51
|
Liu Z, Wu S, Zhang W, Cui H, Zhang J, Yin X, Zheng X, Shen T, Ying H, Chen L, Wang H, Jiang J. Cordycepin mitigates dextran sulfate sodium-induced colitis through improving gut microbiota composition and modulating Th1/Th2 and Th17/Treg balance. Biomed Pharmacother 2024; 180:117394. [PMID: 39395256 DOI: 10.1016/j.biopha.2024.117394] [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: 06/24/2024] [Revised: 08/20/2024] [Accepted: 08/30/2024] [Indexed: 10/14/2024] Open
Abstract
BACKGROUND Imbalances in Th1/Th2 and Th17/Treg immune axes, coupled with disruptions in the gut microbiota (GM), play a pivotal role in the pathogenesis of inflammatory bowel disease (IBD). Cordycepin, a natural anti-inflammatory compound, holds promise in mitigating IBD by rebalancing these immune axes in conjunction with modulating the GM. The aim of this experiment is to investigate the potential of cordycepin in mitigating enteritis and elucidate the underlying mechanisms associated with its ameliorative effects on enteritis. METHODS On the day of inducing experimental colitis with Dextran Sulfate Sodium (DSS), mice in the DSS + Cordycepin and Cordycepin groups received 50 mg/kg/day Cordycepin via intra-gastric administration (i.g.) for seven consecutive days, respectively. Mice in the DSS and control groups were treated with equal volumes of saline. On day 8, all mice were euthanized under pentobarbital sodium anesthesia. RESULTS In a DSS-induced colitis mouse model, Cordycepin treatment led to a significant reduction in the disease activity index (DAI), splenic weight, and colonic pathological injury while simultaneously improving body weight and colonic length. Furthermore, it positively impacted GM composition, resulting in decreased Th1 and Th17 cells, alongside an increase in Th2 and Treg cells. The contents of the mouse colon were extracted for microbial community analysis. Mouse blood was prepared into a single-cell suspension, and flow cytometry was used to assess the expressio of Treg, Th17, Th1, and Th2 immune cells. CONCLUSIONS These results underscored the effective intervention of cordycepin in ameliorating DSS-induced colitis by harmonizing the interplay between GM and immune homeostasis.
Collapse
Affiliation(s)
- Zhilin Liu
- Department of Gastrointestinal Surgery, the Third Affiliated Hospital of Soochow University, Changzhou 213003, China; Department of Tumor Biological Treatment, the Third Affiliated Hospital of Soochow University, Changzhou 213003, China; Jiangsu Engineering Research Center for Tumor Immunotherapy, the Third Affiliated Hospital of Soochow University, Changzhou 213003, China; Institute of Cell Therapy, the Third Affiliated Hospital of Soochow University, Changzhou 213003, China
| | - Shaoxian Wu
- Department of Tumor Biological Treatment, the Third Affiliated Hospital of Soochow University, Changzhou 213003, China; Jiangsu Engineering Research Center for Tumor Immunotherapy, the Third Affiliated Hospital of Soochow University, Changzhou 213003, China; Institute of Cell Therapy, the Third Affiliated Hospital of Soochow University, Changzhou 213003, China
| | - Wenting Zhang
- Affiliated Changzhou Children's Hospital of Nantong University, Changzhou Children's Hospital, Changzhou 213003, China
| | - Hengwei Cui
- Department of Gastrointestinal Surgery, the Third Affiliated Hospital of Soochow University, Changzhou 213003, China
| | - Jingfeng Zhang
- Department of Gastrointestinal Surgery, the Third Affiliated Hospital of Soochow University, Changzhou 213003, China
| | - Xuan Yin
- Department of Tumor Biological Treatment, the Third Affiliated Hospital of Soochow University, Changzhou 213003, China; Jiangsu Engineering Research Center for Tumor Immunotherapy, the Third Affiliated Hospital of Soochow University, Changzhou 213003, China; Institute of Cell Therapy, the Third Affiliated Hospital of Soochow University, Changzhou 213003, China
| | - Xiao Zheng
- Department of Tumor Biological Treatment, the Third Affiliated Hospital of Soochow University, Changzhou 213003, China; Jiangsu Engineering Research Center for Tumor Immunotherapy, the Third Affiliated Hospital of Soochow University, Changzhou 213003, China; Institute of Cell Therapy, the Third Affiliated Hospital of Soochow University, Changzhou 213003, China
| | - Tao Shen
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Jiangsu, Nanjing, China
| | - Hanjie Ying
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Jiangsu, Nanjing, China
| | - Lujun Chen
- Department of Tumor Biological Treatment, the Third Affiliated Hospital of Soochow University, Changzhou 213003, China; Jiangsu Engineering Research Center for Tumor Immunotherapy, the Third Affiliated Hospital of Soochow University, Changzhou 213003, China; Institute of Cell Therapy, the Third Affiliated Hospital of Soochow University, Changzhou 213003, China.
| | - Haitao Wang
- Department of Gastrointestinal Surgery, the Third Affiliated Hospital of Soochow University, Changzhou 213003, China.
| | - Jingting Jiang
- Department of Tumor Biological Treatment, the Third Affiliated Hospital of Soochow University, Changzhou 213003, China; Jiangsu Engineering Research Center for Tumor Immunotherapy, the Third Affiliated Hospital of Soochow University, Changzhou 213003, China; Institute of Cell Therapy, the Third Affiliated Hospital of Soochow University, Changzhou 213003, China.
| |
Collapse
|
52
|
Wong J, Yang Z, Wang L, Sun F, Yue GH. Tbx21 gene and its association with resistance against viral nervous necrosis (VNN) in Asian seabass, Lates calcarifer. MARINE LIFE SCIENCE & TECHNOLOGY 2024; 6:679-689. [PMID: 39620089 PMCID: PMC11602908 DOI: 10.1007/s42995-024-00234-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 04/29/2024] [Indexed: 06/02/2025]
Abstract
Viral nervous necrosis (VNN) caused by a betanodavirus (NNV) is one of the major diseases in Asian seabass (Lates calcarifer) hatcheries. Our previous studies showed that the tbx21 gene was in a QTL for NNV resistance in linkage group 23 in Asian seabass. The expression of this gene was changed in tissues of Asian seabass challenged with NNV. However, the role of tbx21 in NNV resistance remains largely unknown. In this study, tbx21 of Asian seabass was characterized. This gene consists of an ORF of 1866 bp, a 5' UTR of 357 bp, and a 3' UTR of 4674 bp. The TBX21 protein showed substantial amino acid similarity (70-96%) with other fish but exhibited lower identity (47-52%) with mammals. One SNP identified in the first intron was significantly associated with NNV resistance. In healthy fish, tbx21 was expressed in all tissues examined, and was highly expressed in the kidney and liver. The expression of tbx21 increased in the eye, gills, heart, kidney and gut, but decreased in the brain and spleen at five days after NNV challenge. Overexpression of tbx21 reduced the replication of NNV, whereas knockdown increased viral expression and virus titers. These results suggest that tbx21 plays a key role in NNV resistance. The SNP in this gene could be used as a marker to facilitate marker-assisted selection for NNV resistance. Further investigation of polymorphisms in the 5' and 3' UTRs of tbx21 may provide additional insights into the gene's role in NNV resistance. Supplementary Information The online version contains supplementary material available at 10.1007/s42995-024-00234-0.
Collapse
Affiliation(s)
- Joey Wong
- Temasek Life Sciences Laboratory, National University of Singapore, Singapore, 117604 Singapore
| | - Zituo Yang
- Temasek Life Sciences Laboratory, National University of Singapore, Singapore, 117604 Singapore
| | - Le Wang
- Temasek Life Sciences Laboratory, National University of Singapore, Singapore, 117604 Singapore
| | - Fei Sun
- Temasek Life Sciences Laboratory, National University of Singapore, Singapore, 117604 Singapore
| | - Gen Hua Yue
- Temasek Life Sciences Laboratory, National University of Singapore, Singapore, 117604 Singapore
- Department of Biological Sciences, National University of Singapore, Singapore, 117543 Singapore
| |
Collapse
|
53
|
Yee YC, Nakamura A, Okada Y, Mori T, Katayama Y. Establishment of an in vitro evaluation method for immunomodulatory functions of yeast strains. ANAL SCI 2024; 40:2043-2051. [PMID: 39097563 DOI: 10.1007/s44211-024-00641-4] [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: 06/26/2024] [Accepted: 07/23/2024] [Indexed: 08/05/2024]
Abstract
Saccharomyces cerevisiae, a widely studied yeast known for its industrial applications, is increasingly recognized for its potential in immunomodulation. This study aimed to systematically analyze and compare the immune-modulating properties of various S. cerevisiae strains under controlled experimental conditions. Three essential signals crucial for immune response activation were evaluated to elucidate the immunological responses elicited by these strains, i.e., dendritic cells (DC) cytokine secretion profiles, maturation status, and T cell polarization. Analysis of DC cytokine secretion profiles and maturation status revealed that all tested yeast strains induced DC activation, characterized by significant IL-6 secretion and modest IL-10 induction, as well as upregulation of MHC II molecules. Additionally, strain-specific effects were observed, particularly, strain AJM109 and Y1383 uniquely enhanced CD86 and PD-L1 expression, respectively, suggesting differential impacts on DC co-stimulatory signaling. Furthermore, strain Y1383 showed a unique capacity to support Treg-mediated immune suppression, demonstrating its potential in immune tolerance induction. These findings underscore the complexity of S. cerevisiae-based immune modulation and emphasize the importance of standardized evaluation methods to distinguish their specific immunological effects.
Collapse
Affiliation(s)
- Ying Chuin Yee
- Department of Applied Chemistry, Graduate School of Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka, 819-0395, Japan
| | - Akihiro Nakamura
- Research and Development Laboratory, Sanwa Shurui Co., Ltd., 2231-1 Yamamoto, Usa, Oita, 879-0495, Japan
| | - Yoshikiyo Okada
- Department of Internal Medicine, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama, 359-8513, Japan
| | - Takeshi Mori
- Department of Applied Chemistry, Graduate School of Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka, 819-0395, Japan.
- Center for Future Chemistry, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka, 819-0395, Japan.
| | - Yoshiki Katayama
- Department of Applied Chemistry, Graduate School of Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka, 819-0395, Japan.
- Center for Future Chemistry, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka, 819-0395, Japan.
- Center for Molecular Systems, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka, 819-0395, Japan.
- Centre for Advanced Medicine Open Innovation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan.
- Department of Biomedical Engineering, Chung Yuan Christian University, 200 Chung Pei Rd., Chung Li, Taoyüan, 32023, Taiwan, ROC.
| |
Collapse
|
54
|
Hu L, Sun C, Yuan K, Yang P. Expression, regulation, and function of PD-L1 on non-tumor cells in the tumor microenvironment. Drug Discov Today 2024; 29:104181. [PMID: 39278561 DOI: 10.1016/j.drudis.2024.104181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Revised: 09/03/2024] [Accepted: 09/11/2024] [Indexed: 09/18/2024]
Abstract
Antiprogrammed death ligand 1 (PD-L1) therapy is a leading immunotherapy, but only some patients with solid cancers benefit. Overwhelming evidence has revealed that PD-L1 is expressed on various immune cells in the tumor microenvironment (TME), including macrophages, dendritic cells, and regulatory T cells, modulating tumor immunity and influencing tumor progression. PD-L1 can also be located on tumor cell membranes as well as in exosomes and cytoplasm. Accordingly, the dynamic expression and various forms of PD-L1 might explain the therapy's limited efficacy and resistance. Herein a systematic summary of the expression of PD-L1 on different immune cells and their regulatory mechanisms is provided to offer a solid foundation for future studies.
Collapse
Affiliation(s)
- Lingrong Hu
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China; Institute of Innovative Drug Discovery and Development, China Pharmaceutical University, Nanjing 211198, China
| | - Chengliang Sun
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China; Institute of Innovative Drug Discovery and Development, China Pharmaceutical University, Nanjing 211198, China
| | - Kai Yuan
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China; Institute of Innovative Drug Discovery and Development, China Pharmaceutical University, Nanjing 211198, China.
| | - Peng Yang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China; Institute of Innovative Drug Discovery and Development, China Pharmaceutical University, Nanjing 211198, China.
| |
Collapse
|
55
|
Baiju I, Kumar Bharti M, Somal A, Pandey S, Bhat IA, Joseph A, Chandra V, Taru Sharma G. Exploration of immunomodulatory mechanism of caprine Wharton's jelly derived mesenchymal stem cells. Cell Immunol 2024; 405-406:104879. [PMID: 39305581 DOI: 10.1016/j.cellimm.2024.104879] [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: 07/17/2024] [Accepted: 09/15/2024] [Indexed: 12/02/2024]
Abstract
The present study was aimed to explore the possible mechanisms by which caprine Wharton's jelly-derived MSCs (WJ-MSCs) perform their immunomodulatory function. WJ-MSCs were isolated through explants culture and characterized as per ISCT criteria using culture behavior, expression of surface markers by PCR, FACS and immunocytochemical localization (ICC), trilineage differentiation potential etc. Secretory behavior for important biomolecules (IDO, TGFβ1, VEGF, IL6) was evaluated by ICC and western blot assay. Cell-to-cell communication was studied by culturing cells in cell-cell contact and trans-well system. The MSCs when co-cultured with activated Tc and Th cells, down-regulation of T cell cytokine as well as upregulation of immunomodulatory factors (VEGF A, IL10, IL6, IDO, iNOS, PTGS2, HGF, TGFβ, CXCL10, CXCL11) was noticed in both cell-cell contact and trans-well culture system which was significantly higher in cell-cell contact system. Trilineage differentiation of MSCs showed significant upregulation of MHC I (CAHI) and MHC II (CLA DRB3) molecules suggesting better clinical applications of MSCs without differentiation to avoid immune rejection. It can be concluded that WJ-MSCs perform their immunomodulation through the secretion of a battery of biomolecules and work in both cell-cell contact manner and through their secretome.
Collapse
Affiliation(s)
- Indu Baiju
- Division of Physiology & Climatology, ICAR-Indian Veterinary Research Institute, Izatnagar 243122 UP India
| | - Mukesh Kumar Bharti
- Division of Physiology & Climatology, ICAR-Indian Veterinary Research Institute, Izatnagar 243122 UP India; Institute of Agricultural Sciences, RGSC, Banaras Hindu University, Barkachha, Mirzapur 231001 UP India
| | - Anjali Somal
- Division of Physiology & Climatology, ICAR-Indian Veterinary Research Institute, Izatnagar 243122 UP India; Chaudhary Sarwan Kumar Himachal Pradesh Krishi Vishvavidyalaya, Palampur HP India
| | - Sriti Pandey
- Division of Physiology & Climatology, ICAR-Indian Veterinary Research Institute, Izatnagar 243122 UP India; Nanaji Deshmukh Veterinary Science University, Jabalpur 482001 MP India
| | - Irfan A Bhat
- Division of Physiology & Climatology, ICAR-Indian Veterinary Research Institute, Izatnagar 243122 UP India
| | - Anand Joseph
- Division of Physiology & Climatology, ICAR-Indian Veterinary Research Institute, Izatnagar 243122 UP India
| | - Vikash Chandra
- Division of Physiology & Climatology, ICAR-Indian Veterinary Research Institute, Izatnagar 243122 UP India.
| | - G Taru Sharma
- Division of Physiology & Climatology, ICAR-Indian Veterinary Research Institute, Izatnagar 243122 UP India; National Institute of Animal Biotechnology, Hyderabad 500 032 Telangana India.
| |
Collapse
|
56
|
Kopetz S, Murphy DA, Pu J, Ciardiello F, Desai J, Van Cutsem E, Wasan HS, Yoshino T, Saffari H, Zhang X, Hamilton P, Xie T, Yaeger R, Tabernero J. Molecular profiling of BRAF-V600E-mutant metastatic colorectal cancer in the phase 3 BEACON CRC trial. Nat Med 2024; 30:3261-3271. [PMID: 39313594 PMCID: PMC11564101 DOI: 10.1038/s41591-024-03235-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 08/06/2024] [Indexed: 09/25/2024]
Abstract
The BEACON CRC study demonstrated that encorafenib (Enco)+cetuximab (Cetux)±binimetinib (Bini) significantly improved overall survival (OS) versus Cetux + chemotherapy in previously treated patients with BRAF-V600E-mutant mCRC, providing the basis for the approval of the Enco+Cetux regimen in the United States and the European Union. A greater understanding of biomarkers predictive of response to Enco+Cetux±Bini treatment is of clinical relevance. In this prespecified, exploratory biomarker analysis of the BEACON CRC study, we characterize genomic and transcriptomic correlates of clinical outcomes and acquired resistance mechanisms through integrated clinical and molecular analysis, including whole-exome and -transcriptome tissue sequencing and circulating tumor DNA genomic profiling. Tumors with higher immune signatures showed a trend towards increased OS benefit with Enco+Bini+Cetux. RAS, MAP2K1 and MET alterations were most commonly acquired with Enco+Cetux±Bini, and more frequent in patients with a high baseline cell-cycle gene signature; baseline TP53 mutation was associated with acquired MET amplification. Acquired mutations were subclonal and polyclonal, with evidence of increased tumor mutation rate with Enco+Cetux±Bini and mutational signatures (SBS17a/b). These findings support treatment with Enco+Cetux±Bini for patients with BRAF-V600E-mutant mCRC and provide insights into the biology of response and resistance to MAPK-pathway-targeted therapy. ClinicalTrials.gov registration: NCT02928224.
Collapse
Affiliation(s)
- Scott Kopetz
- University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | | | - Jie Pu
- Pfizer, New York, NY, USA
| | | | - Jayesh Desai
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Eric Van Cutsem
- University Hospitals Gasthuisberg Leuven and KU Leuven, Leuven, Belgium
| | | | | | | | | | | | | | - Rona Yaeger
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Josep Tabernero
- Vall d'Hebron Hospital Campus and Vall d'Hebron Institute of Oncology (VHIO), University of Vic-Central University of Catalonia, Barcelona, Spain
| |
Collapse
|
57
|
Maiti G, Frikeche J, Loomis C, Chakravarti S. Paracrine regulations of IFN-γ secreting CD4 + T cells by lumican and biglycan are protective in allergic contact dermatitis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.20.619307. [PMID: 39484444 PMCID: PMC11526879 DOI: 10.1101/2024.10.20.619307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
The extracellular matrix (ECM) is known to regulate innate immune cells but its role in T cell functions is poorly understood. Here, we show a protective role for ECM proteoglycans, lumican and biglycan in hapten-induced contact dermatitis that is achieved through limiting proinflammatory CD4 + T cells. Lumican and biglycan-null mice develop significant inflammation with greater numbers of CD4 + T cells in hapten-challenged ear pinnae, while their draining lymph nodes show increased T-bet-STAT1 signaling, Th1 commitment, and IFN-γ secreting CD4 + T cell proliferation. Wild type mouse lymph node fibroblastic reticular cells secrete lumican, biglycan and decorin, a related proteoglycan, while none are expressed by naive or activated T cells. In vitro , lumican and biglycan co-localize with LFA-1 on T cell surfaces, and all three proteoglycans suppress LFA-1 mediated T cell activation. Overall, this study elucidates a novel paracrine regulation of Th1 cells by ECM proteoglycans to limit inflammation and tissue damage.
Collapse
|
58
|
Daga N, Servaas NH, Kisand K, Moonen D, Arnold C, Reyes-Palomares A, Kaleviste E, Kingo K, Kuuse R, Ulst K, Steinmetz L, Peterson P, Nakic N, Zaugg JB. Integration of genetic and chromatin modification data pinpoints autoimmune-specific remodeling of enhancer landscape in CD4 + T cells. Cell Rep 2024; 43:114810. [PMID: 39388354 DOI: 10.1016/j.celrep.2024.114810] [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: 12/23/2023] [Revised: 07/16/2024] [Accepted: 09/16/2024] [Indexed: 10/12/2024] Open
Abstract
CD4+ T cells play a crucial role in adaptive immune responses and have been implicated in the pathogenesis of autoimmune diseases (ADs). Despite numerous studies, the molecular mechanisms underlying T cell dysregulation in ADs remain incompletely understood. Here, we used chromatin immunoprecipitation (ChIP)-sequencing of active chromatin and transcriptomic data from CD4+ T cells of healthy donors and patients with systemic lupus erythematosus (SLE), psoriasis, juvenile idiopathic arthritis (JIA), and Graves' disease to investigate the role of enhancers in AD pathogenesis. By generating enhancer-based gene regulatory networks (eGRNs), we identified disease-specific dysregulated pathways and potential downstream target genes of enhancers harboring AD-associated single-nucleotide polymorphisms (SNPs), which we also validated using chromatin-capture (HiC) data and CRISPR interference (CRISPRi) in primary CD4+ T cells. Our results suggest that alterations in the regulatory landscapes of CD4+ T cells, including enhancers, contribute to the development of ADs and provide a basis for developing new therapeutic approaches.
Collapse
Affiliation(s)
- Neha Daga
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Nila H Servaas
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Kai Kisand
- Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Dewi Moonen
- Genome Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Christian Arnold
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Armando Reyes-Palomares
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Epp Kaleviste
- Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Külli Kingo
- Department of Dermatology and Venerology, Faculty of Medicine, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia and Dermatology Clinic, Tartu University Hospital, Tartu, Estonia
| | - Reet Kuuse
- Department of Internal Medicine, Tartu University Hospital, Tartu, Estonia
| | - Katrin Ulst
- Department of Internal Medicine, Tartu University Hospital, Tartu, Estonia
| | - Lars Steinmetz
- Genome Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany; Department of Genetics, Stanford University, Stanford, CA, USA
| | - Pärt Peterson
- Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Nikolina Nakic
- Functional Genomics, Medicinal Science and Technology, GSK R&D, Stevenage, UK
| | - Judith B Zaugg
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany.
| |
Collapse
|
59
|
Chia JE, Rousseau RP, Ozturk M, Poswayo SKL, Lucas R, Brombacher F, Parihar SP. The divergent outcome of IL-4Rα signalling on Foxp3 T regulatory cells in listeriosis and tuberculosis. Front Immunol 2024; 15:1427055. [PMID: 39483462 PMCID: PMC11524857 DOI: 10.3389/fimmu.2024.1427055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Accepted: 09/18/2024] [Indexed: 11/03/2024] Open
Abstract
Introduction Forkhead box P3 (Foxp3) T regulatory cells are critical for maintaining self-tolerance, immune homeostasis, and regulating the immune system. Methods We investigated interleukin-4 receptor alpha (IL-4Rα) signalling on T regulatory cells (Tregs) during Listeria monocytogenes (L. monocytogenes) infection using a mouse model on a BALB/c background, specifically with IL-4Rα knockdown in Tregs (Foxp3creIL-4Rα-/lox). Results We showed an impairment of Treg responses, along with a decreased bacterial burden and diminished tissue pathology in the liver and spleen, which translated into better survival. Mechanistically, we observed an enhancement of the Th1 signature, characterised by increased expression of the T-bet transcription factor and a greater number of effector T cells producing IFN-γ, IL-2 following ex-vivo stimulation with heat-killed L. monocytogenes in Foxp3creIL-4Rα-/lox mice. Furthermore, CD8 T cells from Foxp3creIL-4Rα-/lox mice displayed increased cytotoxicity (Granzyme-B) with higher proliferation capacity (Ki-67), better survival (Bcl-2) with concomitant reduced apoptosis (activated caspase 3). In contrast to L. monocytogenes, Foxp3creIL-4Rα-/lox mice displayed similar bacterial burdens, lung pathology and survival during Mycobacterium tuberculosis (M. tuberculosis) infection, despite increased T cell numbers and IFN-γ, TNF and IL-17 production. Conclusion Our results demonstrated that the diminished IL-4Rα signalling on Foxp3+ T regulatory cells resulted in a loss of their functionality, leading to survival benefits in listeriosis but not in tuberculosis.
Collapse
Affiliation(s)
- Julius E. Chia
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town Component, Cape Town, South Africa
- Division of Immunology, Institute of Infectious Diseases and Molecular Medicine (IDM), Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Robert P. Rousseau
- Division of Immunology, Institute of Infectious Diseases and Molecular Medicine (IDM), Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- Centre for Infectious Diseases Research in Africa (CIDRI-Africa), Institute of Infectious Diseases and Molecular Medicine (IDM), Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Mumin Ozturk
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town Component, Cape Town, South Africa
- Division of Immunology, Institute of Infectious Diseases and Molecular Medicine (IDM), Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Sibongiseni K. L. Poswayo
- Division of Immunology, Institute of Infectious Diseases and Molecular Medicine (IDM), Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- Centre for Infectious Diseases Research in Africa (CIDRI-Africa), Institute of Infectious Diseases and Molecular Medicine (IDM), Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Rodney Lucas
- Research Animal Facility (RAF), Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Frank Brombacher
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town Component, Cape Town, South Africa
- Division of Immunology, Institute of Infectious Diseases and Molecular Medicine (IDM), Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- Centre for Infectious Diseases Research in Africa (CIDRI-Africa), Institute of Infectious Diseases and Molecular Medicine (IDM), Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Suraj P. Parihar
- Division of Immunology, Institute of Infectious Diseases and Molecular Medicine (IDM), Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- Centre for Infectious Diseases Research in Africa (CIDRI-Africa), Institute of Infectious Diseases and Molecular Medicine (IDM), Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- Division of Medical Microbiology, Institute of Infectious Diseases and Molecular Medicine (IDM), Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- Division of Human Metabolomics, North-West University, Potchefstroom, South Africa
| |
Collapse
|
60
|
Sung CJ, Whitmore LS, Smith E, Chang J, Tisoncik-Go J, Barber-Axthelm A, Selseth A, Feltham S, Ojha S, Hansen SG, Picker LJ, Gale M. Functional genomic analysis of the 68-1 RhCMV- Mycobacteria tuberculosis vaccine reveals an IL-15 response signature that is conserved with vector attenuation. Front Immunol 2024; 15:1460344. [PMID: 39474415 PMCID: PMC11518738 DOI: 10.3389/fimmu.2024.1460344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 09/26/2024] [Indexed: 11/14/2024] Open
Abstract
Tuberculosis (TB), caused by Mycobacterium tuberculosis (Mtb) is a deadly infectious disease having a major impact on global health. Using the CMV vector for development of novel vaccines is a promising new strategy that elicits strong and durable, high frequency memory T cell responses against heterologous immunogens. We conducted functional transcriptomic analysis of whole blood samples collected from cohorts of rhesus (Rh) macaques that were administered RhCMV/TB vector using a prime-boost strategy. Two modified CMV vectors were used in this study, including 68-1 RhCMV/TB-6Ag (encoding 6 Mtb protein immunogens, including Ag85A, ESAT-6, Rv3407, Rv2626, Rpf A, and Rpf D) and its attenuated variant, 68-1 RhCMV/Δpp71-TB-6Ag (a cell-to-cell spread-deficient vaccine vector lacking the Rh110 gene encoding the pp71 tegument protein). Bulk mRNA sequencing, differential gene expression, and functional enrichment analyses showed that these RhCMV/TB vaccines induce the innate and adaptive immune responses with specific transcriptomic signatures, including the IL-15-induced protective gene signature previously defined to be linked with protection against simian immunodeficiency virus (SIV) by the 68-1 RhCMV/SIV vaccine. While both vectors exhibited a transcriptomic response of the IL-15 protective signature in whole blood, we show that lack of pp71 does not maintain induction of the protective signature for the full duration of the study compared to the parental non-attenuated vector. Our observations indicate that RhCMV vector vaccines induce a transcriptomic response in whole blood that include a conserved IL-15 signature of which vector-encoded pp71 is an important component of response durability that upon future Mtb challenge may define specific vaccine protection outcomes against Mtb infection.
Collapse
Affiliation(s)
- Cheng-Jung Sung
- Center for Innate Immunity and Immune Disease, Department of Immunology, University of Washington, Seattle, WA, United States
| | - Leanne S. Whitmore
- Center for Innate Immunity and Immune Disease, Department of Immunology, University of Washington, Seattle, WA, United States
| | - Elise Smith
- Center for Innate Immunity and Immune Disease, Department of Immunology, University of Washington, Seattle, WA, United States
| | - Jean Chang
- Center for Innate Immunity and Immune Disease, Department of Immunology, University of Washington, Seattle, WA, United States
| | - Jennifer Tisoncik-Go
- Center for Innate Immunity and Immune Disease, Department of Immunology, University of Washington, Seattle, WA, United States
- Washington National Primate Research Center, Seattle, WA, United States
| | - Aaron Barber-Axthelm
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR, United States
| | - Andrea Selseth
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR, United States
| | - Shana Feltham
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR, United States
| | - Sohita Ojha
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR, United States
| | - Scott G. Hansen
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR, United States
| | - Louis J. Picker
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR, United States
| | - Michael Gale
- Center for Innate Immunity and Immune Disease, Department of Immunology, University of Washington, Seattle, WA, United States
- Washington National Primate Research Center, Seattle, WA, United States
| |
Collapse
|
61
|
Xie C, Yang J, Gul A, Li Y, Zhang R, Yalikun M, Lv X, Lin Y, Luo Q, Gao H. Immunologic aspects of asthma: from molecular mechanisms to disease pathophysiology and clinical translation. Front Immunol 2024; 15:1478624. [PMID: 39439788 PMCID: PMC11494396 DOI: 10.3389/fimmu.2024.1478624] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Accepted: 09/18/2024] [Indexed: 10/25/2024] Open
Abstract
In the present review, we focused on recent translational and clinical discoveries in asthma immunology, facilitating phenotyping and stratified or personalized interventions for patients with this condition. The immune processes behind chronic inflammation in asthma exhibit marked heterogeneity, with diverse phenotypes defining discernible features and endotypes illuminating the underlying molecular mechanisms. In particular, two primary endotypes of asthma have been identified: "type 2-high," characterized by increased eosinophil levels in the airways and sputum of patients, and "type 2-low," distinguished by increased neutrophils or a pauci-granulocytic profile. Our review encompasses significant advances in both innate and adaptive immunities, with emphasis on the key cellular and molecular mediators, and delves into innovative biological and targeted therapies for all the asthma endotypes. Recognizing that the immunopathology of asthma is dynamic and continuous, exhibiting spatial and temporal variabilities, is the central theme of this review. This complexity is underscored through the innumerable interactions involved, rather than being driven by a single predominant factor. Integrated efforts to improve our understanding of the pathophysiological characteristics of asthma indicate a trend toward an approach based on disease biology, encompassing the combined examination of the clinical, cellular, and molecular dimensions of the disease to more accurately correlate clinical traits with specific disease mechanisms.
Collapse
Affiliation(s)
- Cong Xie
- Department of Endocrinology and Clinical Immunology, Yuquan Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China
- Department of Integrative Medicine, Huashan Hospital Affiliated to Fudan University, Fudan Institutes of Integrative Medicine, Fudan University Shanghai Medical College, Shanghai, China
| | - Jingyan Yang
- The Third Affiliated Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Aman Gul
- Department of Integrative Medicine, Huashan Hospital Affiliated to Fudan University, Fudan Institutes of Integrative Medicine, Fudan University Shanghai Medical College, Shanghai, China
- Department of Respiratory Medicine, Uyghur Medicines Hospital of Xinjiang Uyghur Autonomous Region, Urumqi, China
- College of Life Science and Technology, Xinjiang University, Urumqi, China
| | - Yifan Li
- Department of Integrative Medicine, Huashan Hospital Affiliated to Fudan University, Fudan Institutes of Integrative Medicine, Fudan University Shanghai Medical College, Shanghai, China
| | - Rui Zhang
- Department of Pulmonary and Critical Care Medicine, Shenzhen Hospital of Guangzhou University of Chinese Medicine (Futian), Shenzhen, China
| | - Maimaititusun Yalikun
- Department of Integrative Medicine, Huashan Hospital Affiliated to Fudan University, Fudan Institutes of Integrative Medicine, Fudan University Shanghai Medical College, Shanghai, China
| | - Xiaotong Lv
- Department of Cardiology, The Second Affiliated Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yuhan Lin
- Department of Endocrinology and Clinical Immunology, Yuquan Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Qingli Luo
- Department of Integrative Medicine, Huashan Hospital Affiliated to Fudan University, Fudan Institutes of Integrative Medicine, Fudan University Shanghai Medical College, Shanghai, China
| | - Huijuan Gao
- Department of Endocrinology and Clinical Immunology, Yuquan Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China
| |
Collapse
|
62
|
Curci D, Lucafò M, Decorti G, Stocco G. Monoclonal antibodies against pediatric ulcerative colitis: a review of clinical progress. Expert Opin Biol Ther 2024; 24:1133-1144. [PMID: 39285823 DOI: 10.1080/14712598.2024.2404076] [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: 05/20/2024] [Accepted: 09/10/2024] [Indexed: 09/21/2024]
Abstract
INTRODUCTION In children, ulcerative colitis (UC) is often more severe and extensive than in adults and hospitalization for acute exacerbations occurs in around a quarter of subjects. There is a need for effective drugs, which could avoid or reduce the use of corticosteroids which, especially in children, are burdened by a number of severe side effects. The introduction in therapy of monoclonal antibodies has completely changed the therapeutic scenario and the prognosis of the disease. AREAS COVERED In this review, the use of the monoclonal antibodies directed against tumor necrosis factor (TNF)α or other inflammatory targets for the treatment of pediatric UC will be discussed. A search of the literature was done using the keywords 'pediatric,' 'ulcerative colitis,' 'inflammatory bowel disease,' 'monoclonal antibodies;' 'infliximab,' 'adalimumab,' 'golimumab,' vedolizumab," 'ustekinumab' and 'risankizumab.' EXPERT OPINION The use of monoclonal antibodies has greatly increased in recent years in pediatric UC, both in patients who did not respond to conventional therapies, and, more often, as initial therapy. Thanks to therapeutic drug monitoring and to the availability of biologics with different targets, therapy has become more targeted and personalized, with a significant improvement in response, in quality of life, and with a good safety profile.
Collapse
Affiliation(s)
- Debora Curci
- Institute for Maternal and Child Health, IRCCS "Burlo Garofolo", Trieste, Italy
| | - Marianna Lucafò
- Department of Life Sciences, University of Trieste, Trieste, Italy
| | - Giuliana Decorti
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Trieste, Italy
| | - Gabriele Stocco
- Institute for Maternal and Child Health, IRCCS "Burlo Garofolo", Trieste, Italy
| |
Collapse
|
63
|
Duran T, Karaselek MA, Kuccukturk S, Gul Y, Sahin A, Guner SN, Keles S, Reisli I. Investigation of Transcription Factor and Cytokine Gene Expression Levels in Helper T Cell Subsets Among Turkish Patients Diagnosed with ICF2 (Novel ZBTB24 gene Variant) and ICF3 (CDCA7 Variant) Syndrome. J Clin Immunol 2024; 45:16. [PMID: 39320531 DOI: 10.1007/s10875-024-01807-5] [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: 03/21/2024] [Accepted: 09/10/2024] [Indexed: 09/26/2024]
Abstract
Immunodeficiency, centromeric region instability, facial anomalies syndrome (ICF), is a rare disease with autosomal recessive inheritance. ICF syndrome. It has been reported that ICF syndrome is caused by mutations in the DNMT3B (ICF1), ZBTB24 (ICF2), CDCA7 (ICF3), and HELLS (ICF4) genes. As a result of literature research, there are no studies on transcription factor and cytokine expressions of helper T cell subsets in ICF syndrome. In the study; Th1 (TBET, STAT1, STAT4), Th2 (GATA3, STAT6), Th17 (RORgt, STAT3), Treg (FoxP3, STAT5) transcription factors and the major cytokines of these cells (Th1; IFNG, Th2; IL4, Th17; IL17A-21-22, Treg; IL10, TGFβ) expressions were aimed to be evaluated by qRT-PCR. Patients (ICF3: three patients; ICF2: two patients), six heterozygous individual and five healthy controls were included in the study. All patients had hypogammaglobulinemia. Except for the CD19 cells of P2 from patients diagnosed with ICF3, the CD3, CD4, CD8, and CD19 cells in the other ICF3 patients were normal. However, the rates of these cells were low in patients with ICF2 syndrome. Factors belonging to patients' Th1, Th17 and Treg cells were significantly lower than the control. Additionally, novel mutation was detected in ZBTB24 gene (c.1121-2 A > T). Our study is the first molecular study on Th cell subsets in patients with ICF syndrome and a new mutation that causes ICF2 syndrome has been identified.
Collapse
Affiliation(s)
- Tugce Duran
- Department of Medical Genetics, Medicine Faculty, KTO Karatay University, Konya, Turkey.
- Department of Pediatric Immunology and Allergy, Medicine Faculty, Necmettin Erbakan University, Konya, Turkey.
| | - Mehmet Ali Karaselek
- Department of Pediatric Immunology and Allergy, Medicine Faculty, Necmettin Erbakan University, Konya, Turkey
| | - Serkan Kuccukturk
- Department of Medical Biology, Medicine Faculty, Karamanoğlu Mehmetbey University, Karaman, Turkey
| | - Yahya Gul
- Department of Pediatric Immunology and Allergy, Diyarbakır Gazi Yasargil Education and Research Hospital, Diyarbakir, Turkey
| | - Ali Sahin
- School of Medicine Faculty, Selcuk University, Konya, Turkey
| | - Sukru Nail Guner
- Department of Pediatric Immunology and Allergy, Medicine Faculty, Necmettin Erbakan University, Konya, Turkey
| | - Sevgi Keles
- Department of Pediatric Immunology and Allergy, Medicine Faculty, Necmettin Erbakan University, Konya, Turkey
| | - Ismail Reisli
- Department of Pediatric Immunology and Allergy, Medicine Faculty, Necmettin Erbakan University, Konya, Turkey
| |
Collapse
|
64
|
Wang Y, Wang Y, Ge Y, Wu Z, Yue X, Li C, Liang X, Ma C, Wang P, Gao L. Tim-4 alleviates acute hepatic injury by modulating homeostasis and function of NKT cells. Clin Exp Immunol 2024; 218:101-110. [PMID: 39036980 PMCID: PMC11404119 DOI: 10.1093/cei/uxae063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 05/29/2024] [Accepted: 07/20/2024] [Indexed: 07/23/2024] Open
Abstract
T-cell immunoglobulin and mucin domain-containing molecule 4 (Tim-4) is an immune checkpoint molecule, which involves in numerous inflammatory diseases. Tim-4 is mainly expressed on antigen-presenting cells. However, increasing evidence has shown that Tim-4 is also expressed on natural killer T (NKT) cells. The role of Tim-4 in maintaining NKT cell homeostasis and function remains unknown. In this study, we explored the effect of Tim-4 on NKT cells in acute liver injury. This study found that Tim-4 expression on hepatic NKT cells was elevated during acute liver injury. Tim-4 deficiency enhanced IFN-γ, TNF-α expression while impaired IL-4 production in NKT cells. Loss of Tim-4 drove NKT-cell effector lineages to be skewed to NKT1 subset. Furthermore, Tim-4 KO mice were more susceptible to α-Galactosylceramide (α-GalCer) challenge. In conclusion, our findings indicate that Tim-4 plays an important role in regulating homeostasis and function of NKT cells in acute liver injury. Therefore, Tim-4 might become a new regulator of NKT cells and a potential target for the therapy of acute hepatitis.
Collapse
Affiliation(s)
- Yingchun Wang
- Key Laboratory for Experimental Teratology of Ministry of Education, Shandong Key Laboratory of Infection and Immunity, and Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, PR China
| | - Yuzhen Wang
- Key Laboratory for Experimental Teratology of Ministry of Education, Shandong Key Laboratory of Infection and Immunity, and Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, PR China
| | - Yutong Ge
- Key Laboratory for Experimental Teratology of Ministry of Education, Shandong Key Laboratory of Infection and Immunity, and Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, PR China
| | - Zhuanchang Wu
- Key Laboratory for Experimental Teratology of Ministry of Education, Shandong Key Laboratory of Infection and Immunity, and Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, PR China
| | - Xuetian Yue
- Department of Cell Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, PR China
| | - Chunyang Li
- Key Laboratory for Experimental Teratology of Ministry of Education and Department of Histology and Embryology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, PR China
| | - Xiaohong Liang
- Key Laboratory for Experimental Teratology of Ministry of Education, Shandong Key Laboratory of Infection and Immunity, and Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, PR China
| | - Chunhong Ma
- Key Laboratory for Experimental Teratology of Ministry of Education, Shandong Key Laboratory of Infection and Immunity, and Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, PR China
| | - Pin Wang
- Department of Neurology, The Second Hospital, Cheeloo College of Medicine, Shandong University. Jinan, Shandong 250033, PR China
| | - Lifen Gao
- Key Laboratory for Experimental Teratology of Ministry of Education, Shandong Key Laboratory of Infection and Immunity, and Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, PR China
| |
Collapse
|
65
|
Miranda S, Vermeesen R, Janssen A, Rehnberg E, Etlioglu E, Baatout S, Tabury K, Baselet B. Effects of simulated space conditions on CD4+ T cells: a multi modal analysis. Front Immunol 2024; 15:1443936. [PMID: 39286254 PMCID: PMC11402665 DOI: 10.3389/fimmu.2024.1443936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 08/08/2024] [Indexed: 09/19/2024] Open
Abstract
Introduction The immune system is an intricate network of cellular components that safeguards against pathogens and aberrant cells, with CD4+ T cells playing a central role in this process. Human space travel presents unique health challenges, such as heavy ion ionizing radiation, microgravity, and psychological stress, which can collectively impede immune function. The aim of this research was to examine the consequences of simulated space stressors on CD4+ T cell activation, cytokine production, and gene expression. Methods CD4+ T cells were obtained from healthy individuals and subjected to Fe ion particle radiation, Photon irradiation, simulated microgravity, and hydrocortisone, either individually or in different combinations. Cytokine levels for Th1 and Th2 cells were determined using multiplex Luminex assays, and RNA sequencing was used to investigate gene expression patterns and identify essential genes and pathways impacted by these stressors. Results Simulated microgravity exposure resulted in an apparent Th1 to Th2 shift, evidenced on the level of cytokine secretion as well as altered gene expression. RNA sequencing analysis showed that several gene pathways were altered, particularly in response to Fe ions irradiation and simulated microgravity exposures. Individually, each space stressor caused differential gene expression, while the combination of stressors revealed complex interactions. Discussion The research findings underscore the substantial influence of the space exposome on immune function, particularly in the regulation of T cell responses. Future work should focus expanding the limited knowledge in this field. Comprehending these modifications will be essential for devising effective strategies to safeguard the health of astronauts during extended space missions. Conclusion The effects of simulated space stressors on CD4+ T cell function are substantial, implying that space travel poses a potential threat to immune health. Additional research is necessary to investigate the intricate relationship between space stressors and to develop effective countermeasures to mitigate these consequences.
Collapse
Affiliation(s)
- Silvana Miranda
- Radiobiology Unit, Institute for Nuclear Medical Applications, Belgian Nuclear Research Centre SCK CEN, Mol, Belgium
- Department of Biotechnology, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
| | - Randy Vermeesen
- Radiobiology Unit, Institute for Nuclear Medical Applications, Belgian Nuclear Research Centre SCK CEN, Mol, Belgium
| | - Ann Janssen
- Radiobiology Unit, Institute for Nuclear Medical Applications, Belgian Nuclear Research Centre SCK CEN, Mol, Belgium
| | - Emil Rehnberg
- Radiobiology Unit, Institute for Nuclear Medical Applications, Belgian Nuclear Research Centre SCK CEN, Mol, Belgium
- Department of Biotechnology, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
| | - Emre Etlioglu
- Radiobiology Unit, Institute for Nuclear Medical Applications, Belgian Nuclear Research Centre SCK CEN, Mol, Belgium
| | - Sarah Baatout
- Radiobiology Unit, Institute for Nuclear Medical Applications, Belgian Nuclear Research Centre SCK CEN, Mol, Belgium
- Department of Biotechnology, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
| | - Kevin Tabury
- Radiobiology Unit, Institute for Nuclear Medical Applications, Belgian Nuclear Research Centre SCK CEN, Mol, Belgium
- Department of Biomedical Engineering, College of Engineering and Computing, University of South Carolina, Columbia, SC, United States
| | - Bjorn Baselet
- Radiobiology Unit, Institute for Nuclear Medical Applications, Belgian Nuclear Research Centre SCK CEN, Mol, Belgium
| |
Collapse
|
66
|
Zhang H, Ren Y, Wang F, Tu X, Tong Z, Liu L, Zheng Y, Zhao P, Cheng J, Li J, Fang W, Liu X. The long-term effectiveness and mechanism of oncolytic virotherapy combined with anti-PD-L1 antibody in colorectal cancer patient. Cancer Gene Ther 2024; 31:1412-1426. [PMID: 39068234 PMCID: PMC11405277 DOI: 10.1038/s41417-024-00807-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 07/02/2024] [Accepted: 07/05/2024] [Indexed: 07/30/2024]
Abstract
Colorectal cancer (CRC) is known to be resistant to immunotherapy. In our phase-I clinical trial, one patient achieved a 313-day prolonged response during the combined treatment of oncolytic virotherapy and immunotherapy. To gain a deeper understanding of the potential molecular mechanisms, we performed a comprehensive multi-omics analysis on this patient and three non-responders. Our investigation unveiled that, initially, the tumor microenvironment (TME) of this responder presented minimal infiltration of T cells and natural killer cells, along with a relatively higher presence of macrophages compared to non-responders. Remarkably, during treatment, there was a progressive increase in CD4+ T cells, CD8+ T cells, and B cells in the responder's tumor tissue. This was accompanied by a significant upregulation of transcription factors associated with T-cell activation and cytotoxicity, including GATA3, EOMES, and RUNX3. Furthermore, dynamic monitoring of peripheral blood samples from the responder revealed a rapid decrease in circulating tumor DNA (ctDNA), suggesting its potential as an early blood biomarker of treatment efficacy. Collectively, our findings demonstrate the effectiveness of combined oncolytic virotherapy and immunotherapy in certain CRC patients and provide molecular evidence that virotherapy can potentially transform a "cold" TME into a "hot" one, thereby improving sensitivity to immunotherapy.
Collapse
Affiliation(s)
- Hangyu Zhang
- Department of Medical Oncology, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, P. R. China
| | - Yiqing Ren
- Department of Medical Oncology, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, P. R. China
| | - Feiyu Wang
- College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, P. R. China
| | - Xiaoxuan Tu
- Department of Medical Oncology, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, P. R. China
| | - Zhou Tong
- Department of Medical Oncology, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, P. R. China
| | - Lulu Liu
- Department of Medical Oncology, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, P. R. China
| | - Yi Zheng
- Department of Medical Oncology, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, P. R. China
| | - Peng Zhao
- Department of Medical Oncology, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, P. R. China
| | - Jinlin Cheng
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, P. R. China
| | - Jianwen Li
- Geneplus-Shenzhen, Shenzhen, P. R. China.
| | - Weijia Fang
- Department of Medical Oncology, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, P. R. China.
| | - Xia Liu
- ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou, Zhejiang, P. R. China.
| |
Collapse
|
67
|
Bączek K, Piotrowski WJ. Lung fibrosis in sarcoidosis. Is there a place for antifibrotics? Front Pharmacol 2024; 15:1445923. [PMID: 39281278 PMCID: PMC11392764 DOI: 10.3389/fphar.2024.1445923] [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: 06/08/2024] [Accepted: 08/22/2024] [Indexed: 09/18/2024] Open
Abstract
Sarcoidosis, an enigmatic disease with unknown etiology, is characterized by inflammation and the potential involvement of various organs, predominantly the lungs and intrathoracic lymph nodes. Non-caseating granulomas can resolve spontaneously in approximately 60% of cases within 2-3 years. However, sarcoidosis-related mortality has increased. Lung fibrosis, affecting up to 20% of sarcoidosis patients, stands out as a primary cause of mortality. Traditionally, fibrosis is viewed because of prolonged inflammation, necessitating anti-inflammatory treatment with systemic steroids, immunosuppressants, and anti-TNF agents to manage the disease. The recent introduction of antifibrotic drugs such as nintedanib and pirfenidone offers new avenues for treating fibrotic sarcoidosis. Nintedanib, effective in idiopathic pulmonary fibrosis (IPF) and systemic sclerosis-related interstitial lung disease (SSc-ILD), has shown promise in patients with various progressive fibrosing interstitial lung diseases (PF-ILD), including those with sarcoidosis. Pirfenidone, also effective in IPF, has demonstrated potential in managing fibrotic sarcoidosis, though results have been inconclusive due to limited participant numbers in studies. This review explores the theoretical and empirical evidence supporting the use of antifibrotics in sarcoidosis, weighing the benefits and drawbacks. While antifibrotics offer a potential therapeutic approach, further randomized controlled trials are essential to determine their efficacy in fibrotic sarcoidosis. Addressing fibrosis as a continuum of chronic inflammation, the role of antifibrotics in managing sarcoidosis remains an area requiring more in-depth research to improve patient outcomes and advance treatment paradigms.
Collapse
Affiliation(s)
- Karol Bączek
- Department of Pneumology, Medical University of Łódź, Łódź, Poland
| | | |
Collapse
|
68
|
Charoensuksira S, Tantiwong S, Pongklaokam J, Hanvivattanakul S, Surinlert P, Krajarng A, Thanasarnaksorn W, Hongeng S, Ponnikorn S. Disturbance of Immune Microenvironment in Androgenetic Alopecia through Spatial Transcriptomics. Int J Mol Sci 2024; 25:9031. [PMID: 39201715 PMCID: PMC11354591 DOI: 10.3390/ijms25169031] [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: 06/03/2024] [Revised: 08/11/2024] [Accepted: 08/15/2024] [Indexed: 09/03/2024] Open
Abstract
Androgenetic alopecia (AGA) is characterized by microinflammation and abnormal immune responses, particularly in the upper segment of hair follicles (HFs). However, the precise patterns of immune dysregulation remain unclear, partly due to limitations in current analysis techniques to preserve tissue architecture. The infundibulum, a major part of the upper segment of HFs, is associated with significant clusters of immune cells. In this study, we investigated immune cells around the infundibulum, referred to as peri-infundibular immune infiltration (PII). We employed spatial transcriptome profiling, a high-throughput analysis technology, to investigate the immunological disruptions within the PII region. Our comprehensive analysis included an evaluation of overall immune infiltrates, gene set enrichment analysis (GSEA), cellular deconvolution, differential expression analysis, over-representation analysis, protein-protein interaction (PPI) networks, and upstream regulator analysis to identify cell types and molecular dysregulation in immune cells. Our results demonstrated significant differences in immune signatures between the PII of AGA patients (PII-A) and the PII of control donors (PII-C). Specifically, PII-A exhibited an enrichment of CD4+ helper T cells, distinct immune response patterns, and a bias toward a T helper (Th) 2 response. Immunohistochemistry revealed disruptions in T cell subpopulations, with more CD4+ T cells displaying an elevated Th2 response and a reduced Th1-cytotoxic response compared to PII-C. These findings reveal the unique immune landscapes of PII-A and PII-C, suggesting potential for the development of innovative treatment approaches.
Collapse
Affiliation(s)
- Sasin Charoensuksira
- Division of Dermatology, Chulabhorn International College of Medicine, Thammasat University, Pathum Thani 12120, Thailand; (S.C.); (S.T.); (J.P.); (W.T.)
| | - Supasit Tantiwong
- Division of Dermatology, Chulabhorn International College of Medicine, Thammasat University, Pathum Thani 12120, Thailand; (S.C.); (S.T.); (J.P.); (W.T.)
| | - Juthapa Pongklaokam
- Division of Dermatology, Chulabhorn International College of Medicine, Thammasat University, Pathum Thani 12120, Thailand; (S.C.); (S.T.); (J.P.); (W.T.)
| | - Sirashat Hanvivattanakul
- Chulabhorn International College of Medicine, Thammasat University, Pathum Thani 12120, Thailand; (S.H.); (P.S.); (A.K.)
| | - Piyaporn Surinlert
- Chulabhorn International College of Medicine, Thammasat University, Pathum Thani 12120, Thailand; (S.H.); (P.S.); (A.K.)
- Research Unit in Synthesis and Applications of Graphene, Thammasat University, Pathum Thani 12120, Thailand
| | - Aungkana Krajarng
- Chulabhorn International College of Medicine, Thammasat University, Pathum Thani 12120, Thailand; (S.H.); (P.S.); (A.K.)
| | - Wilai Thanasarnaksorn
- Division of Dermatology, Chulabhorn International College of Medicine, Thammasat University, Pathum Thani 12120, Thailand; (S.C.); (S.T.); (J.P.); (W.T.)
- Division of Dermatology, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand
| | - Suradej Hongeng
- Division of Hematology and Oncology, Department of Pediatrics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand;
| | - Saranyoo Ponnikorn
- Division of Dermatology, Chulabhorn International College of Medicine, Thammasat University, Pathum Thani 12120, Thailand; (S.C.); (S.T.); (J.P.); (W.T.)
- Chulabhorn International College of Medicine, Thammasat University, Pathum Thani 12120, Thailand; (S.H.); (P.S.); (A.K.)
- Thammasat University, Pattaya Campus, Bang Lamung 20150, Thailand
| |
Collapse
|
69
|
Muratsu A, Oda S, Onishi S, Yoshimura J, Matsumoto H, Togami Y, Mitsuyama Y, Ito H, Okuzaki D, Ogura H, Oda J. Bacterial sepsis causes more dramatic pathogenetic changes in the Th1 pathway than does viral (COVID-19) sepsis: a prospective observational study of whole blood transcriptomes. Virol J 2024; 21:190. [PMID: 39160575 PMCID: PMC11334310 DOI: 10.1186/s12985-024-02451-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 07/30/2024] [Indexed: 08/21/2024] Open
Abstract
OBJECTIVES This study aimed to comprehensively compare host responses of patients with bacterial sepsis and those with viral (COVID-19) sepsis by analyzing messenger RNA (mRNA) and microRNA (miRNA) profiles to shed light on their distinct pathophysiological mechanisms. DESIGN Prospective observational study. SETTING Whole blood RNA sequencing was used to analyze mRNA and miRNA profiles of patients diagnosed as having bacterial sepsis or viral (COVID-19) sepsis at the Department of Trauma and Emergency Medicine, Osaka University Graduate School of Medicine. PATIENTS Twenty-two bacterial sepsis patients, 35 viral (COVID-19) sepsis patients, and 15 healthy subjects admitted to the department were included. We diagnosed bacterial sepsis patients according to the sepsis-3 criterion that the Sequential Organ Failure Assessment score must increase to 2 points or more among patients with suspected infections. Viral (COVID-19) sepsis patients were diagnosed using SARS-CoV-2 RT-PCR testing, and presence of pneumonia was assessed through chest computed tomography scans. INTERVENTIONS None. MEASUREMENTS AND MAIN RESULTS For RNA sequencing, 14,500 mRNAs, 1121 miRNAs, and 2556 miRNA-targeted mRNAs were available for analysis in the bacterial sepsis patients. Numbers of genes showing upregulated: downregulated gene expression (false discovery rate < 0.05, |log2 fold change| > 1.5) were 256:2887 for mRNA, 53:5 for miRNA, and 49:2507 for miRNA-targeted mRNA. Similarly, in viral (COVID-19) sepsis patients, 14,500 mRNAs, 1121 miRNAs, and 327 miRNA-targeted mRNAs were analyzed, with numbers of genes exhibiting upregulated: downregulated gene expression of 672:1147 for mRNA, 3:4 for miRNA, and 165:162 for miRNA-targeted mRNA. This analysis revealed significant differences in the numbers of upregulated and downregulated genes expressed and pathways between the bacterial sepsis and viral (COVID-19) sepsis patients. Bacterial sepsis patients showed activation of the PD-1 and PD-L1 cancer immunotherapy signaling pathway and concurrent suppression of Th1 signaling. CONCLUSION Our study illuminated distinct molecular variances between bacterial sepsis and viral (COVID-19) sepsis. Bacterial sepsis patients had a greater number of upregulated and downregulated genes and pathways compared to viral (COVID-19) sepsis patients. Especially, bacterial sepsis caused more dramatic pathogenetic changes in the Th1 pathway than did viral (COVID-19) sepsis.
Collapse
Affiliation(s)
- Arisa Muratsu
- Department of Traumatology and Acute Critical Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Sayaka Oda
- Department of Oral and Maxillofacial Surgery, Osaka University Graduate School of Dentistry, Osaka, Japan
- Laboratory for Human Immunology (Single Cell Genomics), WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Shinya Onishi
- Department of Traumatology and Acute Critical Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Jumpei Yoshimura
- Department of Traumatology and Acute Critical Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Hisatake Matsumoto
- Department of Traumatology and Acute Critical Medicine, Osaka University Graduate School of Medicine, Osaka, Japan.
| | - Yuki Togami
- Department of Traumatology and Acute Critical Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yumi Mitsuyama
- Department of Traumatology and Acute Critical Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Hiroshi Ito
- Department of Traumatology and Acute Critical Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Daisuke Okuzaki
- Laboratory for Human Immunology (Single Cell Genomics), WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Hiroshi Ogura
- Department of Traumatology and Acute Critical Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Jun Oda
- Department of Traumatology and Acute Critical Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| |
Collapse
|
70
|
Sun X, Gu R, Bai J. Differentiation and regulation of CD4 + T cell subsets in Parkinson's disease. Cell Mol Life Sci 2024; 81:352. [PMID: 39153043 PMCID: PMC11335276 DOI: 10.1007/s00018-024-05402-0] [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: 03/09/2024] [Revised: 08/06/2024] [Accepted: 08/07/2024] [Indexed: 08/19/2024]
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disease, and its hallmark pathological features are the loss of dopaminergic (DA) neurons in the midbrain substantia nigra pars compacta (SNpc) and the accumulation of alpha-synuclein (α-syn). It has been shown that the integrity of the blood-brain barrier (BBB) is damaged in PD patients, and a large number of infiltrating T cells and inflammatory cytokines have been detected in the cerebrospinal fluid (CSF) and brain parenchyma of PD patients and PD animal models, including significant change in the number and proportion of different CD4+ T cell subsets. This suggests that the neuroinflammatory response caused by CD4+ T cells is an important risk factor for the development of PD. Here, we systematically review the differentiation of CD4+ T cell subsets, and focus on describing the functions and mechanisms of different CD4+ T cell subsets and their secreted cytokines in PD. We also summarize the current immunotherapy targeting CD4+ T cells with a view to providing assistance in the diagnosis and treatment of PD.
Collapse
Affiliation(s)
- Xiaowei Sun
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, 650500, China
- Medical School, Kunming University of Science and Technology, Kunming, 650500, China
- Southwest United Graduate School, Kunming, 650500, China
| | - Rou Gu
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, 650500, China
- Medical School, Kunming University of Science and Technology, Kunming, 650500, China
| | - Jie Bai
- Medical School, Kunming University of Science and Technology, Kunming, 650500, China.
- Southwest United Graduate School, Kunming, 650500, China.
| |
Collapse
|
71
|
Ansari AW, Jayakumar MN, Ahmad F, Venkatachalam T, Salameh L, Unnikannan H, Raheed T, Mohammed AK, Mahboub B, Al-Ramadi BK, Hamid Q, Steinhoff M, Hamoudi R. Azithromycin targets the CD27 pathway to modulate CD27hi T-lymphocyte expansion and type-1 effector phenotype. Front Immunol 2024; 15:1447625. [PMID: 39211048 PMCID: PMC11357905 DOI: 10.3389/fimmu.2024.1447625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 07/12/2024] [Indexed: 09/04/2024] Open
Abstract
Macrolide antibiotic azithromycin is widely used in clinical practice to treat respiratory tract infections and inflammatory diseases. However, its mechanism of action is not fully understood. Given the involvement of the CD27 pathway in the pathophysiology of various T-lymphocyte-mediated inflammatory, autoimmune, and lymphoproliferative diseases, we examined the impact of AZM on CD27 regulation and potential consequences on CD4+ and CD8+ T-cell phenotypes. Using cellular immunology approaches on healthy donors' peripheral blood mononuclear cells, we demonstrate AZM-mediated downregulation of surface CD27 expression as well as its extracellular release as soluble CD27. Notably, AZM-exposed CD27high (hi) cells were defective in their ability to expand compared to CD27intermediate (Int) and CD27low (lo) subsets. The defective CD27hi subset expansion was found to be associated with impaired cell proliferation and cell division. At the molecular level, the CD27hi subset exhibited lower mTOR activity than other subsets. Functionally, AZM treatment resulted in marked depletion of helper CD4+ (Th1) and cytotoxic CD8+ T-lymphocyte (Tc1)-associated CXCR3+CD27hi effector cells and inhibition of inflammatory cytokine IFN-γ production. These findings provide mechanistic insights on immunomodulatory features of AZM on T-lymphocyte by altering the CD27 pathway. From a clinical perspective, this study also sheds light on potential clinical benefits observed in patients on prophylactic AZM regimens against various respiratory diseases and opens avenues for future adjunct therapy against Th1- and Tc1-dominated inflammatory and autoimmune diseases.
Collapse
Affiliation(s)
- Abdul Wahid Ansari
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
- Dermatology Institute, Interim Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Manju Nidagodu Jayakumar
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Fareed Ahmad
- Dermatology Institute, Interim Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Thenmozhi Venkatachalam
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Laila Salameh
- Department of Pulmonary Medicine, Rashid Hospital, Dubai, United Arab Emirates
| | - Hema Unnikannan
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Thesni Raheed
- Dermatology Institute, Interim Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Abdul Khader Mohammed
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Bassam Mahboub
- Department of Pulmonary Medicine, Rashid Hospital, Dubai, United Arab Emirates
| | - Basel K. Al-Ramadi
- Department of Medical Microbiology and Immunology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
- Zayed Center for Health Sciences, United Arab Emirates University (UAEU), Al Ain, United Arab Emirates
- ASPIRE Precision Medicine Research Institute Abu Dhabi, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Qutayba Hamid
- Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
- Meakins-Christie Laboratories, Faculty of Medicine, McGill University, Montreal, QC, Canada
| | - Martin Steinhoff
- Dermatology Institute, Interim Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
- Department of Dermatology and Venereology, Hamad Medical Corporation, Doha, Qatar
- Weill Cornell Medicine-Qatar, Doha, Qatar
- College of Medicine, Qatar University, Doha, Qatar
- College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar
| | - Rifat Hamoudi
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
- Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
- Division of Surgery and Interventional Science, University College London, London, United Kingdom
- Biomedically Informed Artificial Intelligence Laboratory (BIMAI-Lab), University of Sharjah, Sharjah, United Arab Emirates
- ASPIRE Precision Medicine Research Institute Abu Dhabi, University of Sharjah, Sharjah, United Arab Emirates
| |
Collapse
|
72
|
Ibraheem Y, Bayarsaikhan G, Macalinao ML, Kimura K, Yui K, Aoshi T, Inoue SI. γδ T cell-mediated activation of cDC1 orchestrates CD4 + Th1 cell priming in malaria. Front Immunol 2024; 15:1426316. [PMID: 39211036 PMCID: PMC11357926 DOI: 10.3389/fimmu.2024.1426316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 07/17/2024] [Indexed: 09/04/2024] Open
Abstract
γδ T cells facilitate the CD4+ T helper 1 (Th1) cell response against Plasmodium infection by activating conventional dendritic cells (cDCs), although the underlying mechanism remains elusive. Our study revealed that γδ T cells promote the complete maturation and production of interleukin-12 and CXCR3-ligands specifically in type 1 cDCs (cDC1), with minimal impact on cDC2 and monocyte derived DCs (Mo-DCs). During the initial infection phase, γδ T cell activation and temporal accumulation in the splenic white pulp, alongside cDC1, occur via CCR7-signaling. Furthermore, cDC1/γδ T cell interactions in the white pulp are amplified through CXCR3 signaling in γδ T cells, optimizing Th1 cell priming by cDC1. We also demonstrated how transitional Th1 cells arise in the white pulp before establishing their presence in the red pulp as fully differentiated Th1 cells. Additionally, we elucidate the reciprocal activation between γδ T cells and cDC1s. These findings suggest that Th1 cell priming is orchestrated by this reciprocal activation in the splenic white pulp during the early phase of blood-stage Plasmodium infection.
Collapse
MESH Headings
- Th1 Cells/immunology
- Dendritic Cells/immunology
- Dendritic Cells/metabolism
- Animals
- Mice
- Lymphocyte Activation/immunology
- Malaria/immunology
- Malaria/parasitology
- Mice, Inbred C57BL
- Receptors, CXCR3/metabolism
- Receptors, Antigen, T-Cell, gamma-delta/metabolism
- Receptors, Antigen, T-Cell, gamma-delta/immunology
- Receptors, CCR7/metabolism
- Receptors, CCR7/immunology
- Signal Transduction
- Spleen/immunology
- Cell Differentiation/immunology
- Female
Collapse
Affiliation(s)
- Yarob Ibraheem
- Department of Immunology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Ganchimeg Bayarsaikhan
- Department of Immunology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | | | - Kazumi Kimura
- Department of Immunology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Katsuyuki Yui
- Department of Immunology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
- School of Tropical Medicine and Global Health, Nagasaki University, Nagasaki, Japan
- Shionogi Global Infectious Diseases Division, Institute of Tropical Medicine, Nagasaki University, Nagasaki, Japan
| | - Taiki Aoshi
- Department of Immunology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Shin-Ichi Inoue
- Department of Immunology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| |
Collapse
|
73
|
Bonetti L, Horkova V, Grusdat M, Longworth J, Guerra L, Kurniawan H, Franchina DG, Soriano-Baguet L, Binsfeld C, Verschueren C, Spath S, Ewen A, Koncina E, Gérardy JJ, Kobayashi T, Dostert C, Farinelle S, Härm J, Fan YT, Chen Y, Harris IS, Lang PA, Vasiliou V, Waisman A, Letellier E, Becher B, Mittelbronn M, Brenner D. A Th17 cell-intrinsic glutathione/mitochondrial-IL-22 axis protects against intestinal inflammation. Cell Metab 2024; 36:1726-1744.e10. [PMID: 38986617 DOI: 10.1016/j.cmet.2024.06.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 02/06/2024] [Accepted: 06/12/2024] [Indexed: 07/12/2024]
Abstract
The intestinal tract generates significant reactive oxygen species (ROS), but the role of T cell antioxidant mechanisms in maintaining intestinal homeostasis is poorly understood. We used T cell-specific ablation of the catalytic subunit of glutamate cysteine ligase (Gclc), which impaired glutathione (GSH) production, crucially reducing IL-22 production by Th17 cells in the lamina propria, which is critical for gut protection. Under steady-state conditions, Gclc deficiency did not alter cytokine secretion; however, C. rodentium infection induced increased ROS and disrupted mitochondrial function and TFAM-driven mitochondrial gene expression, resulting in decreased cellular ATP. These changes impaired the PI3K/AKT/mTOR pathway, reducing phosphorylation of 4E-BP1 and consequently limiting IL-22 translation. The resultant low IL-22 levels led to poor bacterial clearance, severe intestinal damage, and high mortality. Our findings highlight a previously unrecognized, essential role of Th17 cell-intrinsic GSH in promoting mitochondrial function and cellular signaling for IL-22 protein synthesis, which is critical for intestinal integrity and defense against gastrointestinal infections.
Collapse
Affiliation(s)
- Lynn Bonetti
- Experimental and Molecular Immunology, Department of Infection and Immunity (DII), Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg; Immunology & Genetics, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7, Avenue des Hauts Fourneaux, Esch-sur-Alzette, Luxembourg
| | - Veronika Horkova
- Experimental and Molecular Immunology, Department of Infection and Immunity (DII), Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg; Immunology & Genetics, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7, Avenue des Hauts Fourneaux, Esch-sur-Alzette, Luxembourg
| | - Melanie Grusdat
- Experimental and Molecular Immunology, Department of Infection and Immunity (DII), Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg; Immunology & Genetics, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7, Avenue des Hauts Fourneaux, Esch-sur-Alzette, Luxembourg
| | - Joseph Longworth
- Experimental and Molecular Immunology, Department of Infection and Immunity (DII), Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg; Immunology & Genetics, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7, Avenue des Hauts Fourneaux, Esch-sur-Alzette, Luxembourg
| | - Luana Guerra
- Experimental and Molecular Immunology, Department of Infection and Immunity (DII), Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg; Immunology & Genetics, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7, Avenue des Hauts Fourneaux, Esch-sur-Alzette, Luxembourg
| | - Henry Kurniawan
- Experimental and Molecular Immunology, Department of Infection and Immunity (DII), Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg; Immunology & Genetics, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7, Avenue des Hauts Fourneaux, Esch-sur-Alzette, Luxembourg
| | - Davide G Franchina
- Experimental and Molecular Immunology, Department of Infection and Immunity (DII), Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg; Immunology & Genetics, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7, Avenue des Hauts Fourneaux, Esch-sur-Alzette, Luxembourg
| | - Leticia Soriano-Baguet
- Experimental and Molecular Immunology, Department of Infection and Immunity (DII), Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg; Immunology & Genetics, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7, Avenue des Hauts Fourneaux, Esch-sur-Alzette, Luxembourg
| | - Carole Binsfeld
- Experimental and Molecular Immunology, Department of Infection and Immunity (DII), Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg; Immunology & Genetics, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7, Avenue des Hauts Fourneaux, Esch-sur-Alzette, Luxembourg
| | - Charlène Verschueren
- Experimental and Molecular Immunology, Department of Infection and Immunity (DII), Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg; Immunology & Genetics, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7, Avenue des Hauts Fourneaux, Esch-sur-Alzette, Luxembourg
| | - Sabine Spath
- Institute of Experimental Immunology, Inflammation Research, University of Zurich, 8057 Zurich, Switzerland; Center for Fundamental Immunology, Benaroya Research Institute, Seattle, WA 98101, USA
| | - Anouk Ewen
- Experimental and Molecular Immunology, Department of Infection and Immunity (DII), Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg; Immunology & Genetics, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7, Avenue des Hauts Fourneaux, Esch-sur-Alzette, Luxembourg
| | - Eric Koncina
- Molecular Disease Mechanisms Group, Department of Life Sciences and Medicine, University of Luxembourg, Belval, Luxembourg
| | - Jean-Jacques Gérardy
- National Center of Pathology (NCP), Laboratoire National de Santé (LNS), Dudelange, Luxembourg; Luxembourg Center of Neuropathology (LCNP), 3555 Dudelange, Luxembourg
| | - Takumi Kobayashi
- Experimental and Molecular Immunology, Department of Infection and Immunity (DII), Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg; Immunology & Genetics, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7, Avenue des Hauts Fourneaux, Esch-sur-Alzette, Luxembourg
| | - Catherine Dostert
- Experimental and Molecular Immunology, Department of Infection and Immunity (DII), Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg; Immunology & Genetics, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7, Avenue des Hauts Fourneaux, Esch-sur-Alzette, Luxembourg
| | - Sophie Farinelle
- Experimental and Molecular Immunology, Department of Infection and Immunity (DII), Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg; Immunology & Genetics, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7, Avenue des Hauts Fourneaux, Esch-sur-Alzette, Luxembourg
| | - Janika Härm
- Experimental and Molecular Immunology, Department of Infection and Immunity (DII), Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg; Immunology & Genetics, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7, Avenue des Hauts Fourneaux, Esch-sur-Alzette, Luxembourg
| | - Yu-Tong Fan
- Experimental and Molecular Immunology, Department of Infection and Immunity (DII), Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg; Immunology & Genetics, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7, Avenue des Hauts Fourneaux, Esch-sur-Alzette, Luxembourg
| | - Ying Chen
- Department of Environmental Health Sciences, Yale School of Public Health, New Haven, CT, USA
| | - Isaac S Harris
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Philipp A Lang
- Department of Molecular Medicine II, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Vasilis Vasiliou
- Department of Environmental Health Sciences, Yale School of Public Health, New Haven, CT, USA
| | - Ari Waisman
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Elisabeth Letellier
- Molecular Disease Mechanisms Group, Department of Life Sciences and Medicine, University of Luxembourg, Belval, Luxembourg
| | - Burkhard Becher
- Institute of Experimental Immunology, Inflammation Research, University of Zurich, 8057 Zurich, Switzerland
| | - Michel Mittelbronn
- National Center of Pathology (NCP), Laboratoire National de Santé (LNS), Dudelange, Luxembourg; Luxembourg Center of Neuropathology (LCNP), 3555 Dudelange, Luxembourg; Department of Life Sciences and Medicine (DLSM), University of Luxembourg, Esch-sur-Alzette, Luxembourg; Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 4362 Esch-sur-Alzette, Luxembourg; Faculty of Science, Technology and Medicine (FSTM), University of Luxembourg, Esch-sur-Alzette, Luxembourg; Department of Cancer Research (DoCR), Luxembourg Institute of Health (LIH), 1526 Luxembourg, Luxembourg
| | - Dirk Brenner
- Experimental and Molecular Immunology, Department of Infection and Immunity (DII), Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg; Immunology & Genetics, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7, Avenue des Hauts Fourneaux, Esch-sur-Alzette, Luxembourg; Odense Research Center for Anaphylaxis (ORCA), Department of Dermatology and Allergy Center, Odense University Hospital, University of Southern Denmark, Odense, Denmark.
| |
Collapse
|
74
|
Mohammad Taheri M, Javan F, Poudineh M, Athari SS. Beyond CAR-T: The rise of CAR-NK cell therapy in asthma immunotherapy. J Transl Med 2024; 22:736. [PMID: 39103889 PMCID: PMC11302387 DOI: 10.1186/s12967-024-05534-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Accepted: 07/23/2024] [Indexed: 08/07/2024] Open
Abstract
Asthma poses a major public health burden. While existing asthma drugs manage symptoms for many, some patients remain resistant. The lack of a cure, especially for severe asthma, compels exploration of novel therapies. Cancer immunotherapy successes with CAR-T cells suggest its potential for asthma treatment. Researchers are exploring various approaches for allergic diseases including membrane-bound IgE, IL-5, PD-L2, and CTLA-4 for asthma, and Dectin-1 for fungal asthma. NK cells offer several advantages over T cells for CAR-based immunotherapy. They offer key benefits: (1) HLA compatibility, meaning they can be used in a wider range of patients without the need for matching tissue types. (2) Minimal side effects (CRS and GVHD) due to their limited persistence and cytokine profile. (3) Scalability for "off-the-shelf" production from various sources. Several strategies have been introduced that highlight the superiority and challenges of CAR-NK cell therapy for asthma treatment including IL-10, IFN-γ, ADCC, perforin-granzyme, FASL, KIR, NCRs (NKP46), DAP, DNAM-1, TGF-β, TNF-α, CCL, NKG2A, TF, and EGFR. Furthermore, we advocate for incorporating AI for CAR design optimization and CRISPR-Cas9 gene editing technology for precise gene manipulation to generate highly effective CAR constructs. This review will delve into the evolution and production of CAR designs, explore pre-clinical and clinical studies of CAR-based therapies in asthma, analyze strategies to optimize CAR-NK cell function, conduct a comparative analysis of CAR-T and CAR-NK cell therapy with their respective challenges, and finally present established novel CAR designs with promising potential for asthma treatment.
Collapse
Affiliation(s)
| | - Fatemeh Javan
- Student Research Committee, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Mohadeseh Poudineh
- Student Research Committee, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Seyed Shamseddin Athari
- Cancer Gene Therapy Research Center, Zanjan University of Medical Sciences, Zanjan, Iran.
- Department of Immunology, Zanjan School of Medicine, Zanjan University of Medical Sciences, 12th Street, Shahrake Karmandan, Zanjan, 45139-561111, Iran.
| |
Collapse
|
75
|
McGrath S, Grimstad K, Thorarinsdottir K, Forslind K, Glinatsi D, Leu Agelii M, Aranburu A, Sundell T, Jonsson CA, Camponeschi A, Hultgård Ekwall AK, Tilevik A, Gjertsson I, Mårtensson IL. Correlation of Professional Antigen-Presenting Tbet +CD11c + B Cells With Bone Destruction in Untreated Rheumatoid Arthritis. Arthritis Rheumatol 2024; 76:1263-1277. [PMID: 38570939 DOI: 10.1002/art.42857] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 02/09/2024] [Accepted: 04/01/2024] [Indexed: 04/05/2024]
Abstract
OBJECTIVE Subsets of CD21-/low memory B cells (MBCs), including double-negative (DN, CD27-IgD-) and Tbet+CD11c+ cells, are expanded in chronic inflammatory diseases. In rheumatoid arthritis (RA), CD21-/low MBCs correlate with joint destruction. However, whether this is due to the Tbet+CD11c+ subset, its function and pathogenic contribution to RA are unknown. This study aims to investigate the association between CD21-/lowTbet+CD11c+ MBCs and joint destruction as well as other clinical parameters and to elucidate their functional properties in patients with untreated RA (uRA). METHODS Clinical observations were combined with flow cytometry (n = 36) and single-cell RNA sequencing (scRNA-seq) and V(D)J sequencing (n = 4) of peripheral blood (PB) MBCs from patients with uRA. The transcriptome of circulating Tbet+CD11c+ MBCs was compared with scRNA-seq data of synovial B cells. In vitro coculture of Tbet+CD11c+ B cells with T cells was used to assess costimulatory capacity. RESULTS CD21-/lowTbet+CD11c+ MBCs in PB correlated with bone destruction but no other clinical parameters analyzed. The Tbet+CD11c+ MBCs have undergone clonal expansion and express somatically mutated V genes. Gene expression analysis of these cells identified a unique signature of more than 150 up-regulated genes associated with antigen presentation functions, including B cell receptor activation and clathrin-mediated antigen internalization; regulation of actin filaments, endosomes, and lysosomes; antigen processing, loading, presentation, and costimulation; a transcriptome mirrored in their synovial tissue counterparts. In vitro, Tbet+CD11c+ B cells induced retinoic acid receptor-related orphan nuclear receptor γT expression in CD4+ T cells, thereby polarizing to Th17 cells, a T cell subset critical for osteoclastogenesis and associated with bone destruction. CONCLUSION This study suggests that Tbet+CD11c+ MBCs contribute to the pathogenesis of RA by promoting bone destruction through antigen presentation, T cell activation, and Th17 polarization.
Collapse
Affiliation(s)
- Sarah McGrath
- Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Kristoffer Grimstad
- Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden, and School of Bioscience, University of Skövde, Skövde, Sweden
| | - Katrin Thorarinsdottir
- Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Kristina Forslind
- Lund University, Lund, Sweden, and Spenshult Research and Development Centre, Halmstad, Sweden
| | | | - Monica Leu Agelii
- Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Alaitz Aranburu
- Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Timothy Sundell
- Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Charlotte A Jonsson
- Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Alessandro Camponeschi
- Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Anna-Karin Hultgård Ekwall
- Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden, and Sahlgrenska University Hospital, Gothenburg, Sweden
| | | | - Inger Gjertsson
- Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden, and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Inga-Lill Mårtensson
- Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
76
|
Moss P. A tumor suppressor of CLL: all (T-)bets are on. Blood 2024; 144:467-469. [PMID: 39088232 DOI: 10.1182/blood.2024025015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/02/2024] Open
Affiliation(s)
- Paul Moss
- University of Birmingham
- University Hospitals Birmingham NHS Foundation Trust
| |
Collapse
|
77
|
Nellore A, Zumaquero E, Seifert M. T-bet + B Cells in Humans: Protective and Pathologic Functions. Transplantation 2024; 108:1709-1714. [PMID: 38051131 PMCID: PMC11150333 DOI: 10.1097/tp.0000000000004889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 10/28/2023] [Accepted: 10/31/2023] [Indexed: 12/07/2023]
Abstract
The humoral immune system comprises B cells and plasma cells, which play important roles in organ transplantation, ranging from the production of both protective and injurious antibodies as well as cytokines that can promote operational tolerance. Recent data from conditions outside of transplantation have identified a novel human B-cell subset that expresses the transcription factor T-bet and exerts pleiotropic functions by disease state. Here, we review the generation, activation, and functions of the T-bet + B-cell subset outside of allotransplantation, and consider the relevance of this subset as mediators of allograft injury.
Collapse
Affiliation(s)
- Anoma Nellore
- Division of Infectious Diseases, University of Alabama at Birmingham, Birmingham, AL
| | - Esther Zumaquero
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL
| | - Michael Seifert
- Division of Pediatric Nephrology, University of Alabama at Birmingham, Birmingham, AL
| |
Collapse
|
78
|
Zou H, Liu C, Ruan Y, Fang L, Wu T, Han S, Dang T, Meng H, Zhang Y. Colorectal medullary carcinoma: a pathological subtype with intense immune response and potential to benefit from immune checkpoint inhibitors. Expert Rev Clin Immunol 2024; 20:997-1008. [PMID: 38459764 DOI: 10.1080/1744666x.2024.2328746] [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: 12/03/2023] [Accepted: 03/06/2024] [Indexed: 03/10/2024]
Abstract
INTRODUCTION Different pathological types of colorectal cancer have distinguished immune landscape, and the efficacy of immunotherapy will be completely different. Colorectal medullary carcinoma, accounting for 2.2-3.2%, is characterized by massive lymphocyte infiltration. However, the attention to the immune characteristics of colorectal medullary carcinoma is insufficient. AREA COVERED We searched the literature about colorectal medullary carcinoma on PubMed through November 2023to investigate the hallmarks of colorectal medullary carcinoma's immune landscape, compare medullary carcinoma originating from different organs and provide theoretical evidence for precise treatment, including applying immunotherapy and BRAF inhibitors. EXPERT OPINION Colorectal medullary carcinoma is a pathological subtype with intense immune response, with six immune characteristics and has the potential to benefit from immunotherapy. Mismatch repair deficiency, ARID1A missing and BRAF V600E mutation often occurs. IFN-γ pathway is activated and PD-L1 expression is increased. Abundant lymphocyte infiltration performs tumor killing function. In addition, BRAF mutation plays an important role in the occurrence and development, and we can consider the combination of BRAF inhibitors and immunotherapy in patients with BRAF mutant. The exploration of colorectal medullary carcinoma will arouse researchers' attention to the correlation between pathological subtypes and immune response, and promote the process of precise immunotherapy.
Collapse
Affiliation(s)
- Haoyi Zou
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Chao Liu
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
- Key Laboratory of Tumor Immunology in Heilongjiang, Harbin Medical University Cancer Hospital, Harbin, China
| | - Yuli Ruan
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
- Key Laboratory of Tumor Immunology in Heilongjiang, Harbin Medical University Cancer Hospital, Harbin, China
| | - Lin Fang
- Phase I Clinical Research Center, The Affiliated Hospital of Qingdao University in Shandong, Qingdao, China
| | - Tong Wu
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Shuling Han
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Tianjiao Dang
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Hongxue Meng
- Department of Pathology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Yanqiao Zhang
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
- Key Laboratory of Tumor Immunology in Heilongjiang, Harbin Medical University Cancer Hospital, Harbin, China
- Clinical Research Center for Colorectal Cancer in Heilongjiang, Harbin Medical University Cancer Hospital, Harbin, China
| |
Collapse
|
79
|
Dong L, Tang Y, Wen S, He Y, Li F, Deng Y, Tao Z. Fecal Microbiota Transplantation Alleviates Allergic Rhinitis via CD4 + T Cell Modulation Through Gut Microbiota Restoration. Inflammation 2024; 47:1278-1297. [PMID: 38294580 DOI: 10.1007/s10753-024-01975-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Revised: 01/07/2024] [Accepted: 01/10/2024] [Indexed: 02/01/2024]
Abstract
Allergic rhinitis (AR) is an allergic condition of the upper respiratory tract with a complex pathogenesis, including epithelial barrier disruption, immune regulation, and gut microbiota, which is not yet fully understood. Gut microbiota is closely linked to allergic diseases, including AR. Fecal microbiota transplantation (FMT) has recently been recognized as a potentially effective therapy for allergic diseases. However, the efficacy and mechanism of action of FMT in AR remain unknown. Herein, we aimed to observe the implications of gut microbiota on epithelial barrier function and T cell homeostasis, as well as the effect of FMT in AR, using the ovalbumin (OVA)-induced AR mice. The intestinal microbiota of recipient mice was cleared using an antibiotic cocktail; thereafter, FMT was performed. Subsequently, the nasal symptom scores and histopathological features of colon and nasal mucosa tissues of mice were monitored, and serum OVA-sIgE and cytokines of IL-4, IFNγ, IL-17A, and IL-10 cytokine concentrations were examined. Thereafter, tight junction protein and CD4+ T cell-related transcription factor and cytokine expressions were observed in the colon and nasal mucosa, and changes in the expression of PI3K/AKT/mTOR and NFκB signaling pathway were detected by WB assay in each group. Fecal DNA was extracted from the four mice groups for high-throughput 16S rRNA sequencing. FMT ameliorated nasal symptoms and reduced nasal mucosal inflammation in AR mice. Moreover, according to 16S rRNA sequencing, FMT restored the disordered gut microbiota in AR mice. Following FMT, ZO-1 and claudin-1 and Th1/Th2/Th17-related transcription factor and cytokine expressions were upregulated, whereas Treg cell-related Foxp3 and IL-10 expressions were downregulated. Mechanistic studies have revealed that FMT also inhibited PI3K/AKT/mTOR and NF-κB pathway protein phosphorylation in AR mouse tissues. FMT alleviates allergic inflammation in AR by repairing the epithelial barrier and modulating CD4+ T cell balance and exerts anti-inflammatory effects through the PI3K/AKT/mTOR and NF-κB signaling pathways. Moreover, gut microbiota disorders are involved in AR pathogenesis. Disturbed gut microbiota causes an altered immune-inflammatory state in mice and increases susceptibility to AR. This study suggested the regulatory role of the gut-nose axis in the pathogenesis of AR is an emerging field, which provides novel directions and ideas for the treatment of AR.
Collapse
Affiliation(s)
- Lin Dong
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, 238 Jie-Fang Road, Wuhan, Hubei, 430060, People's Republic of China
| | - Yulei Tang
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, 238 Jie-Fang Road, Wuhan, Hubei, 430060, People's Republic of China
| | - Silu Wen
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, 238 Jie-Fang Road, Wuhan, Hubei, 430060, People's Republic of China
| | - Yan He
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, 238 Jie-Fang Road, Wuhan, Hubei, 430060, People's Republic of China
| | - Fen Li
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, 238 Jie-Fang Road, Wuhan, Hubei, 430060, People's Republic of China
- Department of Otolaryngology-Head and Neck Surgery, Central Laboratory, Renmin Hospital of Wuhan University, 238 Jie-Fang Road, Wuhan, Hubei, 430060, People's Republic of China
| | - Yuqin Deng
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, 238 Jie-Fang Road, Wuhan, Hubei, 430060, People's Republic of China.
| | - Zezhang Tao
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, 238 Jie-Fang Road, Wuhan, Hubei, 430060, People's Republic of China.
- Department of Otolaryngology-Head and Neck Surgery, Central Laboratory, Renmin Hospital of Wuhan University, 238 Jie-Fang Road, Wuhan, Hubei, 430060, People's Republic of China.
| |
Collapse
|
80
|
Roessner PM, Seufert I, Chapaprieta V, Jayabalan R, Briesch H, Massoni-Badosa R, Boskovic P, Benckendorff J, Roider T, Arseni L, Coelho M, Chakraborty S, Vaca AM, Sivina M, Muckenhuber M, Rodriguez-Rodriguez S, Bonato A, Herbst SA, Zapatka M, Sun C, Kretzmer H, Naake T, Bruch PM, Czernilofsky F, ten Hacken E, Schneider M, Helm D, Yosifov DY, Kauer J, Danilov AV, Bewarder M, Heyne K, Schneider C, Stilgenbauer S, Wiestner A, Mallm JP, Burger JA, Efremov DG, Lichter P, Dietrich S, Martin-Subero JI, Rippe K, Seiffert M. T-bet suppresses proliferation of malignant B cells in chronic lymphocytic leukemia. Blood 2024; 144:510-524. [PMID: 38684038 PMCID: PMC11307267 DOI: 10.1182/blood.2023021990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 03/28/2024] [Accepted: 04/13/2024] [Indexed: 05/02/2024] Open
Abstract
ABSTRACT The T-box transcription factor T-bet is known as a master regulator of the T-cell response but its role in malignant B cells has not been sufficiently explored. Here, we conducted single-cell resolved multi-omics analyses of malignant B cells from patients with chronic lymphocytic leukemia (CLL) and studied a CLL mouse model with a genetic knockout of Tbx21. We found that T-bet acts as a tumor suppressor in malignant B cells by decreasing their proliferation rate. NF-κB activity, induced by inflammatory signals provided by the microenvironment, triggered T-bet expression, which affected promoter-proximal and distal chromatin coaccessibility and controlled a specific gene signature by mainly suppressing transcription. Gene set enrichment analysis identified a positive regulation of interferon signaling and negative control of proliferation by T-bet. In line, we showed that T-bet represses cell cycling and is associated with longer overall survival of patients with CLL. Our study uncovered a novel tumor suppressive role of T-bet in malignant B cells via its regulation of inflammatory processes and cell cycling, which has implications for the stratification and therapy of patients with CLL. Linking T-bet activity to inflammation explains the good prognostic role of genetic alterations in the inflammatory signaling pathways in CLL.
Collapse
MESH Headings
- Leukemia, Lymphocytic, Chronic, B-Cell/pathology
- Leukemia, Lymphocytic, Chronic, B-Cell/genetics
- Leukemia, Lymphocytic, Chronic, B-Cell/immunology
- Leukemia, Lymphocytic, Chronic, B-Cell/metabolism
- T-Box Domain Proteins/genetics
- T-Box Domain Proteins/metabolism
- Animals
- Humans
- Cell Proliferation
- Mice
- B-Lymphocytes/pathology
- B-Lymphocytes/metabolism
- B-Lymphocytes/immunology
- Mice, Knockout
- Gene Expression Regulation, Leukemic
- NF-kappa B/metabolism
Collapse
Affiliation(s)
- Philipp M. Roessner
- Division of Molecular Genetics, German Cancer Research Center, Heidelberg, Germany
| | - Isabelle Seufert
- Division of Chromatin Networks, German Cancer Research Center and BioQuant, Heidelberg, Germany
- Faculty of Biosciences, University of Heidelberg, Heidelberg, Germany
| | | | - Ruparoshni Jayabalan
- Division of Molecular Genetics, German Cancer Research Center, Heidelberg, Germany
- Faculty of Biosciences, University of Heidelberg, Heidelberg, Germany
| | - Hannah Briesch
- Division of Molecular Genetics, German Cancer Research Center, Heidelberg, Germany
- Faculty of Biosciences, University of Heidelberg, Heidelberg, Germany
| | - Ramon Massoni-Badosa
- Division of Molecular Genetics, German Cancer Research Center, Heidelberg, Germany
- Single Cell Genomics, Centre for Genomic Regulation, Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Pavle Boskovic
- Division of Molecular Genetics, German Cancer Research Center, Heidelberg, Germany
- Faculty of Biosciences, University of Heidelberg, Heidelberg, Germany
| | | | - Tobias Roider
- Department of Medicine V, Heidelberg University Hospital, Heidelberg, Germany
- Molecular Medicine Partnership Unit, Heidelberg, Germany
| | - Lavinia Arseni
- Division of Molecular Genetics, German Cancer Research Center, Heidelberg, Germany
| | - Mariana Coelho
- Division of Molecular Genetics, German Cancer Research Center, Heidelberg, Germany
- Faculty of Biosciences, University of Heidelberg, Heidelberg, Germany
| | - Supriya Chakraborty
- Molecular Hematology, International Centre for Genetic Engineering and Biotechnology, Trieste, Italy
| | - Alicia M. Vaca
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Mariela Sivina
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Markus Muckenhuber
- Division of Chromatin Networks, German Cancer Research Center and BioQuant, Heidelberg, Germany
| | | | - Alice Bonato
- Molecular Hematology, International Centre for Genetic Engineering and Biotechnology, Trieste, Italy
| | - Sophie A. Herbst
- Department of Medicine V, Heidelberg University Hospital, Heidelberg, Germany
- Molecular Medicine Partnership Unit, Heidelberg, Germany
| | - Marc Zapatka
- Division of Molecular Genetics, German Cancer Research Center, Heidelberg, Germany
| | - Clare Sun
- Laboratory of Lymphoid Malignancies, Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Helene Kretzmer
- Department of Genome Regulation, Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Thomas Naake
- Genome Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Peter-Martin Bruch
- Department of Medicine V, Heidelberg University Hospital, Heidelberg, Germany
- Molecular Medicine Partnership Unit, Heidelberg, Germany
- Department of Hematology, Oncology and Clinical Immunology, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Felix Czernilofsky
- Department of Medicine V, Heidelberg University Hospital, Heidelberg, Germany
- Molecular Medicine Partnership Unit, Heidelberg, Germany
| | | | - Martin Schneider
- Proteomics Core Facility, German Cancer Research Center, Heidelberg, Germany
| | - Dominic Helm
- Proteomics Core Facility, German Cancer Research Center, Heidelberg, Germany
| | - Deyan Y. Yosifov
- Division of Chronic Lymphocytic Leukemia, Department of Internal Medicine III, Ulm University, Ulm, Germany
- Cooperation Unit Mechanisms of Leukemogenesis, German Cancer Research Center, Heidelberg, Germany
| | - Joseph Kauer
- Department of Medicine V, Heidelberg University Hospital, Heidelberg, Germany
- Molecular Medicine Partnership Unit, Heidelberg, Germany
| | - Alexey V. Danilov
- Department of Hematology, City of Hope National Medical Center, Duarte, CA
| | - Moritz Bewarder
- José Carreras Center for Immuno- and Gene Therapy and Internal Medicine I, Saarland University Medical School, Homburg/Saar, Germany
| | - Kristina Heyne
- José Carreras Center for Immuno- and Gene Therapy and Internal Medicine I, Saarland University Medical School, Homburg/Saar, Germany
| | - Christof Schneider
- Division of Chronic Lymphocytic Leukemia, Department of Internal Medicine III, Ulm University, Ulm, Germany
| | - Stephan Stilgenbauer
- Division of Chronic Lymphocytic Leukemia, Department of Internal Medicine III, Ulm University, Ulm, Germany
| | - Adrian Wiestner
- Laboratory of Lymphoid Malignancies, Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Jan-Philipp Mallm
- Division of Chromatin Networks, German Cancer Research Center and BioQuant, Heidelberg, Germany
| | - Jan A. Burger
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Dimitar G. Efremov
- Molecular Hematology, International Centre for Genetic Engineering and Biotechnology, Trieste, Italy
| | - Peter Lichter
- Division of Molecular Genetics, German Cancer Research Center, Heidelberg, Germany
| | - Sascha Dietrich
- Department of Medicine V, Heidelberg University Hospital, Heidelberg, Germany
- Molecular Medicine Partnership Unit, Heidelberg, Germany
- Department of Hematology, Oncology and Clinical Immunology, University Hospital Düsseldorf, Düsseldorf, Germany
| | - José I. Martin-Subero
- Institut d’Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats, Barcelona, Spain
| | - Karsten Rippe
- Division of Chromatin Networks, German Cancer Research Center and BioQuant, Heidelberg, Germany
| | - Martina Seiffert
- Division of Molecular Genetics, German Cancer Research Center, Heidelberg, Germany
| |
Collapse
|
81
|
Cardani-Boulton A, Lin F, Bergmann CC. CD6 Regulates CD4 T Follicular Helper Cell Differentiation and Humoral Immunity During Murine Coronavirus Infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.26.605237. [PMID: 39091786 PMCID: PMC11291160 DOI: 10.1101/2024.07.26.605237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/04/2024]
Abstract
During activation the T cell transmembrane receptor CD6 becomes incorporated into the T cell immunological synapse where it can exert both co-stimulatory and co-inhibitory functions. Given the ability of CD6 to carry out opposing functions, this study sought to determine how CD6 regulates early T cell activation in response to viral infection. Infection of CD6 deficient mice with a neurotropic murine coronavirus resulted in greater activation and expansion of CD4 T cells in the draining lymph nodes. Further analysis demonstrated that there was also preferential differentiation of CD4 T cells into T follicular helper cells, resulting in accelerated germinal center responses and emergence of high affinity virus specific antibodies. Given that CD6 conversely supports CD4 T cell activation in many autoimmune models, we probed potential mechanisms of CD6 mediated suppression of CD4 T cell activation during viral infection. Analysis of CD6 binding proteins revealed that infection induced upregulation of Ubash3a, a negative regulator of T cell receptor signaling, was hindered in CD6 deficient lymph nodes. Consistent with greater T cell activation and reduced UBASH3a activity, the T cell receptor signal strength was intensified in CD6 deficient CD4 T cells. These results reveal a novel immunoregulatory role for CD6 in limiting CD4 T cell activation and deterring CD4 T follicular helper cell differentiation, thereby attenuating antiviral humoral immunity.
Collapse
Affiliation(s)
- Amber Cardani-Boulton
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
- Case Western Reserve University, Cleveland Clinic, Lerner College of Medicine, Cleveland, OH
| | - Feng Lin
- Case Western Reserve University, Cleveland Clinic, Lerner College of Medicine, Cleveland, OH
- Department of Immunity and Inflammation, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
| | - Cornelia C Bergmann
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
- Case Western Reserve University, Cleveland Clinic, Lerner College of Medicine, Cleveland, OH
| |
Collapse
|
82
|
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
|
83
|
Hai S, Li X, Xie E, Wu W, Gao Q, Yu B, Hu J, Xu F, Zheng X, Zhang BH, Wu D, Yan W, Ning Q, Wang X. Intestinal IL-33 promotes microbiota-derived trimethylamine N -oxide synthesis and drives metabolic dysfunction-associated steatotic liver disease progression by exerting dual regulation on HIF-1α. Hepatology 2024:01515467-990000000-00950. [PMID: 38985971 DOI: 10.1097/hep.0000000000000985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 06/13/2024] [Indexed: 07/12/2024]
Abstract
BACKGROUND AND AIMS Gut microbiota plays a prominent role in the pathogenesis of metabolic dysfunction-associated steatotic liver disease (MASLD). IL-33 is highly expressed at mucosal barrier sites and regulates intestinal homeostasis. Herein, we aimed to investigate the role and mechanism of intestinal IL-33 in MASLD. APPROACH AND RESULTS In both humans and mice with MASLD, hepatic expression of IL-33 and its receptor suppression of tumorigenicity 2 (ST2) showed no significant change compared to controls, while serum soluble ST2 levels in humans, as well as intestinal IL-33 and ST2 expression in mice were significantly increased in MASLD. Deletion of global or intestinal IL-33 in mice alleviated metabolic disorders, inflammation, and fibrosis associated with MASLD by reducing intestinal barrier permeability and rectifying gut microbiota dysbiosis. Transplantation of gut microbiota from IL-33 deficiency mice prevented MASLD progression in wild-type mice. Moreover, IL-33 deficiency resulted in a decrease in the abundance of trimethylamine N -oxide-producing bacteria. Inhibition of trimethylamine N -oxide synthesis by 3,3-dimethyl-1-butanol mitigated hepatic oxidative stress in mice with MASLD. Nuclear IL-33 bound to hypoxia-inducible factor-1α and suppressed its activation, directly damaging the integrity of the intestinal barrier. Extracellular IL-33 destroyed the balance of intestinal Th1/Th17 and facilitated Th1 differentiation through the ST2- Hif1a - Tbx21 axis. Knockout of ST2 resulted in a diminished MASLD phenotype resembling that observed in IL-33 deficiency mice. CONCLUSIONS Intestinal IL-33 enhanced gut microbiota-derived trimethylamine N -oxide synthesis and aggravated MASLD progression through dual regulation on hypoxia-inducible factor-1α. Targeting IL-33 and its associated microbiota may provide a potential therapeutic strategy for managing MASLD.
Collapse
Affiliation(s)
- Suping Hai
- Department of Infectious Diseases, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonostic Infectious Disease, Huazhong University of Science and Technology, Wuhan, China
| | - Xitang Li
- Department of Infectious Diseases, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonostic Infectious Disease, Huazhong University of Science and Technology, Wuhan, China
| | - Erliang Xie
- Department of Infectious Diseases, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonostic Infectious Disease, Huazhong University of Science and Technology, Wuhan, China
| | - Wenhui Wu
- Department of Infectious Diseases, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonostic Infectious Disease, Huazhong University of Science and Technology, Wuhan, China
| | - Qiang Gao
- Department of Infectious Diseases, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonostic Infectious Disease, Huazhong University of Science and Technology, Wuhan, China
| | - Binghui Yu
- Department of Infectious Diseases, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonostic Infectious Disease, Huazhong University of Science and Technology, Wuhan, China
| | - Junjian Hu
- Department of Infectious Diseases, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonostic Infectious Disease, Huazhong University of Science and Technology, Wuhan, China
| | - Feiyang Xu
- Department of Infectious Diseases, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonostic Infectious Disease, Huazhong University of Science and Technology, Wuhan, China
| | - Xizhe Zheng
- Department of Infectious Diseases, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonostic Infectious Disease, Huazhong University of Science and Technology, Wuhan, China
| | - Bin-Hao Zhang
- Department of Surgery, Hepatic Surgery Center, Institute of Hepato-Pancreato-Biliary Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Di Wu
- Department of Infectious Diseases, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonostic Infectious Disease, Huazhong University of Science and Technology, Wuhan, China
| | - Weiming Yan
- Department of Infectious Diseases, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonostic Infectious Disease, Huazhong University of Science and Technology, Wuhan, China
| | - Qin Ning
- Department of Infectious Diseases, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonostic Infectious Disease, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaojing Wang
- Department of Infectious Diseases, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonostic Infectious Disease, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
84
|
Tolomeo M, Cascio A. The Complex Dysregulations of CD4 T Cell Subtypes in HIV Infection. Int J Mol Sci 2024; 25:7512. [PMID: 39062756 PMCID: PMC11276885 DOI: 10.3390/ijms25147512] [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: 06/04/2024] [Revised: 07/04/2024] [Accepted: 07/07/2024] [Indexed: 07/28/2024] Open
Abstract
Human immunodeficiency virus (HIV) infection remains an important global public health problem. About 40 million people are infected with HIV, and this infection caused about 630,000 deaths in 2022. The hallmark of HIV infection is the depletion of CD4+ T helper lymphocytes (Th cells). There are at least seven different Th subtypes, and not all are the main targets of HIV. Moreover, the effect of the virus in a specific subtype can be completely different from that of the others. Although the most compromised Th subtype in HIV infection is Th17, HIV can induce important dysregulations in other subtypes, such as follicular Th (Tfh) cells and regulatory Th cells (Treg cells or Tregs). Several studies have shown that HIV can induce an increase in the immunosuppressive activity of Tregs without causing a significant reduction in their numbers, at least in the early phase of infection. The increased activity of this Th subtype seems to play an important role in determining the immunodeficiency status of HIV-infected patients, and Tregs may represent a new target for innovative anti-HIV therapies, including the so-called "Kick and Kill" therapeutic method whose goal is the complete elimination of the virus and the healing of HIV infection. In this review, we report the most important findings on the effects of HIV on different CD4+ T cell subtypes, the molecular mechanisms by which the virus impairs the functions of these cells, and the implications for new anti-HIV therapeutic strategies.
Collapse
Affiliation(s)
- Manlio Tolomeo
- Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties, University of Palermo, 90127 Palermo, Italy;
- Department of Infectious Diseases, A.O.U.P. Palermo, 90127 Palermo, Italy
| | - Antonio Cascio
- Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties, University of Palermo, 90127 Palermo, Italy;
- Department of Infectious Diseases, A.O.U.P. Palermo, 90127 Palermo, Italy
| |
Collapse
|
85
|
Chen L, Yi H, Li Q, Duan T, Liu X, Li L, Wang HY, Xing C, Wang R. T-bet Regulates Ion Channels and Transporters and Induces Apoptosis in Intestinal Epithelial Cells. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2401654. [PMID: 38650111 PMCID: PMC11267362 DOI: 10.1002/advs.202401654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 03/22/2024] [Indexed: 04/25/2024]
Abstract
T-bet, encoded by TBX21, is extensively expressed across various immune cell types, and orchestrates critical functions in their development, survival, and physiological activities. However, the role of T-bet in non-immune compartments, notably the epithelial cells, remains obscure. Herein, a Tet-O-T-bet transgenic mouse strain is generated for doxycycline-inducible T-bet expression in adult animals. Unexpectedly, ubiquitous T-bet overexpression causes acute diarrhea, intestinal damage, and rapid mortality. Cell-type-specific analyses reveal that T-bet-driven pathology is not attributable to its overexpression in CD4+ T cells or myeloid lineages. Instead, inducible T-bet overexpression in the intestinal epithelial cells is the critical determinant of the observed lethal phenotype. Mechanistically, T-bet overexpression modulates ion channel and transporter profiles in gut epithelial cells, triggering profound fluid secretion and subsequent lethal dehydration. Furthermore, ectopic T-bet expression enhances gut epithelial cell apoptosis and markedly suppresses colon cancer development in xenograft models. Collectively, the findings unveil a previously unrecognized role of T-bet in intestinal epithelial cells for inducing apoptosis, diarrhea, and local inflammation, thus implicating its potential as a therapeutic target for the treatment of cancer and inflammatory diseases.
Collapse
Affiliation(s)
- Lang Chen
- Center for Inflammation and EpigeneticsHouston Methodist Research InstituteHoustonTX77030USA
- Department of General SurgeryThird Xiangya HospitalXiangya School of MedicineCentral South UniversityChangsha410013China
| | - Hongwei Yi
- Center for Inflammation and EpigeneticsHouston Methodist Research InstituteHoustonTX77030USA
- Department of PharmacologySchool of MedicineSoutheast UniversityNanjing210009China
| | - Qingtian Li
- Center for Inflammation and EpigeneticsHouston Methodist Research InstituteHoustonTX77030USA
- Department of MedicineBaylor College of MedicineHoustonTX77030USA
| | - Tianhao Duan
- Center for Inflammation and EpigeneticsHouston Methodist Research InstituteHoustonTX77030USA
- Department of MedicineKeck School of MedicineUniversity of Southern CaliforniaLos AngelesCA90033USA
| | - Xin Liu
- Center for Inflammation and EpigeneticsHouston Methodist Research InstituteHoustonTX77030USA
- Department of MedicineKeck School of MedicineUniversity of Southern CaliforniaLos AngelesCA90033USA
| | - Linfeng Li
- Center for Inflammation and EpigeneticsHouston Methodist Research InstituteHoustonTX77030USA
- Department of Thoracic SurgeryXiangya HospitalCentral South UniversityChangsha410008China
| | - Helen Y. Wang
- Center for Inflammation and EpigeneticsHouston Methodist Research InstituteHoustonTX77030USA
- Department of MedicineKeck School of MedicineUniversity of Southern CaliforniaLos AngelesCA90033USA
| | - Changsheng Xing
- Center for Inflammation and EpigeneticsHouston Methodist Research InstituteHoustonTX77030USA
- Department of MedicineKeck School of MedicineUniversity of Southern CaliforniaLos AngelesCA90033USA
| | - Rong‐Fu Wang
- Center for Inflammation and EpigeneticsHouston Methodist Research InstituteHoustonTX77030USA
- Department of MedicineKeck School of MedicineUniversity of Southern CaliforniaLos AngelesCA90033USA
- Department of PediatricsChildren's Hospital Los AngelesKeck School of MedicineUniversity of Southern CaliforniaLos AngelesCA90027USA
- Norris Comprehensive Cancer CenterKeck School of MedicineUniversity of Southern CaliforniaLos AngelesCA90033USA
| |
Collapse
|
86
|
Pal S, Dhar R. Living in a noisy world-origins of gene expression noise and its impact on cellular decision-making. FEBS Lett 2024; 598:1673-1691. [PMID: 38724715 DOI: 10.1002/1873-3468.14898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 03/23/2024] [Accepted: 03/27/2024] [Indexed: 07/23/2024]
Abstract
The expression level of a gene can vary between genetically identical cells under the same environmental condition-a phenomenon referred to as gene expression noise. Several studies have now elucidated a central role of transcription factors in the generation of expression noise. Transcription factors, as the key components of gene regulatory networks, drive many important cellular decisions in response to cellular and environmental signals. Therefore, a very relevant question is how expression noise impacts gene regulation and influences cellular decision-making. In this Review, we summarize the current understanding of the molecular origins of expression noise, highlighting the role of transcription factors in this process, and discuss the ways in which noise can influence cellular decision-making. As advances in single-cell technologies open new avenues for studying expression noise as well as gene regulatory circuits, a better understanding of the influence of noise on cellular decisions will have important implications for many biological processes.
Collapse
Affiliation(s)
- Sampriti Pal
- Department of Bioscience and Biotechnology, IIT Kharagpur, India
| | - Riddhiman Dhar
- Department of Bioscience and Biotechnology, IIT Kharagpur, India
| |
Collapse
|
87
|
He J, Chen D, Xiong W, Hou X, Quan Y, Yang M, Dong Z. Eomesodermin spatiotemporally orchestrates the early and late stages of NK cell development by targeting KLF2 and T-bet, respectively. Cell Mol Immunol 2024; 21:662-673. [PMID: 38740922 PMCID: PMC11214621 DOI: 10.1038/s41423-024-01164-8] [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: 07/22/2023] [Accepted: 04/07/2024] [Indexed: 05/16/2024] Open
Abstract
Eomesodermin (Eomes) is a critical factor in the development of natural killer (NK) cells, but its precise role in temporal and spatial coordination during this process remains unclear. Our study revealed that Eomes plays distinct roles during the early and late stages of NK cell development. Specifically, the early deletion of Eomes via the CD122-Cre transgene resulted in significant blockade at the progenitor stage due to the downregulation of KLF2, another important transcription factor. ChIP-seq revealed direct binding of Eomes to the conserved noncoding sequence (CNS) of Klf2. Utilizing the CHimeric IMmune Editing (CHIME) technique, we found that deletion of the CNS region of Klf2 via CRISPRi led to a reduction in the NK cell population and developmental arrest. Moreover, constitutive activation of this specific CNS region through CRISPRa significantly reversed the severe defects in NK cell development caused by Eomes deficiency. Conversely, Ncr1-Cre-mediated terminal deletion of Eomes expedited the transition of NK cell subsets from the CD27+CD11b+ phenotype to the CD27-CD11b+ phenotype. Late-stage deficiency of Eomes led to a significant increase in T-bet expression, which subsequently increased the expression of the transcription factor Zeb2. Genetic deletion of one allele of Tbx21, encoding T-bet, effectively reversed the aberrant differentiation of Eomes-deficient NK cells. In summary, we utilized two innovative genetic models to elucidate the intricate mechanisms underlying Eomes-mediated NK cell commitment and differentiation.
Collapse
Affiliation(s)
- Junming He
- The First Affiliated Hospital of Anhui Medical University and Institute for Clinical Immunology, Anhui Medical University, Anhui, 230032, China
- State Key Laboratory of Membrane Biology, School of Medicine and Institute for Immunology, Tsinghua University, 100084, Beijing, China
| | - Donglin Chen
- State Key Laboratory of Membrane Biology, School of Medicine and Institute for Immunology, Tsinghua University, 100084, Beijing, China
| | - Wei Xiong
- State Key Laboratory of Membrane Biology, School of Medicine and Institute for Immunology, Tsinghua University, 100084, Beijing, China
| | - Xinlei Hou
- State Key Laboratory of Membrane Biology, School of Medicine and Institute for Immunology, Tsinghua University, 100084, Beijing, China
| | - Yuhe Quan
- State Key Laboratory of Membrane Biology, School of Medicine and Institute for Immunology, Tsinghua University, 100084, Beijing, China
| | - Meixiang Yang
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital Affiliated with Jinan University, Jinan University, Zhuhai, 519000, China.
- The Biomedical Translational Research Institute. Key Laboratory of Ministry of Education for Viral Pathogenesis & Infection Prevention and Control (Jinan University). Guangzhou Key Laboratory for Germ-Free Animals and Microbiota Application. School of Medicine. Jinan University, Guangzhou, 510632, China.
| | - Zhongjun Dong
- The First Affiliated Hospital of Anhui Medical University and Institute for Clinical Immunology, Anhui Medical University, Anhui, 230032, China.
- State Key Laboratory of Membrane Biology, School of Medicine and Institute for Immunology, Tsinghua University, 100084, Beijing, China.
- Innovative Institute of Tumor Immunity and Medicine (ITIM), Hefei, 230032, China.
- Anhui Province Key Laboratory of Tumor Immune Microenvironment and Immunotherapy, Hefei, 230032, China.
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, 230032, China.
| |
Collapse
|
88
|
Yang AYP, Wistuba-Hamprecht K, Greten TF, Ruf B. Innate-like T cells in liver disease. Trends Immunol 2024; 45:535-548. [PMID: 38879436 DOI: 10.1016/j.it.2024.05.008] [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: 04/28/2024] [Revised: 05/27/2024] [Accepted: 05/27/2024] [Indexed: 07/14/2024]
Abstract
Mammalian innate-like T cells (ILTCs), including mucosal-associated invariant T (MAIT), natural killer T (NKT), and γδ T cells, are abundant tissue-resident lymphocytes that have recently emerged as orchestrators of hepatic inflammation, tissue repair, and immune homeostasis. This review explores the involvement of different ILTC subsets in liver diseases. We explore the mechanisms underlying the pro- and anti-inflammatory effector functions of ILTCs in a context-dependent manner. We highlight latest findings regarding the dynamic interplay between ILTC functional subsets and other immune and parenchymal cells which may inform candidate immunomodulatory strategies to achieve improved clinical outcomes in liver diseases. We present new insights into how distinct gene expression programs in hepatic ILTCs are induced, maintained, and reprogrammed in a context- and disease stage-dependent manner.
Collapse
Affiliation(s)
- Albert Ying-Po Yang
- Department of Internal Medicine I, University Hospital Tübingen, Eberhard Karls University of Tübingen, Tübingen, Germany; M3 Research Center for Malignome, Metabolome, and Microbiome, Faculty of Medicine, University of Tübingen, Tübingen, Germany
| | - Kilian Wistuba-Hamprecht
- Department of Internal Medicine I, University Hospital Tübingen, Eberhard Karls University of Tübingen, Tübingen, Germany; M3 Research Center for Malignome, Metabolome, and Microbiome, Faculty of Medicine, University of Tübingen, Tübingen, Germany; Cluster of Excellence iFIT (EXC 2180) - Image-Guided and Functionally Instructed Tumor Therapies, University of Tübingen, Tübingen, Germany; Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany; Department of Dermatology, Venereology, and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, Mannheim, Germany; DKFZ Hector Cancer Institute at the University Medical Center Mannheim, Mannheim, Germany
| | - Tim F Greten
- Gastrointestinal Malignancy Section, Thoracic and Gastrointestinal Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA; Center for Cancer Research (CCR) Liver Cancer Program, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Benjamin Ruf
- Department of Internal Medicine I, University Hospital Tübingen, Eberhard Karls University of Tübingen, Tübingen, Germany; M3 Research Center for Malignome, Metabolome, and Microbiome, Faculty of Medicine, University of Tübingen, Tübingen, Germany; Cluster of Excellence iFIT (EXC 2180) - Image-Guided and Functionally Instructed Tumor Therapies, University of Tübingen, Tübingen, Germany.
| |
Collapse
|
89
|
Elsner RA, Smita S, Shlomchik MJ. IL-12 induces a B cell-intrinsic IL-12/IFNγ feed-forward loop promoting extrafollicular B cell responses. Nat Immunol 2024; 25:1283-1295. [PMID: 38862796 PMCID: PMC11992614 DOI: 10.1038/s41590-024-01858-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 04/26/2024] [Indexed: 06/13/2024]
Abstract
While some infections elicit germinal centers, others produce only extrafollicular responses. The mechanisms controlling these dichotomous fates are poorly understood. We identify IL-12 as a cytokine switch, acting directly on B cells to promote extrafollicular and suppress germinal center responses. IL-12 initiates a B cell-intrinsic feed-forward loop between IL-12 and IFNγ, amplifying IFNγ production, which promotes proliferation and plasmablast differentiation from mouse and human B cells, in synergy with IL-12. IL-12 sustains the expression of a portion of IFNγ-inducible genes. Together, they also induce unique gene changes, reflecting both IFNγ amplification and cooperative effects between both cytokines. In vivo, cells lacking both IL-12 and IFNγ receptors are more impaired in plasmablast production than those lacking either receptor alone. Further, B cell-derived IL-12 enhances both plasmablast responses and T helper 1 cell commitment. Thus, B cell-derived IL-12, acting on T and B cells, determines the immune response mode, with implications for vaccines, pathogen protection and autoimmunity.
Collapse
Affiliation(s)
- Rebecca A Elsner
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA.
| | - Shuchi Smita
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Mark J Shlomchik
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
90
|
Wang X, Li N, Zhang J, Wang J, Wei Y, Yang J, Sun D, Liu L, Nian H, Wei R. AS101 regulates the Teff/Treg balance to alleviate rabbit autoimmune dacryoadenitis through modulating NFATc2. Exp Eye Res 2024; 244:109937. [PMID: 38782179 DOI: 10.1016/j.exer.2024.109937] [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: 02/22/2024] [Revised: 04/27/2024] [Accepted: 05/20/2024] [Indexed: 05/25/2024]
Abstract
Sjögren's syndrome (SS) dry eye can cause ocular surface inflammation and lacrimal gland (LG) damage, leading to discomfort and potential vision problems. The existing treatment options for SS dry eye are currently constrained. We investigated the possible therapeutic effect and the underlying mechanism of AS101 in autoimmune dry eye. AS101 was injected subconjunctivally into a rabbit model of autoimmune dacryoadenitis and its therapeutic effects were determined by evaluating clinical and histological scores. The expressions of effector T cells (Teff)/regulatory T cells (Treg)-related transcription factors and cytokines, inflammation mediators, and transcription factor NFATc2 were measured by quantitative real-time PCR and/or Western blot both in vivo and in vitro. Additionally, the role of NFATc2 in the immunomodulatory effects of AS101 on T cells was explored by co-culturing activated peripheral blood lymphocytes (PBLs) transfected with NFATc2 overexpression lentiviral plasmid with AS101. AS101 treatment potently ameliorated the clinical severity and reduced the inflammation of LG. Further investigation revealed that AS101 treatment led to decreased expression of Th1-related genes (T-bet and IFN-γ) and Th17-related genes (RORC, IL-17A, IL-17F, and GM-CSF) and increased expression of Treg-related gene Foxp3 in vivo and in vitro. Meanwhile, AS101 suppressed the expression of TNF-α, IL-1β, IL-23, IL-6, MMP-2, and MMP-9. Mechanistically, AS101 downregulated the expression of NFATc2 in inflamed LGs. Overexpression of NFATc2 in activated PBLs partially blunted the effect of AS101 on Teff suppression and Treg promotion. In conclusion, AS101 is a potential regulator of Teff/Treg cell balance and could be an effective treatment agent for SS dry eye.
Collapse
Affiliation(s)
- Xiu Wang
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin China, China
| | - Na Li
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin China, China
| | - Jiawen Zhang
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin China, China
| | - Jiali Wang
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin China, China
| | - Yankai Wei
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin China, China
| | - Jun Yang
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin China, China
| | - Deming Sun
- Doheny Eye Institute, And Department of Ophthalmology, David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, CA, United States
| | - Lin Liu
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin China, China
| | - Hong Nian
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin China, China.
| | - Ruihua Wei
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin China, China.
| |
Collapse
|
91
|
Kannan N, Choi A, Rivera De Jesus MA, Wei PM, Sahler JM, Curley SM, August A, DeLisa MP, Whittaker GR, Putnam D. Intranasal Vaccination with Recombinant TLR2-Active Outer Membrane Vesicles Containing Sequential M2e Epitopes Protects against Lethal Influenza a Challenge. Vaccines (Basel) 2024; 12:724. [PMID: 39066362 PMCID: PMC11281606 DOI: 10.3390/vaccines12070724] [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: 05/23/2024] [Revised: 06/14/2024] [Accepted: 06/20/2024] [Indexed: 07/28/2024] Open
Abstract
Influenza is a highly contagious respiratory disease, resulting in an estimated 3 to 5 million cases of severe illness annually. While most influenza vaccines are administered parenterally via injection, one shortcoming is that they do not generate a strong immune response at the site of infection, which can become important in a pandemic. Intranasal vaccines can generate both local and systemic protective immune responses, can reduce costs, and enhance ease of administration. Previous studies showed that parenterally administered outer membrane vesicles (OMVs) that carry sequences of the M2e protein (OMV-M2e) protect against influenza A/PR8 challenge in mice and ferrets. In the current study, we measured the effectiveness of the intranasal route of the OMV-M2e vaccine against the influenza A/PR8 strain in mice. We observed high anti-M2e IgG and IgA titers post-challenge in mice vaccinated intranasally with OMV-M2e. In addition, we observed a Th1/Tc1 bias in the vaccinated mice, and an increased Th17/Tc17 response, both of which correlated with survival to A/PR8 challenge and significantly lower lung viral titers. We conclude that the intranasal-route administration of the OMV-M2e vaccine is a promising approach toward generating protection against influenza A as it leads to an increased proinflammatory immune response correlating with survival to viral challenge.
Collapse
Affiliation(s)
- Nisha Kannan
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA; (N.K.); (M.A.R.D.J.); (P.M.W.); (S.M.C.)
| | - Annette Choi
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY 14853, USA; (A.C.); (J.M.S.); (A.A.); (G.R.W.)
| | - Mariela A. Rivera De Jesus
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA; (N.K.); (M.A.R.D.J.); (P.M.W.); (S.M.C.)
| | - Peter Male Wei
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA; (N.K.); (M.A.R.D.J.); (P.M.W.); (S.M.C.)
| | - Julie Marie Sahler
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY 14853, USA; (A.C.); (J.M.S.); (A.A.); (G.R.W.)
| | - Stephanie Marie Curley
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA; (N.K.); (M.A.R.D.J.); (P.M.W.); (S.M.C.)
| | - Avery August
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY 14853, USA; (A.C.); (J.M.S.); (A.A.); (G.R.W.)
| | - Matthew P. DeLisa
- Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, NY 14853, USA;
| | - Gary R. Whittaker
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY 14853, USA; (A.C.); (J.M.S.); (A.A.); (G.R.W.)
| | - David Putnam
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA; (N.K.); (M.A.R.D.J.); (P.M.W.); (S.M.C.)
- Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, NY 14853, USA;
| |
Collapse
|
92
|
Contreras-Castillo E, García-Rasilla VY, García-Patiño MG, Licona-Limón P. Stability and plasticity of regulatory T cells in health and disease. J Leukoc Biol 2024; 116:33-53. [PMID: 38428948 DOI: 10.1093/jleuko/qiae049] [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: 11/14/2023] [Revised: 02/09/2024] [Accepted: 02/19/2024] [Indexed: 03/03/2024] Open
Abstract
The mechanisms that negatively regulate inflammation upon a pathogenic stimulus are crucial for the maintenance of tissue integrity and organ function. T regulatory cells are one of the main drivers in controlling inflammation. The ability of T regulatory cells to adapt to different inflammatory cues and suppress inflammation is one of the relevant features of T regulatory cells. During this process, T regulatory cells express different transcription factors associated with their counterparts, Th helper cells, including Tbx21, GATA-3, Bcl6, and Rorc. The acquisition of this transcription factor helps the T regulatory cells to suppress and migrate to the different inflamed tissues. Additionally, the T regulatory cells have different mechanisms that preserve stability while acquiring a particular T regulatory cell subtype. This review focuses on describing T regulatory cell subtypes and the mechanisms that maintain their identity in health and diseases.
Collapse
Affiliation(s)
- Eugenio Contreras-Castillo
- Departamento de Biología Celular y del Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Circuito exterior s/n, CU Coyoacán, México City 04510, Mexico
| | - Verónica Yutsil García-Rasilla
- Departamento de Biología Celular y del Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Circuito exterior s/n, CU Coyoacán, México City 04510, Mexico
| | - María Guadalupe García-Patiño
- Departamento de Biología Celular y del Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Circuito exterior s/n, CU Coyoacán, México City 04510, Mexico
| | - Paula Licona-Limón
- Departamento de Biología Celular y del Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Circuito exterior s/n, CU Coyoacán, México City 04510, Mexico
| |
Collapse
|
93
|
Caldirola MS, Daiana E, Gomez Raccio AC, García AL, Bernacchia A, Medín M, Gaillard MI, Di Giovanni D. Case Report: Common variable immunodeficiency phenotype and granulomatous-lymphocytic interstitial lung disease with a novel SOCS1 variant. Front Pediatr 2024; 12:1423858. [PMID: 39005503 PMCID: PMC11239428 DOI: 10.3389/fped.2024.1423858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 06/10/2024] [Indexed: 07/16/2024] Open
Abstract
Common variable immunodeficiency is a heterogeneous symptomatic group of inborn errors of immunity that mainly affects antibodies production and/or function, predisposing patients to recurrent and severe infections. More than half of them usually develop autoimmunity, lymphoproliferation, enteropathy, and malignancies. Among these conditions, chronic lung disease such as granulomatous-lymphocytic interstitial lung disease is one of the leading causes of death in these patients. Recently, many genes that play a key role in B and T cells' development, maintenance, and/or cytokines signaling pathways have been implicated in the pathogenesis of the disease. Here, we describe the first Argentinian patient presenting with common variable immunodeficiency and granulomatous-lymphocytic interstitial lung disease, harboring two in cis heterozygous variants in the SOCS1 gene.
Collapse
Affiliation(s)
- María Soledad Caldirola
- Servicio de Inmunología, Hospital de Niños “Dr. Ricardo Gutiérrez”, Buenos Aires, Argentina
- Instituto Multidisciplinario de Investigaciones en Patologías Pediátricas (IMIPP—CONICET-GCBA), Buenos Aires, Argentina
| | - Espantoso Daiana
- Servicio de Inmunología, Hospital de Niños “Dr. Ricardo Gutiérrez”, Buenos Aires, Argentina
| | | | - Ana Luz García
- Servicio de Inmunología, Hospital de Niños “Dr. Ricardo Gutiérrez”, Buenos Aires, Argentina
| | - Agustin Bernacchia
- Servicio de Inmunología, Hospital de Niños “Dr. Ricardo Gutiérrez”, Buenos Aires, Argentina
| | - Martín Medín
- Servicio de Anatomía Patológica, Hospital de Niños “Dr. Ricardo Gutiérrez”, Buenos Aires, Argentina
| | - Maria Isabel Gaillard
- Servicio de Inmunología, Hospital de Niños “Dr. Ricardo Gutiérrez”, Buenos Aires, Argentina
| | - Daniela Di Giovanni
- Servicio de Inmunología, Hospital de Niños “Dr. Ricardo Gutiérrez”, Buenos Aires, Argentina
| |
Collapse
|
94
|
Iijima N. The emerging role of effector functions exerted by tissue-resident memory T cells. OXFORD OPEN IMMUNOLOGY 2024; 5:iqae006. [PMID: 39193473 PMCID: PMC11213632 DOI: 10.1093/oxfimm/iqae006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 04/14/2024] [Accepted: 06/04/2024] [Indexed: 08/29/2024] Open
Abstract
The magnitude of the effector functions of memory T cells determines the consequences of the protection against invading pathogens and tumor development or the pathogenesis of autoimmune and allergic diseases. Tissue-resident memory T cells (TRM cells) are unique T-cell populations that persist in tissues for long periods awaiting re-encounter with their cognate antigen. Although TRM cell reactivation primarily requires the presentation of cognate antigens, recent evidence has shown that, in addition to the conventional concept, TRM cells can be reactivated without the presentation of cognate antigens. Non-cognate TRM cell activation is triggered by cross-reactive antigens or by several combinations of cytokines, including interleukin (IL)-2, IL-7, IL-12, IL-15 and IL-18. The activation mode of TRM cells reinforces their cytotoxic activity and promotes the secretion of effector cytokines (such as interferon-gamma and tumor necrosis factor-alpha). This review highlights the key features of TRM cell maintenance and reactivation and discusses the importance of effector functions that TRM cells exert upon being presented with cognate and/or non-cognate antigens, as well as cytokines secreted by TRM and non-TRM cells within the tissue microenvironment.
Collapse
Affiliation(s)
- Norifumi Iijima
- Center for Drug Design Research, National Institutes of Biomedical Innovation, Health and Nutrition (NIBN), Ibaraki, Osaka, Japan
| |
Collapse
|
95
|
Çubuk C, Lau R, Cutillas P, Rajeeve V, John CR, Surace AEA, Hands R, Fossati-Jimack L, Lewis MJ, Pitzalis C. Phosphoproteomic profiling of early rheumatoid arthritis synovium reveals active signalling pathways and differentiates inflammatory pathotypes. Arthritis Res Ther 2024; 26:120. [PMID: 38867295 PMCID: PMC11167927 DOI: 10.1186/s13075-024-03351-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 06/06/2024] [Indexed: 06/14/2024] Open
Abstract
BACKGROUND Kinases are intracellular signalling mediators and key to sustaining the inflammatory process in rheumatoid arthritis (RA). Oral inhibitors of Janus Kinase family (JAKs) are widely used in RA, while inhibitors of other kinase families e.g. phosphoinositide 3-kinase (PI3K) are under development. Most current biomarker platforms quantify mRNA/protein levels, but give no direct information on whether proteins are active/inactive. Phosphoproteome analysis has the potential to measure specific enzyme activation status at tissue level. METHODS We validated the feasibility of phosphoproteome and total proteome analysis on 8 pre-treatment synovial biopsies from treatment-naive RA patients using label-free mass spectrometry, to identify active cell signalling pathways in synovial tissue which might explain failure to respond to RA therapeutics. RESULTS Differential expression analysis and functional enrichment revealed clear separation of phosphoproteome and proteome profiles between lymphoid and myeloid RA pathotypes. Abundance of specific phosphosites was associated with the degree of inflammatory state. The lymphoid pathotype was enriched with lymphoproliferative signalling phosphosites, including Mammalian Target Of Rapamycin (MTOR) signalling, whereas the myeloid pathotype was associated with Mitogen-Activated Protein Kinase (MAPK) and CDK mediated signalling. This analysis also highlighted novel kinases not previously linked to RA, such as Protein Kinase, DNA-Activated, Catalytic Subunit (PRKDC) in the myeloid pathotype. Several phosphosites correlated with clinical features, such as Disease-Activity-Score (DAS)-28, suggesting that phosphosite analysis has potential for identifying novel biomarkers at tissue-level of disease severity and prognosis. CONCLUSIONS Specific phosphoproteome/proteome signatures delineate RA pathotypes and may have clinical utility for stratifying patients for personalised medicine in RA.
Collapse
Affiliation(s)
- Cankut Çubuk
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Queen Mary University of London and Barts NIHR BRC & NHS Trust, Charterhouse Square, London, EC1M 6BQ, UK
| | - Rachel Lau
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Queen Mary University of London and Barts NIHR BRC & NHS Trust, Charterhouse Square, London, EC1M 6BQ, UK
| | - Pedro Cutillas
- Cell Signalling and Proteomics Group, Centre for Genomics and Computational Biology, Barts Cancer Institute, Queen Mary University of London, London, EC1M 6BQ, UK
| | - Vinothini Rajeeve
- Cell Signalling and Proteomics Group, Centre for Genomics and Computational Biology, Barts Cancer Institute, Queen Mary University of London, London, EC1M 6BQ, UK
| | - Christopher R John
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Queen Mary University of London and Barts NIHR BRC & NHS Trust, Charterhouse Square, London, EC1M 6BQ, UK
| | - Anna E A Surace
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Queen Mary University of London and Barts NIHR BRC & NHS Trust, Charterhouse Square, London, EC1M 6BQ, UK
| | - Rebecca Hands
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Queen Mary University of London and Barts NIHR BRC & NHS Trust, Charterhouse Square, London, EC1M 6BQ, UK
| | - Liliane Fossati-Jimack
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Queen Mary University of London and Barts NIHR BRC & NHS Trust, Charterhouse Square, London, EC1M 6BQ, UK
| | - Myles J Lewis
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Queen Mary University of London and Barts NIHR BRC & NHS Trust, Charterhouse Square, London, EC1M 6BQ, UK.
| | - Costantino Pitzalis
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Queen Mary University of London and Barts NIHR BRC & NHS Trust, Charterhouse Square, London, EC1M 6BQ, UK.
- IRCCS Istituto Clinico Humanitas, Via Manzoni 56, Rozzao, Milan, Italy.
| |
Collapse
|
96
|
Nguyen C, Kudek M, Zander R, Niu H, Shen J, Bauer A, Alson D, Khatun A, Chen Y, Sun J, Drobyski W, Edelson BT, Cui W. Bhlhe40 Promotes CD4+ T Helper 1 Cell and Suppresses T Follicular Helper Cell Differentiation during Viral Infection. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:1829-1842. [PMID: 38619295 DOI: 10.4049/jimmunol.2300355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 03/18/2024] [Indexed: 04/16/2024]
Abstract
In response to acute infection, naive CD4+ T cells primarily differentiate into T helper 1 (Th1) or T follicular helper (Tfh) cells that play critical roles in orchestrating cellular or humoral arms of immunity, respectively. However, despite the well established role of T-bet and BCL-6 in driving Th1 and Tfh cell lineage commitment, respectively, whether additional transcriptional circuits also underlie the fate bifurcation of Th1 and Tfh cell subsets is not fully understood. In this article, we study how the transcriptional regulator Bhlhe40 dictates the Th1/Tfh differentiation axis in mice. CD4+ T cell-specific deletion of Bhlhe40 abrogates Th1 but augments Tfh differentiation. We also assessed an increase in germinal center B cells and Ab production, suggesting that deletion of Bhlhe40 in CD4+ T cells not only alters Tfh differentiation but also their capacity to provide help to B cells. To identify molecular mechanisms by which Bhlhe40 regulates Th1 versus Tfh lineage choice, we first performed epigenetic profiling in the virus specific Th1 and Tfh cells following LCMV infection, which revealed distinct promoter and enhancer activities between the two helper cell lineages. Furthermore, we identified that Bhlhe40 directly binds to cis-regulatory elements of Th1-related genes such as Tbx21 and Cxcr6 to activate their expression while simultaneously binding to regions of Tfh-related genes such as Bcl6 and Cxcr5 to repress their expression. Collectively, our data suggest that Bhlhe40 functions as a transcription activator to promote Th1 cell differentiation and a transcription repressor to suppress Tfh cell differentiation.
Collapse
Affiliation(s)
- Christine Nguyen
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI
- Versiti Blood Research Institute, Versiti Wisconsin, Milwaukee, WI
| | - Matthew Kudek
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI
- Versiti Blood Research Institute, Versiti Wisconsin, Milwaukee, WI
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI
| | - Ryan Zander
- Versiti Blood Research Institute, Versiti Wisconsin, Milwaukee, WI
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA
| | - Hongshen Niu
- Department of Pathology, Northwestern University, Chicago, IL
| | - Jian Shen
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI
- Versiti Blood Research Institute, Versiti Wisconsin, Milwaukee, WI
- Department of Pathology, Northwestern University, Chicago, IL
| | - Ashley Bauer
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI
- Versiti Blood Research Institute, Versiti Wisconsin, Milwaukee, WI
- Department of Pathology, Northwestern University, Chicago, IL
| | - Donia Alson
- Versiti Blood Research Institute, Versiti Wisconsin, Milwaukee, WI
| | - Achia Khatun
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI
- Versiti Blood Research Institute, Versiti Wisconsin, Milwaukee, WI
| | - Yao Chen
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI
- Versiti Blood Research Institute, Versiti Wisconsin, Milwaukee, WI
- Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jie Sun
- University of Virginia School of Medicine, Charlottesville, VA
| | - William Drobyski
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI
| | - Brian T Edelson
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO
| | - Weiguo Cui
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI
- Versiti Blood Research Institute, Versiti Wisconsin, Milwaukee, WI
- Department of Pathology, Northwestern University, Chicago, IL
| |
Collapse
|
97
|
Gao Y, Chen S, Jiao S, Fan Y, Li X, Tan N, Fang J, Xu L, Huang Y, Zhao J, Guo S, Liu T, Xu W. ATG5-regulated CCL2/MCP-1 production in myeloid cells selectively modulates anti-malarial CD4 + Th1 responses. Autophagy 2024; 20:1398-1417. [PMID: 38368631 PMCID: PMC11210915 DOI: 10.1080/15548627.2024.2319512] [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: 09/28/2023] [Revised: 02/06/2024] [Accepted: 02/12/2024] [Indexed: 02/20/2024] Open
Abstract
Parasite-specific CD4+ Th1 cell responses are the predominant immune effector for controlling malaria infection; however, the underlying regulatory mechanisms remain largely unknown. This study demonstrated that ATG5 deficiency in myeloid cells can significantly inhibit the growth of rodent blood-stage malarial parasites by selectively enhancing parasite-specific CD4+ Th1 cell responses. This effect was independent of ATG5-mediated canonical and non-canonical autophagy. Mechanistically, ATG5 deficiency suppressed FAS-mediated apoptosis of LY6G- ITGAM/CD11b+ ADGRE1/F4/80- cells and subsequently increased CCL2/MCP-1 production in parasite-infected mice. LY6G- ITGAM+ ADGRE1- cell-derived CCL2 selectively interacted with CCR2 on CD4+ Th1 cells for their optimized responses through the JAK2-STAT4 pathway. The administration of recombinant CCL2 significantly promoted parasite-specific CD4+ Th1 responses and suppressed malaria infection. Conclusively, our study highlights the previously unrecognized role of ATG5 in modulating myeloid cells apoptosis and sequentially affecting CCL2 production, which selectively promotes CD4+ Th1 cell responses. Our findings provide new insights into the development of immune interventions and effective anti-malarial vaccines.Abbreviations: ATG5: autophagy related 5; CBA: cytometric bead array; CCL2/MCP-1: C-C motif chemokine ligand 2; IgG: immunoglobulin G; IL6: interleukin 6; IL10: interleukin 10; IL12: interleukin 12; MFI: mean fluorescence intensity; JAK2: Janus kinase 2; LAP: LC3-associated phagocytosis; MAP1LC3/LC3: microtubule-associated protein 1 light chain 3; pRBCs: parasitized red blood cells; RUBCN: RUN domain and cysteine-rich domain containing, Beclin 1-interacting protein; STAT4: signal transducer and activator of transcription 4; Th1: T helper 1 cell; Tfh: follicular helper cell; ULK1: unc-51 like kinase 1.
Collapse
Affiliation(s)
- Yuanli Gao
- Department of Pathogenic Biology, Army Medical University (Third Military Medical University), Chongqing, China
| | - Suilin Chen
- Department of Pathogenic Biology, Army Medical University (Third Military Medical University), Chongqing, China
- Clinical Laboratory Diagnostic Center, General Hospital of Xinjiang Military Region, Urumqi, China
| | - Shiming Jiao
- Department of Pathogenic Biology, Army Medical University (Third Military Medical University), Chongqing, China
| | - Yongling Fan
- Department of Pathogenic Biology, Army Medical University (Third Military Medical University), Chongqing, China
| | - Xiuxiu Li
- Department of Pathogenic Biology, Army Medical University (Third Military Medical University), Chongqing, China
- The School of Medicine, Chongqing University, Chongqing, China
| | - Nie Tan
- Department of Pathogenic Biology, Army Medical University (Third Military Medical University), Chongqing, China
| | - Jiaqin Fang
- Department of Pathogenic Biology, Army Medical University (Third Military Medical University), Chongqing, China
| | - Luming Xu
- Provincial Key Laboratory of Immune Regulation and Immunotherapy, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Yi Huang
- Biomedical Analysis Center, Army Medical University (Third Military Medical University), Chongqing, China
| | - Jing Zhao
- Biomedical Analysis Center, Army Medical University (Third Military Medical University), Chongqing, China
| | - Shuai Guo
- Department of Pathogenic Biology, Army Medical University (Third Military Medical University), Chongqing, China
| | - Taiping Liu
- Department of Pathogenic Biology, Army Medical University (Third Military Medical University), Chongqing, China
- Key Laboratory of Extreme Environmental Medicine, Ministry of Education of China, Chongqing, China
| | - Wenyue Xu
- Department of Pathogenic Biology, Army Medical University (Third Military Medical University), Chongqing, China
- The School of Medicine, Chongqing University, Chongqing, China
- Key Laboratory of Extreme Environmental Medicine, Ministry of Education of China, Chongqing, China
| |
Collapse
|
98
|
Fischer F, Ermer MR, Howanski J, Yin Z, Bauer M, Wagner M, Fink B, Zenclussen AC, Schumacher A. Single and mixture effects of bisphenol A and benzophenone-3 on in vitro T helper cell differentiation. Chem Biol Interact 2024; 395:111011. [PMID: 38653352 DOI: 10.1016/j.cbi.2024.111011] [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: 01/23/2024] [Revised: 04/15/2024] [Accepted: 04/18/2024] [Indexed: 04/25/2024]
Abstract
Immune homeostasis is key to guarantee that the immune system can elicit effector functions against pathogens and at the same time raise tolerance towards other antigens. A disturbance of this delicate balance may underlie or at least trigger pathologies. Endocrine disrupting chemicals (EDCs) are increasingly recognized as risk factors for immune dysregulation. However, the immunotoxic potential of specific EDCs and their mixtures is still poorly understood. Thus, we aimed to investigate the effect of bisphenol A (BPA) and benzophenone-3 (BP-3), alone and in combination, on in vitro differentiation of T helper (TH)17 cells and regulatory T (Treg) cells. Naïve T cells were isolated from mouse lymphoid tissues and differentiated into the respective TH population in the presence of 0.001-10 μM BP-3 and/or 0.01-100 μM BPA. Cell viability, proliferation and the expression of TH lineage specific transcription factors and cytokines was measured by flow cytometry and CBA/ELISA. Moreover, the transcription of hormone receptors as direct targets of EDCs was quantified by RT-PCR. We found that the highest BPA concentration adversely affected TH cell viability and proliferation. Moreover, the general differentiation potential of both TH populations was not altered in the presence of both EDCs. However, EDC exposure modulated the emergence of TH17 and Treg cell intermediate states. While BPA and BP-3 promoted the development of TH1-like TH17 cells under TH17-differentiating conditions, TH2-like Treg cells occurred under Treg polarization. Interestingly, differential effects could be observed in mixtures of the two tested compounds compared with the individual compounds. Notably, estrogen receptor β expression was decreased under TH17-differentiating conditions in the presence of BPA and BP-3 as mixture. In conclusion, our study provides solid evidence for both, the immune disruptive potential and the existence of cumulative effects of real nature EDC mixtures on T cell in vitro differentiation.
Collapse
Affiliation(s)
- Florence Fischer
- Department of Environmental Immunology, Helmholtz Centre for Environmental Research GmbH - UFZ, Permoserstraße 15, 04318, Leipzig, Germany; Perinatal Immunology, Saxonian Incubator for Clinical Translation (SIKT), Medical Faculty, Leipzig University, Philipp-Rosenthal-Straße 55, 04103, Leipzig, Germany
| | - Miriam Rebecca Ermer
- Department of Environmental Immunology, Helmholtz Centre for Environmental Research GmbH - UFZ, Permoserstraße 15, 04318, Leipzig, Germany; Perinatal Immunology, Saxonian Incubator for Clinical Translation (SIKT), Medical Faculty, Leipzig University, Philipp-Rosenthal-Straße 55, 04103, Leipzig, Germany
| | - Julia Howanski
- Department of Environmental Immunology, Helmholtz Centre for Environmental Research GmbH - UFZ, Permoserstraße 15, 04318, Leipzig, Germany; Perinatal Immunology, Saxonian Incubator for Clinical Translation (SIKT), Medical Faculty, Leipzig University, Philipp-Rosenthal-Straße 55, 04103, Leipzig, Germany
| | - Ziran Yin
- Department of Environmental Immunology, Helmholtz Centre for Environmental Research GmbH - UFZ, Permoserstraße 15, 04318, Leipzig, Germany; Perinatal Immunology, Saxonian Incubator for Clinical Translation (SIKT), Medical Faculty, Leipzig University, Philipp-Rosenthal-Straße 55, 04103, Leipzig, Germany
| | - Mario Bauer
- Department of Environmental Immunology, Helmholtz Centre for Environmental Research GmbH - UFZ, Permoserstraße 15, 04318, Leipzig, Germany
| | - Marita Wagner
- Department of Environmental Immunology, Helmholtz Centre for Environmental Research GmbH - UFZ, Permoserstraße 15, 04318, Leipzig, Germany
| | - Beate Fink
- Department of Environmental Immunology, Helmholtz Centre for Environmental Research GmbH - UFZ, Permoserstraße 15, 04318, Leipzig, Germany
| | - Ana C Zenclussen
- Department of Environmental Immunology, Helmholtz Centre for Environmental Research GmbH - UFZ, Permoserstraße 15, 04318, Leipzig, Germany; Perinatal Immunology, Saxonian Incubator for Clinical Translation (SIKT), Medical Faculty, Leipzig University, Philipp-Rosenthal-Straße 55, 04103, Leipzig, Germany
| | - Anne Schumacher
- Department of Environmental Immunology, Helmholtz Centre for Environmental Research GmbH - UFZ, Permoserstraße 15, 04318, Leipzig, Germany; Perinatal Immunology, Saxonian Incubator for Clinical Translation (SIKT), Medical Faculty, Leipzig University, Philipp-Rosenthal-Straße 55, 04103, Leipzig, Germany.
| |
Collapse
|
99
|
Liu J, Li Y, Shen D, Li X, Wang K, Nagaoka K, Li C. Gut microbiota intervention alleviates pulmonary inflammation in broilers exposed to fine particulate matter from broiler house. Appl Environ Microbiol 2024; 90:e0217423. [PMID: 38656183 PMCID: PMC11107152 DOI: 10.1128/aem.02174-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 03/31/2024] [Indexed: 04/26/2024] Open
Abstract
The gut microbiota of poultry is influenced by a variety of factors, including feed, drinking water, airborne dust, and footpads, among others. Gut microbiota can affect the immune reaction and inflammation in the lungs. To investigate the effect of gut microbiota variation on lung inflammation induced by PM2.5 (fine particulate matter) in broilers, 36 Arbor Acres (AA) broilers were randomly assigned to three groups: control group (CON), PM2.5 exposure group (PM), and PM2.5 exposure plus oral antibiotics group (PMA). We used non-absorbable antibiotics (ABX: neomycin and amikacin) to modify the microbiota composition in the PMA group. The intervention was conducted from the 18th to the 28th day of age. Broilers in the PM and PMA groups were exposed to PM by a systemic exposure method from 21 to 28 days old, and the concentration of PM2.5 was controlled at 2 mg/m3. At 28 days old, the lung injury score, relative mRNA expression of inflammatory factors, T-cell differentiation, and dendritic cell function were significantly increased in the PM group compared to the CON group, and those of the PMA group were significantly decreased compared to the PM group. There were significant differences in both α and β diversity of cecal microbiota among these three groups. Numerous bacterial genera showed significant differences in relative abundance among the three groups. In conclusion, gut microbiota could affect PM2.5-induced lung inflammation in broilers by adjusting the capacity of antigen-presenting cells to activate T-cell differentiation. IMPORTANCE Gut microbes can influence the development of lung inflammation, and fine particulate matter collected from broiler houses can lead to lung inflammation in broilers. In this study, we explored the effect of gut microbes modified by intestinal non-absorbable antibiotics on particulate matter-induced lung inflammation. The results showed that modification in the composition of gut microbiota could alleviate lung inflammation by attenuating the ability of dendritic cells to stimulate T-cell differentiation, which provides a new way to protect lung health in poultry farms.
Collapse
Affiliation(s)
- Junze Liu
- Research Centre for Livestock Environmental Control and Smart Production, National Center for International Research on Animal Gut Nutrition, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Yuan Li
- Research Centre for Livestock Environmental Control and Smart Production, National Center for International Research on Animal Gut Nutrition, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Dan Shen
- Research Centre for Livestock Environmental Control and Smart Production, National Center for International Research on Animal Gut Nutrition, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Xiaoqing Li
- Research Centre for Livestock Environmental Control and Smart Production, National Center for International Research on Animal Gut Nutrition, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Kai Wang
- Research Centre for Livestock Environmental Control and Smart Production, National Center for International Research on Animal Gut Nutrition, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Kentaro Nagaoka
- Laboratory of Veterinary Physiology, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - Chunmei Li
- Research Centre for Livestock Environmental Control and Smart Production, National Center for International Research on Animal Gut Nutrition, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
100
|
Zhou Y, Chen B, Fu Y, Wan C, Li H, Wang L, Huang X, Wu Z, Li G, Xiong L, Qin D. Cang-ai volatile oil alleviates nasal inflammation via Th1/Th2 cell imbalance regulation in a rat model of ovalbumin-induced allergic rhinitis. Front Pharmacol 2024; 15:1332036. [PMID: 38835658 PMCID: PMC11148258 DOI: 10.3389/fphar.2024.1332036] [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: 11/02/2023] [Accepted: 04/23/2024] [Indexed: 06/06/2024] Open
Abstract
We previously revealed that Cang-ai volatile oil (CAVO) regulates T-cell activity, enhancing the immune response in people with chronic respiratory diseases. However, the effects of CAVO on allergic rhinitis (AR) have not been investigated. Herein, we established an ovalbumin (OVA)-induced AR rat model to determine these effects. Sprague-Dawley (SD) rats were exposed to OVA for 3 weeks. CAVO or loratadine (positive control) was given orally once daily for 2 weeks to OVA-exposed rats. Behavior modeling nasal allergies was observed. Nasal mucosa, serum, and spleen samples of AR rats were analyzed. CAVO treatment significantly reduced the number of nose rubs and sneezes, and ameliorated several hallmarks of nasal mucosa tissue remodeling: inflammation, eosinophilic infiltration, goblet cell metaplasia, and mast cell hyperplasia. CAVO administration markedly upregulated expressions of interferon-γ, interleukin (IL)-2, and IL-12, and downregulated expressions of serum tumor necrosis factor-α, IL-4, IL-5, IL-6, IL-13, immunoglobulin-E, and histamine. CAVO therapy also increased production of IFN-γ and T-helper type 1 (Th1)-specific T-box transcription factor (T-bet) of the cluster of differentiation-4+ T-cells in splenic lymphocytes, and protein and mRNA expressions of T-bet in nasal mucosa. In contrast, levels of the Th2 cytokine IL-4 and Th2-specific transcription factor GATA binding protein-3 were suppressed by CAVO. These cumulative findings demonstrate that CAVO therapy can alleviate AR by regulating the balance between Th1 and Th2 cells.
Collapse
Affiliation(s)
- Yang Zhou
- The First School of Clinical Medicine, Yunnan University of Chinese Medicine, Kunming, China
- Key Laboratory of Traditional Chinese Medicine for Prevention and Treatment of Neuropsychiatric Diseases, Yunnan University of Chinese Medicine, Kunming, China
| | - Bojun Chen
- Yunnan Provincial University Key Laboratory of Aromatic Chinese Herb Research, Kunming, China
- Yunnan Innovation Team of Application Research on Traditional Chinese Medicine Theory of Disease Prevention at Yunnan University of TCM, Kunming, China
| | - Yi Fu
- The Third Affiliated Hospital, Yunnan University of Chinese Medicine, Kunming, China
| | - Chunping Wan
- The First School of Clinical Medicine, Yunnan University of Chinese Medicine, Kunming, China
| | - Huayan Li
- The First School of Clinical Medicine, Yunnan University of Chinese Medicine, Kunming, China
| | - Lin Wang
- School of Pharmacy, Yunnan University of Chinese Medicine, Kunming, China
| | - Xiaoyi Huang
- Key Laboratory of Traditional Chinese Medicine for Prevention and Treatment of Neuropsychiatric Diseases, Yunnan University of Chinese Medicine, Kunming, China
| | - Zhao Wu
- School of Pharmacy, Yunnan University of Chinese Medicine, Kunming, China
| | - Gang Li
- Yunnan Provincial University Key Laboratory of Aromatic Chinese Herb Research, Kunming, China
- Yunnan Innovation Team of Application Research on Traditional Chinese Medicine Theory of Disease Prevention at Yunnan University of TCM, Kunming, China
| | - Lei Xiong
- Yunnan Provincial University Key Laboratory of Aromatic Chinese Herb Research, Kunming, China
- Yunnan Innovation Team of Application Research on Traditional Chinese Medicine Theory of Disease Prevention at Yunnan University of TCM, Kunming, China
| | - Dongdong Qin
- Key Laboratory of Traditional Chinese Medicine for Prevention and Treatment of Neuropsychiatric Diseases, Yunnan University of Chinese Medicine, Kunming, China
| |
Collapse
|