Paulo, Ribeirão Preto, Brazil
| | - Abílio Fragata
- Laboratório de Investigação Molecular em Cardiologia, Instituto de Cardiologia Dante Pazzanese (IDPC), São Paulo, Brazil
| | - Ricardo Costa Fernandes Zaniratto
- Laboratory of Immunology, Heart Institute Instituto do Coração (InCor), University of São Paulo, School of Medicine, São Paulo, Brazil
| | - Sergio Siqueira
- Pacemaker Clinic, Heart Institute Instituto do Coração (InCor), School of Medicine, University of São Paulo, São Paulo, Brazil
| | - Giselle De Lima Peixoto
- Pacemaker Clinic, Heart Institute Instituto do Coração (InCor), School of Medicine, University of São Paulo, São Paulo, Brazil
| | - Vagner Oliveira-Carvalho Rigaud
- Laboratory of Immunology, Heart Institute Instituto do Coração (InCor), University of São Paulo, School of Medicine, São Paulo, Brazil
- Heart Failure Unit, Heart Institute Instituto do Coração (InCor) School of Medicine, University of Sao Paulo, Sao Paulo, Brazil
| | - Fernando Bacal
- Division of Transplantation, Heart Institute Instituto do Coração (InCor), University of São Paulo, School of Medicine, São Paulo, Brazil
| | - Paula Buck
- Heart Institute (InCor), School of Medicine, University of São Paulo, São Paulo, São Paulo, Brazil
| | - Rafael Ribeiro Almeida
- Laboratory of Immunology, Heart Institute Instituto do Coração (InCor), University of São Paulo, School of Medicine, São Paulo, Brazil
- Division of Clinical Immunology and Allergy, University of São Paulo, School of Medicine, São Paulo, Brazil
- Instituto Nacional de Ciência e Tecnologia, INCT, III- Institute for Investigation in Immunology, São Paulo, Brazil
| | - Hui Tzu Lin-Wang
- Laboratório de Investigação Molecular em Cardiologia, Instituto de Cardiologia Dante Pazzanese (IDPC), São Paulo, Brazil
| | - André Schmidt
- School of Medicine of Ribeirão Preto Faculdade de Medicina de Ribeirão Preto (FMRP), University of São Paulo, Ribeirão Preto, Brazil
| | - Martino Martinelli
- Pacemaker Clinic, Heart Institute Instituto do Coração (InCor), School of Medicine, University of São Paulo, São Paulo, Brazil
| | - Mario Hiroyuki Hirata
- Department of Clinical and Toxicological Analyses, Faculty of Pharmaceutical Sciences, University of São Paulo (USP), São Paulo, Brazil
| | - Eduardo Antonio Donadi
- School of Medicine of Ribeirão Preto Faculdade de Medicina de Ribeirão Preto (FMRP), University of São Paulo, Ribeirão Preto, Brazil
| | - Alexandre Costa Pereira
- Heart Institute (InCor), School of Medicine, University of São Paulo, São Paulo, São Paulo, Brazil
| | | | - Denis Puthier
- Institut National de la Santé Et de la Recherche Médicale (INSERM), Unité Mixte de Recherche (UMR)_1090, Aix Marseille Université, TAGC Theories and Approaches of Genomic Complexity, Institut MarMaRa, Marseille, France
| | - Jorge Kalil
- Laboratory of Immunology, Heart Institute Instituto do Coração (InCor), University of São Paulo, School of Medicine, São Paulo, Brazil
- Division of Clinical Immunology and Allergy, University of São Paulo, School of Medicine, São Paulo, Brazil
- Instituto Nacional de Ciência e Tecnologia, INCT, III- Institute for Investigation in Immunology, São Paulo, Brazil
| | - Lionel Spinelli
- Institut National de la Santé Et de la Recherche Médicale (INSERM), Unité Mixte de Recherche (UMR)_1090, Aix Marseille Université, TAGC Theories and Approaches of Genomic Complexity, Institut MarMaRa, Marseille, France
- Aix Marseille Université, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy, Marseille, France
- *Correspondence: Christophe Chevillard, ; Edecio Cunha-Neto, ; Lionel Spinelli,
| | - Edecio Cunha-Neto
- Laboratory of Immunology, Heart Institute Instituto do Coração (InCor), University of São Paulo, School of Medicine, São Paulo, Brazil
- Division of Clinical Immunology and Allergy, University of São Paulo, School of Medicine, São Paulo, Brazil
- Instituto Nacional de Ciência e Tecnologia, INCT, III- Institute for Investigation in Immunology, São Paulo, Brazil
- *Correspondence: Christophe Chevillard, ; Edecio Cunha-Neto, ; Lionel Spinelli,
| | - Christophe Chevillard
- Institut National de la Santé Et de la Recherche Médicale (INSERM), Unité Mixte de Recherche (UMR)_1090, Aix Marseille Université, TAGC Theories and Approaches of Genomic Complexity, Institut MarMaRa, Marseille, France
- *Correspondence: Christophe Chevillard, ; Edecio Cunha-Neto, ; Lionel Spinelli,
| |
Collapse
|
34
|
Johari M, Vihola A, Palmio J, Jokela M, Jonson PH, Sarparanta J, Huovinen S, Savarese M, Hackman P, Udd B. Comprehensive transcriptomic analysis shows disturbed calcium homeostasis and deregulation of T lymphocyte apoptosis in inclusion body myositis. J Neurol 2022; 269:4161-4173. [PMID: 35237874 PMCID: PMC9293871 DOI: 10.1007/s00415-022-11029-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 02/09/2022] [Accepted: 02/13/2022] [Indexed: 12/11/2022]
Abstract
OBJECTIVE Inclusion body myositis (IBM) has an unclear molecular etiology exhibiting both characteristic inflammatory T-cell activity and rimmed-vacuolar degeneration of muscle fibers. Using in-depth gene expression and splicing studies, we aimed at understanding the different components of the molecular pathomechanisms in IBM. METHODS We performed RNA-seq on RNA extracted from skeletal muscle biopsies of clinically and histopathologically defined IBM (n = 24), tibial muscular dystrophy (n = 6), and histopathologically normal group (n = 9). In a comprehensive transcriptomics analysis, we analyzed the differential gene expression, differential splicing and exon usage, downstream pathway analysis, and the interplay between coding and non-coding RNAs (micro RNAs and long non-coding RNAs). RESULTS We observe dysregulation of genes involved in calcium homeostasis, particularly affecting the T-cell activity and regulation, causing disturbed Ca2+-induced apoptotic pathways of T cells in IBM muscles. Additionally, LCK/p56, which is an essential gene in regulating the fate of T-cell apoptosis, shows increased expression and altered splicing usage in IBM muscles. INTERPRETATION Our analysis provides a novel understanding of the molecular mechanisms in IBM by showing a detailed dysregulation of genes involved in calcium homeostasis and its effect on T-cell functioning in IBM muscles. Loss of T-cell regulation is hypothesized to be involved in the consistent observation of no response to immune therapies in IBM patients. Our results show that loss of apoptotic control of cytotoxic T cells could indeed be one component of their abnormal cytolytic activity in IBM muscles.
Collapse
Affiliation(s)
- Mridul Johari
- Folkhälsan Research Center, Helsinki, Finland.
- Department of Medical and Clinical Genetics, Medicum, University of Helsinki, Helsinki, Finland.
| | - Anna Vihola
- Folkhälsan Research Center, Helsinki, Finland
- Department of Medical and Clinical Genetics, Medicum, University of Helsinki, Helsinki, Finland
- Neuromuscular Research Center, Department of Genetics, Fimlab Laboratories, Tampere, Finland
| | - Johanna Palmio
- Neuromuscular Research Center, Department of Neurology, Tampere University and University Hospital, Tampere, Finland
| | - Manu Jokela
- Neuromuscular Research Center, Department of Genetics, Fimlab Laboratories, Tampere, Finland
- Division of Clinical Neurosciences, Department of Neurology, Turku University Hospital, Turku, Finland
| | - Per Harald Jonson
- Folkhälsan Research Center, Helsinki, Finland
- Department of Medical and Clinical Genetics, Medicum, University of Helsinki, Helsinki, Finland
| | - Jaakko Sarparanta
- Folkhälsan Research Center, Helsinki, Finland
- Department of Medical and Clinical Genetics, Medicum, University of Helsinki, Helsinki, Finland
| | - Sanna Huovinen
- Department of Pathology, Fimlab Laboratories, Tampere University Hospital, Tampere, Finland
| | - Marco Savarese
- Folkhälsan Research Center, Helsinki, Finland
- Department of Medical and Clinical Genetics, Medicum, University of Helsinki, Helsinki, Finland
| | - Peter Hackman
- Folkhälsan Research Center, Helsinki, Finland
- Department of Medical and Clinical Genetics, Medicum, University of Helsinki, Helsinki, Finland
| | - Bjarne Udd
- Folkhälsan Research Center, Helsinki, Finland
- Department of Medical and Clinical Genetics, Medicum, University of Helsinki, Helsinki, Finland
- Neuromuscular Research Center, Department of Neurology, Tampere University and University Hospital, Tampere, Finland
- Department of Neurology, Vaasa Central Hospital, Vaasa, Finland
| |
Collapse
|
35
|
Kim DS, Na HS, Cho KH, Lee KH, Choi J, Kwok SK, Bae YS, Cho ML, Park SH. Sphingosylphosphorylcholine ameliorates experimental sjögren's syndrome by regulating salivary gland inflammation and hypofunction, and regulatory B cells. Immunol Lett 2022; 248:62-69. [PMID: 35732207 DOI: 10.1016/j.imlet.2022.06.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 06/14/2022] [Accepted: 06/18/2022] [Indexed: 11/24/2022]
Abstract
Sjögren syndrome (SS) is an autoimmune disease in which immune cells infiltrate the exocrine gland. Since SS is caused by a disorder of the immune system, treatments should regulate the immune response. Sphingosylphosphorylcholine (SPC) is a sphingolipid that mediates cellular signaling. In immune cells, SPC has several immunomodulatory functions. Accordingly, this study verifies the immunomodulatory ability and therapeutic effect of SPC in SS. To understand the function of SPC in SS, we treated SPC in female NOD/ShiJcl (NOD) mice. The mice were monitored for 10 weeks, and inflammation in the salivary glands was checked. After SPC treatment, we detected the expression of regulatory B (Breg) cells in mouse splenocytes and the level of salivary secretion-related genes in human submandibular gland (HSG) cells. Salivary flow rate was maintained in the SPC-treated group compared to the vehicle-treated group, and inflammation in the salivary gland tissues was relieved by SPC. SPC treatment in mouse cells and HSG cells enhanced Breg cells and salivary secretion markers, respectively. This study revealed that SPC can be considered as a new therapeutic agent against SS.
Collapse
Affiliation(s)
- Da Som Kim
- Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, 222, Banpo-Daero, Seocho-gu, Seoul, 06591, Republic of Korea; Lab of Translational ImmunoMedicine, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea; Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Hyun Sik Na
- Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, 222, Banpo-Daero, Seocho-gu, Seoul, 06591, Republic of Korea; Lab of Translational ImmunoMedicine, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea; Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Keun-Hyung Cho
- Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, 222, Banpo-Daero, Seocho-gu, Seoul, 06591, Republic of Korea; Lab of Translational ImmunoMedicine, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea; Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Kun Hee Lee
- Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, 222, Banpo-Daero, Seocho-gu, Seoul, 06591, Republic of Korea; Lab of Translational ImmunoMedicine, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea; Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - JeongWon Choi
- Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, 222, Banpo-Daero, Seocho-gu, Seoul, 06591, Republic of Korea; Lab of Translational ImmunoMedicine, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Seung-Ki Kwok
- Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, 222, Banpo-Daero, Seocho-gu, Seoul, 06591, Republic of Korea; Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, 222, Banpo-Daero, Seocho-gu, Seoul, 06591, Republic of Korea
| | - Yoe-Sik Bae
- Department of Biological Sciences, Sungkyunkwan University, Suwon, 16419, Republic of Korea; Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, 06355, Republic of Korea.
| | - Mi-La Cho
- Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, 222, Banpo-Daero, Seocho-gu, Seoul, 06591, Republic of Korea; Lab of Translational ImmunoMedicine, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea; Department of Medical Life Sciences, College of Medicine, The Catholic University of Korea, 222, Banpo-daero, Seocho-gu, Seoul, 06591, Republic of Korea.
| | - Sung-Hwan Park
- Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, 222, Banpo-Daero, Seocho-gu, Seoul, 06591, Republic of Korea; Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, 222, Banpo-Daero, Seocho-gu, Seoul, 06591, Republic of Korea.
| |
Collapse
|
36
|
Ahmed WUR, Kleeman S, Ng M, Wang W, Auton A, Lee R, Handa A, Zondervan KT, Wiberg A, Furniss D. Genome-wide association analysis and replication in 810,625 individuals with varicose veins. Nat Commun 2022; 13:3065. [PMID: 35654884 PMCID: PMC9163161 DOI: 10.1038/s41467-022-30765-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Accepted: 05/17/2022] [Indexed: 12/30/2022] Open
Abstract
Varicose veins affect one-third of Western society, with a significant subset of patients developing venous ulceration, costing $14.9 billion annually in the USA. Current management consists of either compression stockings, or surgical ablation for more advanced disease. Most varicose veins patients report a positive family history, and heritability is ~17%. We describe the largest two-stage genome-wide association study of varicose veins in 401,656 individuals from UK Biobank, and replication in 408,969 individuals from 23andMe (total 135,514 cases and 675,111 controls). Forty-nine signals at 46 susceptibility loci were discovered. We map 237 genes to these loci, several of which are biologically plausible and tractable to therapeutic targeting. Pathway analysis identified enrichment in extracellular matrix biology, inflammation, (lymph)angiogenesis, vascular smooth muscle cell migration, and apoptosis. Using a polygenic risk score (PRS) derived in an independent cohort, we demonstrate its predictive utility and correlation with varicose veins surgery.
Collapse
Affiliation(s)
- Waheed-Ul-Rahman Ahmed
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Botnar Research Centre, Windmill Road, Oxford, OX3 7LD, UK
| | - Sam Kleeman
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, 11724, USA
| | - Michael Ng
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Botnar Research Centre, Windmill Road, Oxford, OX3 7LD, UK
| | - Wei Wang
- 23andMe, Inc., Sunnyvale, CA, USA
| | | | | | - Regent Lee
- Nuffield Department of Surgical Sciences, University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DU, UK
| | - Ashok Handa
- Nuffield Department of Surgical Sciences, University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DU, UK
| | - Krina T Zondervan
- Nuffield Department of Women's & Reproductive Health, University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DU, UK.,Wellcome Centre for Human Genetics, University of Oxford, Old Road Campus, Roosevelt Drive, Oxford, OX3 7BN, UK
| | - Akira Wiberg
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Botnar Research Centre, Windmill Road, Oxford, OX3 7LD, UK.,Department of Plastic and Reconstructive Surgery, Oxford University Hospitals NHS Foundation Trust, John Radcliffe Hospital, Oxford, OX3 9DU, UK
| | - Dominic Furniss
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Botnar Research Centre, Windmill Road, Oxford, OX3 7LD, UK. .,Department of Plastic and Reconstructive Surgery, Oxford University Hospitals NHS Foundation Trust, John Radcliffe Hospital, Oxford, OX3 9DU, UK.
| |
Collapse
|
37
|
Photopharmacological modulation of native CRAC channels using azoboronate photoswitches. Proc Natl Acad Sci U S A 2022; 119:e2118160119. [PMID: 35312368 PMCID: PMC9060504 DOI: 10.1073/pnas.2118160119] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Calcium release–activated calcium (CRAC) channels play key roles in the regulation of cellular signaling, transcription, and migration. Here, we describe the design, chemical synthesis, and characterization of photoswitchable channel inhibitors that can be switched on and off depending on the wavelength of light used. We use the compounds to induce light-dependent modulation of channel activity and downstream gene expression in human immune cells. We further expand the usage of the compounds to control seeding of cancer cells in target tissue and regulation of response to noxious stimuli in vivo in mice. Store-operated calcium entry through calcium release–activated calcium (CRAC) channels replenishes intracellular calcium stores and plays a critical role in cellular calcium signaling. CRAC channels are activated by tightly regulated interaction between the endoplasmic reticulum (ER) calcium sensor STIM proteins and plasma membrane (PM) Orai channels. Our current understanding of the role of STIM–Orai-dependent calcium signals under physiologically relevant conditions remains limited in part due to a lack of spatiotemporally precise methods for direct manipulation of endogenous CRAC channels. Here, we report the synthesis and characterization of azoboronate light-operated CRAC channel inhibitors (LOCIs) that allow for a dynamic and fully reversible remote modulation of the function of native CRAC channels using ultraviolet (UV) and visible light. We demonstrate the use of LOCI-1 to modulate gene expression in T lymphocytes, cancer cell seeding at metastatic sites, and pain-related behavior.
Collapse
|
38
|
Bieerkehazhi S, Fan Y, West SJ, Tewari R, Ko J, Mills T, Boehning D, Akimzhanov AM. Ca2+-dependent protein acyltransferase DHHC21 controls activation of CD4+ T cells. J Cell Sci 2022; 135:jcs258186. [PMID: 34080635 PMCID: PMC8214660 DOI: 10.1242/jcs.258186] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 04/13/2021] [Indexed: 11/20/2022] Open
Abstract
Despite the recognized significance of reversible protein lipidation (S-acylation) for T cell receptor signal transduction, the enzymatic control of this post-translational modification in T cells remains poorly understood. Here, we demonstrate that DHHC21 (also known as ZDHHC21), a member of the DHHC family of mammalian protein acyltransferases, mediates T cell receptor-induced S-acylation of proximal T cell signaling proteins. Using Zdhhc21dep mice, which express a functionally deficient version of DHHC21, we show that DHHC21 is a Ca2+/calmodulin-dependent enzyme critical for activation of naïve CD4+ T cells in response to T cell receptor stimulation. We find that disruption of the Ca2+/calmodulin-binding domain of DHHC21 does not affect thymic T cell development but prevents differentiation of peripheral CD4+ T cells into Th1, Th2 and Th17 effector T helper lineages. Our findings identify DHHC21 as an essential component of the T cell receptor signaling machinery and define a new role for protein acyltransferases in regulation of T cell-mediated immunity.
Collapse
Affiliation(s)
- Shayahati Bieerkehazhi
- Department of Biochemistry and Molecular Biology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Ying Fan
- Department of Biochemistry and Molecular Biology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
- Cooper Medical School of Rowan University, Camden, NJ 08103, USA
| | - Savannah J. West
- Department of Biochemistry and Molecular Biology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
- MD Anderson Cancer Center and University of Texas Health Science at Houston Graduate School, Houston, TX 77030, USA
| | - Ritika Tewari
- Department of Biochemistry and Molecular Biology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Junsuk Ko
- Department of Biochemistry and Molecular Biology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
- MD Anderson Cancer Center and University of Texas Health Science at Houston Graduate School, Houston, TX 77030, USA
| | - Tingting Mills
- Department of Biochemistry and Molecular Biology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Darren Boehning
- Cooper Medical School of Rowan University, Camden, NJ 08103, USA
| | - Askar M. Akimzhanov
- Department of Biochemistry and Molecular Biology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| |
Collapse
|
39
|
Berlansky S, Sallinger M, Grabmayr H, Humer C, Bernhard A, Fahrner M, Frischauf I. Calcium Signals during SARS-CoV-2 Infection: Assessing the Potential of Emerging Therapies. Cells 2022; 11:253. [PMID: 35053369 PMCID: PMC8773957 DOI: 10.3390/cells11020253] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 01/05/2022] [Accepted: 01/11/2022] [Indexed: 01/09/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a positive-sense single-stranded RNA virus that causes coronavirus disease 2019 (COVID-19). This respiratory illness was declared a pandemic by the world health organization (WHO) in March 2020, just a few weeks after being described for the first time. Since then, global research effort has considerably increased humanity's knowledge about both viruses and disease. It has also spawned several vaccines that have proven to be key tools in attenuating the spread of the pandemic and severity of COVID-19. However, with vaccine-related skepticism being on the rise, as well as breakthrough infections in the vaccinated population and the threat of a complete immune escape variant, alternative strategies in the fight against SARS-CoV-2 are urgently required. Calcium signals have long been known to play an essential role in infection with diverse viruses and thus constitute a promising avenue for further research on therapeutic strategies. In this review, we introduce the pivotal role of calcium signaling in viral infection cascades. Based on this, we discuss prospective calcium-related treatment targets and strategies for the cure of COVID-19 that exploit viral dependence on calcium signals.
Collapse
Affiliation(s)
| | | | | | | | | | - Marc Fahrner
- Institute of Biophysics, Johannes Kepler University Linz, Gruberstrasse 40, 4020 Linz, Austria; (S.B.); (M.S.); (H.G.); (C.H.); (A.B.)
| | - Irene Frischauf
- Institute of Biophysics, Johannes Kepler University Linz, Gruberstrasse 40, 4020 Linz, Austria; (S.B.); (M.S.); (H.G.); (C.H.); (A.B.)
| |
Collapse
|
40
|
Al-Aghbar MA, Jainarayanan AK, Dustin ML, Roffler SR. The interplay between membrane topology and mechanical forces in regulating T cell receptor activity. Commun Biol 2022; 5:40. [PMID: 35017678 PMCID: PMC8752658 DOI: 10.1038/s42003-021-02995-1] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 12/21/2021] [Indexed: 12/20/2022] Open
Abstract
T cells are critically important for host defense against infections. T cell activation is specific because signal initiation requires T cell receptor (TCR) recognition of foreign antigen peptides presented by major histocompatibility complexes (pMHC) on antigen presenting cells (APCs). Recent advances reveal that the TCR acts as a mechanoreceptor, but it remains unclear how pMHC/TCR engagement generates mechanical forces that are converted to intracellular signals. Here we propose a TCR Bending Mechanosignal (TBM) model, in which local bending of the T cell membrane on the nanometer scale allows sustained contact of relatively small pMHC/TCR complexes interspersed among large surface receptors and adhesion molecules on the opposing surfaces of T cells and APCs. Localized T cell membrane bending is suggested to increase accessibility of TCR signaling domains to phosphorylation, facilitate selective recognition of agonists that form catch bonds, and reduce noise signals associated with slip bonds.
Collapse
Affiliation(s)
- Mohammad Ameen Al-Aghbar
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
- Department of Translational Medicine, Sidra Medicine, Doha, Qatar
| | - Ashwin K Jainarayanan
- Interdisciplinary Bioscience Doctoral Training Program and Exeter College, University of Oxford, Oxford, UK
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Michael L Dustin
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK.
| | - Steve R Roffler
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan.
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.
| |
Collapse
|
41
|
Lee D, Ha J, Kang M, Yang Z, Jiang W, Kim BYS. Strategies of Perturbing Ion Homeostasis for Cancer Therapy. ADVANCED THERAPEUTICS 2022. [DOI: 10.1002/adtp.202100189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- DaeYong Lee
- Department of Neurosurgery The University of Texas MD Anderson Cancer Center Houston TX 77030 USA
| | - JongHoon Ha
- Department of Radiation Oncology The University of Texas MD Anderson Cancer Center Houston TX 77030 USA
| | - Minjeong Kang
- Department of Radiation Oncology The University of Texas MD Anderson Cancer Center Houston TX 77030 USA
| | - Zhaogang Yang
- Department of Radiation Oncology The University of Texas MD Anderson Cancer Center Houston TX 77030 USA
| | - Wen Jiang
- Department of Radiation Oncology The University of Texas MD Anderson Cancer Center Houston TX 77030 USA
| | - Betty Y. S. Kim
- Department of Neurosurgery The University of Texas MD Anderson Cancer Center Houston TX 77030 USA
| |
Collapse
|
42
|
A Alfheeaid H, Imam Rabbani S. COVID-19: A Review on the Role of Trace Elements Present in Saudi Arabian Traditional Dietary Supplements. Pak J Biol Sci 2022; 25:1-8. [PMID: 35001569 DOI: 10.3923/pjbs.2022.1.8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
The novel coronavirus infection is also called COVID-19 (coronavirus disease 2019). The infection has affected millions of people worldwide and caused morbidity as well mortality in patients with pre-existing chronic conditions such as metabolic, respiratory and cardiovascular disorders. The severity of the disease is mostly seen in people with low immunity and chronic sufferers of respiratory, cardiovascular and metabolic disorders. To date, there is no specific treatment available for COVID-19. Precaution and prevention are the most recommended options followed for controlling the spread of infection. Trace elements such as zinc, calcium, iron and magnesium play an important role in boosting the immunity of the host system. These components assist in the development and functioning of lymphocytes, cytokines, free radicals, inflammatory mediators and endothelial functioning. This review summarizes the common dietary supplements that are regularly consumed in Saudi Arabia and are known to contain these vital trace elements. Data available in Google Scholar, NCBI, PUBMED, EMBASE and Web of Science about COVID-19, micronutrients, trace elements and nutritional supplements of Saudi Arabia was collected. By highlighting the traditionally used dietary components containing the essential elements, this review could provide useful knowledge crucial for building immunity in the population.
Collapse
|
43
|
Zack DE, Stern DA, Willis AL, Kim AS, Mansfield CJ, Reed DR, Brooks SG, Adappa ND, Palmer JN, Cohen NA, Chiu AG, Song BH, Le CH, Chang EH. The GSDMB rs7216389 SNP is associated with chronic rhinosinusitis in a multi-institutional cohort. Int Forum Allergy Rhinol 2021; 11:1647-1653. [PMID: 34076350 PMCID: PMC8636513 DOI: 10.1002/alr.22824] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 04/23/2021] [Accepted: 05/05/2021] [Indexed: 11/11/2022]
Abstract
BACKGROUND Chronic rhinosinusitis (CRS) is a multifactorial disease with a high co-occurrence with asthma. In this multicohort study, we tested whether single nucleotide polymorphisms (SNPs) associated with childhood asthma and rhinovirus (RV)-associated disease are related to an increased susceptibility to adult CRS in a multicohort retrospective case-control study. METHODS Participants at two tertiary academic rhinology centers, University of Arizona (UofA) and University of Pennsylvania (UPenn) were recruited. Cases were defined as those with physician diagnosed CRS (UofA, n = 149; UPenn, n = 250), and healthy controls were those without CRS (UofA, n = 66; UPenn, n = 275). Genomic DNA was screened for the GSDMB rs7216389 SNP and CDHR3 rs6967330 SNP. Gene dosage, or the number of combined risk alleles in a single subject was calculated. Meta-analysis of the association between GSDMB or CDHR3 genotypes and CRS was performed and additive gene dosage effect for each population calculated using p for trend. RESULTS A meta-analysis revealed a combined increased risk for CRS in subjects with the GSDMB rs7216389 SNP (odds ratio [OR] 1.40; 95% confidence interval [CI], 1.16-1.76; p = 0.004). Both the UofA (OR 1.73; 95% CI, 1.23-2.43; p = 0.002) and UPenn (OR 1.27; 95% CI, 1.02-1.58; p = 0.035) populations showed a significant positive association between the number of combined risk alleles of GSDMB rs7216389 SNP and CDHR3 rs6967330 SNP and risk for CRS. CONCLUSION Carriers of the GSDMB rs7216389 SNP and CDHR3 rs6967330 SNP are at increased susceptibility for CRS. These data suggest that therapeutic approaches to target aberrant responses to RV infection may play a role in the treatment of unified airway disease.
Collapse
Affiliation(s)
- Dana E Zack
- Department of Otolaryngology, University of Arizona, Tucson, Arizona, USA
| | - Debra A Stern
- Asthma and Airway Disease Research Center, University of Arizona, Tucson, Arizona, USA
| | - Amanda L Willis
- Department of Otolaryngology, University of Arizona, Tucson, Arizona, USA
| | - Alexander S Kim
- Department of Otolaryngology, University of Arizona, Tucson, Arizona, USA
| | - Corinne J Mansfield
- Department of Otorhinolaryngology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Danielle R Reed
- Monell Chemical Senses Center, Philadelphia, Pennsylvania, USA
| | - Steven G Brooks
- Department of Otorhinolaryngology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Nithin D Adappa
- Department of Otorhinolaryngology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - James N Palmer
- Department of Otorhinolaryngology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Noam A Cohen
- Department of Otorhinolaryngology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Monell Chemical Senses Center, Philadelphia, Pennsylvania, USA
| | - Alexander G Chiu
- Department of Otolaryngology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Brian H Song
- Department of Otolaryngology, University of Arizona, Tucson, Arizona, USA
| | - Chris H Le
- Department of Otolaryngology, University of Arizona, Tucson, Arizona, USA
| | - Eugene H Chang
- Department of Otolaryngology, University of Arizona, Tucson, Arizona, USA
| |
Collapse
|
44
|
Sato N, Marubashi S. Induction of Immune Tolerance in Islet Transplantation Using Apoptotic Donor Leukocytes. J Clin Med 2021; 10:5306. [PMID: 34830586 PMCID: PMC8625503 DOI: 10.3390/jcm10225306] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Revised: 10/31/2021] [Accepted: 11/10/2021] [Indexed: 11/17/2022] Open
Abstract
Allogeneic islet transplantation has become an effective treatment option for severe Type 1 diabetes with intractable impaired awareness due to hypoglycemic events. Although current immunosuppressive protocols effectively prevent the acute rejection associated with initial T cell activation in recipients, chronic rejection has remained an obstacle for achieving long-term allogeneic islet engraftment. The development of donor-specific immune tolerance to the allograft is the ultimate goal given its potential ability to overcome chronic rejection and disregard the need for maintenance immunosuppression, which may be toxic to islet grafts. Recently, a breakthrough in tolerance induction during allogeneic islet transplantation using apoptotic donor lymphocytes (ADLs) in a non-human primate model had been reported. Several studies have suggested that the clonal depletion, anergy, and expansion of the antigen-specific regulatory immune network are the mechanisms for donor-specific tolerance with ADLs, which act synergistically to induce robust transplant tolerance. This achievement represents a huge step forward toward the clinical application of immune tolerance induction. We herein summarize the reported operational induction therapies in islet transplantation using the ADLs. Moreover, a few obstacles for the engraftment of transplanted islets, such as islet immunogenicity and instant blood-mediated response, which need to be resolved in the future, are also discussed.
Collapse
Affiliation(s)
| | - Shigeru Marubashi
- Department of Hepato–Biliary–Pancreatic and Transplant Surgery, Fukushima Medical University, Hikagigaoka-1, Fukushima 960-1295, Japan;
| |
Collapse
|
45
|
Li X, Zeng Q, Wang S, Li M, Chen X, Huang Y, Chen B, Zhou M, Lai Y, Guo C, Zhao S, Zhang H, Yang N. CRAC Channel Controls the Differentiation of Pathogenic B Cells in Lupus Nephritis. Front Immunol 2021; 12:779560. [PMID: 34745151 PMCID: PMC8569388 DOI: 10.3389/fimmu.2021.779560] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Accepted: 10/05/2021] [Indexed: 12/02/2022] Open
Abstract
Store-operated Ca2+ release-activated Ca2+ (CRAC) channel is the main Ca2+ influx pathway in lymphocytes and is essential for immune response. Lupus nephritis (LN) is an autoimmune disease characterized by the production of autoantibodies due to widespread loss of immune tolerance. In this study, RNA-seq analysis revealed that calcium transmembrane transport and calcium channel activity were enhanced in naive B cells from patients with LN. The increased expression of ORAI1, ORAI2, and STIM2 in naive B cells from patients with LN was confirmed by flow cytometry and Western blot, implying a role of CRAC channel in B-cell dysregulation in LN. For in vitro study, CRAC channel inhibition by YM-58483 or downregulation by ORAI1-specific small-interfering RNA (siRNA) decreased the phosphorylation of Ca2+/calmodulin-dependent protein kinase2 (CaMK2) and suppressed Blimp-1 expression in primary human B cells, resulting in decreased B-cell differentiation and immunoglobulin G (IgG) production. B cells treated with CaMK2-specific siRNA showed defects in plasma cell differentiation and IgG production. For in vivo study, YM-58483 not only ameliorated the progression of LN but also prevented the development of LN. MRL/lpr lupus mice treated with YM-58483 showed lower percentage of plasma cells in the spleen and reduced concentration of anti-double-stranded DNA antibodies in the sera significantly. Importantly, mice treated with YM-58483 showed decreased immune deposition in the glomeruli and alleviated kidney damage, which was further confirmed in NZM2328 lupus mice. Collectively, CRAC channel controlled the differentiation of pathogenic B cells and promoted the progression of LN. This study provides insights into the pathogenic mechanisms of LN and that CRAC channel could serve as a potential therapeutic target for LN.
Collapse
Affiliation(s)
- Xue Li
- Department of Rheumatology, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Qin Zeng
- Department of Rheumatology, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Shuyi Wang
- Department of Rheumatology, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Mengyuan Li
- Department of Rheumatology, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xionghui Chen
- Department of Nephrology, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yuefang Huang
- Department of Pediatrics, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Binfeng Chen
- Department of Rheumatology, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Mianjing Zhou
- Department of Rheumatology, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yimei Lai
- Department of Rheumatology, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Chaohuan Guo
- Department of Rheumatology, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Siyuan Zhao
- Department of Rheumatology, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Hui Zhang
- Department of Rheumatology, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Institute of Precision Medicine, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Niansheng Yang
- Department of Rheumatology, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
46
|
Santos DKDDN, Barros BRDS, Filho IJDC, Júnior NDSB, da Silva PR, Nascimento PHDB, Lima MDCAD, Napoleão TH, de Melo CML. Pectin-like polysaccharide extracted from the leaves of Conocarpus erectus Linnaeus promotes antioxidant, immunomodulatory and prebiotic effects. ACTA ACUST UNITED AC 2021. [DOI: 10.1016/j.bcdf.2021.100263] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
47
|
Deng X, Yao XQ, Berglund K, Dong B, Ouedraogo D, Ghane MA, Zhuo Y, McBean C, Wei ZZ, Gozem S, Yu SP, Wei L, Fang N, Mabb AM, Gadda G, Hamelberg D, Yang JJ. Tuning Protein Dynamics to Sense Rapid Endoplasmic-Reticulum Calcium Dynamics. Angew Chem Int Ed Engl 2021; 60:23289-23298. [PMID: 34436811 DOI: 10.1002/anie.202108443] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Indexed: 11/11/2022]
Abstract
Multi-scale calcium (Ca2+ ) dynamics, exhibiting wide-ranging temporal kinetics, constitutes a ubiquitous mode of signal transduction. We report a novel endoplasmic-reticulum (ER)-targeted Ca2+ indicator, R-CatchER, which showed superior kinetics in vitro (koff ≥2×103 s-1 , kon ≥7×106 M-1 s-1 ) and in multiple cell types. R-CatchER captured spatiotemporal ER Ca2+ dynamics in neurons and hotspots at dendritic branchpoints, enabled the first report of ER Ca2+ oscillations mediated by calcium sensing receptors (CaSRs), and revealed ER Ca2+ -based functional cooperativity of CaSR. We elucidate the mechanism of R-CatchER and propose a principle to rationally design genetically encoded Ca2+ indicators with a single Ca2+ -binding site and fast kinetics by tuning rapid fluorescent-protein dynamics and the electrostatic potential around the chromophore. The design principle is supported by the development of G-CatchER2, an upgrade of our previous (G-)CatchER with improved dynamic range. Our work may facilitate protein design, visualizing Ca2+ dynamics, and drug discovery.
Collapse
Affiliation(s)
- Xiaonan Deng
- Department of Chemistry, Center for Diagnostics and Therapeutics, Advanced Translational Imaging Facility, Georgia State University, 50 Decatur Street, 552 NSC, Atlanta, GA, 30303, USA
| | - Xin-Qiu Yao
- Department of Chemistry, Center for Diagnostics and Therapeutics, Advanced Translational Imaging Facility, Georgia State University, 50 Decatur Street, 552 NSC, Atlanta, GA, 30303, USA
| | - Ken Berglund
- Department of Neurosurgery, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Bin Dong
- Department of Chemistry, Center for Diagnostics and Therapeutics, Advanced Translational Imaging Facility, Georgia State University, 50 Decatur Street, 552 NSC, Atlanta, GA, 30303, USA
| | - Daniel Ouedraogo
- Department of Chemistry, Center for Diagnostics and Therapeutics, Advanced Translational Imaging Facility, Georgia State University, 50 Decatur Street, 552 NSC, Atlanta, GA, 30303, USA
| | - Mohammad A Ghane
- Neuroscience Institute, Georgia State University, Atlanta, GA, 30303, USA.,Center for Behavioral Neuroscience, Georgia State University, Atlanta, GA, 30303, USA
| | - You Zhuo
- Department of Chemistry, Center for Diagnostics and Therapeutics, Advanced Translational Imaging Facility, Georgia State University, 50 Decatur Street, 552 NSC, Atlanta, GA, 30303, USA
| | - Cheyenne McBean
- Department of Chemistry, Center for Diagnostics and Therapeutics, Advanced Translational Imaging Facility, Georgia State University, 50 Decatur Street, 552 NSC, Atlanta, GA, 30303, USA
| | - Zheng Zachory Wei
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Samer Gozem
- Department of Chemistry, Center for Diagnostics and Therapeutics, Advanced Translational Imaging Facility, Georgia State University, 50 Decatur Street, 552 NSC, Atlanta, GA, 30303, USA
| | - Shan P Yu
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Ling Wei
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Ning Fang
- Department of Chemistry, Center for Diagnostics and Therapeutics, Advanced Translational Imaging Facility, Georgia State University, 50 Decatur Street, 552 NSC, Atlanta, GA, 30303, USA
| | - Angela M Mabb
- Neuroscience Institute, Georgia State University, Atlanta, GA, 30303, USA.,Center for Behavioral Neuroscience, Georgia State University, Atlanta, GA, 30303, USA
| | - Giovanni Gadda
- Department of Chemistry, Center for Diagnostics and Therapeutics, Advanced Translational Imaging Facility, Georgia State University, 50 Decatur Street, 552 NSC, Atlanta, GA, 30303, USA
| | - Donald Hamelberg
- Department of Chemistry, Center for Diagnostics and Therapeutics, Advanced Translational Imaging Facility, Georgia State University, 50 Decatur Street, 552 NSC, Atlanta, GA, 30303, USA
| | - Jenny J Yang
- Department of Chemistry, Center for Diagnostics and Therapeutics, Advanced Translational Imaging Facility, Georgia State University, 50 Decatur Street, 552 NSC, Atlanta, GA, 30303, USA
| |
Collapse
|
48
|
Deng X, Yao X, Berglund K, Dong B, Ouedraogo D, Ghane MA, Zhuo Y, McBean C, Wei ZZ, Gozem S, Yu SP, Wei L, Fang N, Mabb AM, Gadda G, Hamelberg D, Yang JJ. Tuning Protein Dynamics to Sense Rapid Endoplasmic‐Reticulum Calcium Dynamics. Angew Chem Int Ed Engl 2021. [DOI: 10.1002/ange.202108443] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Xiaonan Deng
- Department of Chemistry Center for Diagnostics and Therapeutics Advanced Translational Imaging Facility Georgia State University 50 Decatur Street, 552 NSC Atlanta GA 30303 USA
| | - Xin‐Qiu Yao
- Department of Chemistry Center for Diagnostics and Therapeutics Advanced Translational Imaging Facility Georgia State University 50 Decatur Street, 552 NSC Atlanta GA 30303 USA
| | - Ken Berglund
- Department of Neurosurgery Emory University School of Medicine Atlanta GA 30322 USA
| | - Bin Dong
- Department of Chemistry Center for Diagnostics and Therapeutics Advanced Translational Imaging Facility Georgia State University 50 Decatur Street, 552 NSC Atlanta GA 30303 USA
| | - Daniel Ouedraogo
- Department of Chemistry Center for Diagnostics and Therapeutics Advanced Translational Imaging Facility Georgia State University 50 Decatur Street, 552 NSC Atlanta GA 30303 USA
| | - Mohammad A. Ghane
- Neuroscience Institute Georgia State University Atlanta GA 30303 USA
- Center for Behavioral Neuroscience Georgia State University Atlanta GA 30303 USA
| | - You Zhuo
- Department of Chemistry Center for Diagnostics and Therapeutics Advanced Translational Imaging Facility Georgia State University 50 Decatur Street, 552 NSC Atlanta GA 30303 USA
| | - Cheyenne McBean
- Department of Chemistry Center for Diagnostics and Therapeutics Advanced Translational Imaging Facility Georgia State University 50 Decatur Street, 552 NSC Atlanta GA 30303 USA
| | - Zheng Zachory Wei
- Department of Anesthesiology Emory University School of Medicine Atlanta GA 30322 USA
| | - Samer Gozem
- Department of Chemistry Center for Diagnostics and Therapeutics Advanced Translational Imaging Facility Georgia State University 50 Decatur Street, 552 NSC Atlanta GA 30303 USA
| | - Shan P. Yu
- Department of Anesthesiology Emory University School of Medicine Atlanta GA 30322 USA
| | - Ling Wei
- Department of Anesthesiology Emory University School of Medicine Atlanta GA 30322 USA
| | - Ning Fang
- Department of Chemistry Center for Diagnostics and Therapeutics Advanced Translational Imaging Facility Georgia State University 50 Decatur Street, 552 NSC Atlanta GA 30303 USA
| | - Angela M. Mabb
- Neuroscience Institute Georgia State University Atlanta GA 30303 USA
- Center for Behavioral Neuroscience Georgia State University Atlanta GA 30303 USA
| | - Giovanni Gadda
- Department of Chemistry Center for Diagnostics and Therapeutics Advanced Translational Imaging Facility Georgia State University 50 Decatur Street, 552 NSC Atlanta GA 30303 USA
| | - Donald Hamelberg
- Department of Chemistry Center for Diagnostics and Therapeutics Advanced Translational Imaging Facility Georgia State University 50 Decatur Street, 552 NSC Atlanta GA 30303 USA
| | - Jenny J. Yang
- Department of Chemistry Center for Diagnostics and Therapeutics Advanced Translational Imaging Facility Georgia State University 50 Decatur Street, 552 NSC Atlanta GA 30303 USA
| |
Collapse
|
49
|
Markakis I, Charitakis I, Beeton C, Galani M, Repousi E, Aggeloglou S, Sfikakis PP, Pennington MW, Chandy KG, Poulopoulou C. Kv1.3 Channel Up-Regulation in Peripheral Blood T Lymphocytes of Patients With Multiple Sclerosis. Front Pharmacol 2021; 12:714841. [PMID: 34630091 PMCID: PMC8495199 DOI: 10.3389/fphar.2021.714841] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 09/09/2021] [Indexed: 11/02/2022] Open
Abstract
Voltage-gated Kv1.3 potassium channels are key regulators of T lymphocyte activation, proliferation and cytokine production, by providing the necessary membrane hyper-polarization for calcium influx following immune stimulation. It is noteworthy that an accumulating body of in vivo and in vitro evidence links these channels to multiple sclerosis pathophysiology. Here we studied the electrophysiological properties and the transcriptional and translational expression of T lymphocyte Kv1.3 channels in multiple sclerosis, by combining patch clamp recordings, reverse transcription polymerase chain reaction and flow cytometry on freshly isolated peripheral blood T lymphocytes from two patient cohorts with multiple sclerosis, as well as from healthy and disease controls. Our data demonstrate that T lymphocytes in MS, manifest a significant up-regulation of Kv1.3 mRNA, Kv1.3 membrane protein and Kv1.3 current density and therefore of functional membrane channel protein, compared to control groups (p < 0.001). Interestingly, patient sub-grouping shows that Kv1.3 channel density is significantly higher in secondary progressive, compared to relapsing-remitting multiple sclerosis (p < 0.001). Taking into account the tight connection between Kv1.3 channel activity and calcium-dependent processes, our data predict and could partly explain the reported alterations of T lymphocyte function in multiple sclerosis, while they highlight Kv1.3 channels as potential therapeutic targets and peripheral biomarkers for the disease.
Collapse
Affiliation(s)
- Ioannis Markakis
- National and Kapodistrian University of Athens, Medical School, Athens, Greece.,Department of Neurology, "St. Panteleimon" General State Hospital, Nikaia, Greece
| | - Ioannis Charitakis
- National and Kapodistrian University of Athens, Medical School, Athens, Greece
| | - Christine Beeton
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, United States.,Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, United States
| | - Melpomeni Galani
- National and Kapodistrian University of Athens, Medical School, Athens, Greece
| | - Elpida Repousi
- National and Kapodistrian University of Athens, Medical School, Athens, Greece.,Department of Neurology, "St. Panteleimon" General State Hospital, Nikaia, Greece
| | - Stella Aggeloglou
- Department of Neurology, "St. Panteleimon" General State Hospital, Nikaia, Greece
| | - Petros P Sfikakis
- National and Kapodistrian University of Athens, Medical School, Athens, Greece
| | | | - K George Chandy
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, United States.,Lee Kong Chian School of Medicine, Nanyang Technological University, Nanyang Ave, Singapore
| | | |
Collapse
|
50
|
Bohmwald K, Gálvez NMS, Andrade CA, Mora VP, Muñoz JT, González PA, Riedel CA, Kalergis AM. Modulation of Adaptive Immunity and Viral Infections by Ion Channels. Front Physiol 2021; 12:736681. [PMID: 34690811 PMCID: PMC8531258 DOI: 10.3389/fphys.2021.736681] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 09/10/2021] [Indexed: 12/15/2022] Open
Abstract
Most cellular functions require of ion homeostasis and ion movement. Among others, ion channels play a crucial role in controlling the homeostasis of anions and cations concentration between the extracellular and intracellular compartments. Calcium (Ca2+) is one of the most relevant ions involved in regulating critical functions of immune cells, allowing the appropriate development of immune cell responses against pathogens and tumor cells. Due to the importance of Ca2+ in inducing the immune response, some viruses have evolved mechanisms to modulate intracellular Ca2+ concentrations and the mobilization of this cation through Ca2+ channels to increase their infectivity and to evade the immune system using different mechanisms. For instance, some viral infections require the influx of Ca2+ through ionic channels as a first step to enter the cell, as well as their replication and budding. Moreover, through the expression of viral proteins on the surface of infected cells, Ca2+ channels function can be altered, enhancing the pathogen evasion of the adaptive immune response. In this article, we review those ion channels and ion transporters that are essential for the function of immune cells. Specifically, cation channels and Ca2+ channels in the context of viral infections and their contribution to the modulation of adaptive immune responses.
Collapse
Affiliation(s)
- Karen Bohmwald
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Nicolás M. S. Gálvez
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Catalina A. Andrade
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Valentina P. Mora
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - José T. Muñoz
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Pablo A. González
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Claudia A. Riedel
- Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Millennium Institute on Immunology and Immunotherapy, Universidad Andres Bello, Santiago, Chile
| | - Alexis M. Kalergis
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
- Departamento de Endocrinología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|