1
|
Alenezi FO, Nader MA, El-Kashef DH, Abdelmageed ME. Dapansutrile mitigates concanavalin A- induced autoimmune hepatitis: Involvement of NLRP3/IL-1β and JNK/ p38 MAPK pathways. Biomed Pharmacother 2025; 186:118026. [PMID: 40164046 DOI: 10.1016/j.biopha.2025.118026] [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: 01/06/2025] [Revised: 03/25/2025] [Accepted: 03/27/2025] [Indexed: 04/02/2025] Open
Abstract
AIM Dapansutrile (Dapan) is a newly developed anti-inflammatory molecule that supresses the production of NLRP3 inflammasome-dependent IL-1β. Its hepatoprotective effects against autoimmune hepatitis (AIH) have not yet been explored. Hence, this study was conducted to examine the possible protective effects of Dapan against concanavalin A (Con A)-induced hepatitis in mice. MAIN METHODS Mice were randomly divided into five groups (n = 6): control, Con A (15 mg/kg), Dapan (60 mg/kg), Dapan (6 mg/kg) + Con A, and Dapan (60 mg/kg) + Con A. Mice were euthanised at the end of the study, and blood and hepatic tissues were collected. KEY FINDINGS Hepatic function testing using lactate dehydrogenase, alanine aminotransferase, and aspartate aminotransferase levels, in addition to hepatic tissue histological examination, revealed that intraperitoneal administration of Dapan noticeably ameliorated Con A-induced hepatic enzyme impairment and histopathological disruption. Moreover, Dapan-treated mice had significantly lower malondialdehyde hepatic content and elevated reduced glutathione, superoxide dismutase, and total antioxidant capacity levels than non-treated mice in a dose-dependent manner. The Dapan-treated groups showed significantly lower levels of the inflammatory mediators, NLRP3, TNF-α, IL-6, and IL-1β, in addition to the immunomodulators CD8, CD4, INF-γ, and NFκB and inhibition of JNK and p38 MAPK levels compared to the Con A-treated group. SIGNIFICANCE Our results showed that intraperitoneal administration of Dapan could be a therapeutic opportunity to inhibit the development of AIH via inhibition of inflammatory pathways.
Collapse
Affiliation(s)
- Fahad O Alenezi
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt; Forensic Toxicology Services Center, Ministry of health, Qassim, Saudi Arabia
| | - Manar A Nader
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Dalia H El-Kashef
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Marwa E Abdelmageed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt.
| |
Collapse
|
2
|
Paton H, Sarkar P, Gurung P. An overview of host immune responses against Leishmania spp. infections. Hum Mol Genet 2025:ddaf043. [PMID: 40287829 DOI: 10.1093/hmg/ddaf043] [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: 12/25/2024] [Revised: 03/18/2025] [Accepted: 03/24/2025] [Indexed: 04/29/2025] Open
Abstract
Leishmania spp. infections pose a significant global health challenge, affecting approximately 1 billion people across more than 88 endemic countries. This unicellular, obligate intracellular parasite causes a spectrum of diseases, ranging from localized cutaneous lesions to systemic visceral infections. Despite advancements in modern medicine and increased understanding of the parasite's etiology and associated diseases, treatment options remain limited to pentavalent antimonials, liposomal amphotericin B, and miltefosine. A deeper understanding of the interactions between immune and non-immune cells involved in the clearance of Leishmania spp. infections could uncover novel therapeutic strategies for this debilitating disease. This review highlights recent progress in elucidating how various cell types contribute to the regulation and resolution of Leishmania spp. infections.
Collapse
Affiliation(s)
- Hanna Paton
- Inflammation Program, University of Iowa, 431 Newton Road, Iowa City, IA 52242, United States
- Department of Internal Medicine, University of Iowa, 431 Newton Road, Iowa City, IA 52442, United States
- Immunology Graduate Program, University of Iowa, 431 Newton Road, Iowa City, IA 52242, United States
| | - Prabuddha Sarkar
- Inflammation Program, University of Iowa, 431 Newton Road, Iowa City, IA 52242, United States
- Department of Internal Medicine, University of Iowa, 431 Newton Road, Iowa City, IA 52442, United States
| | - Prajwal Gurung
- Inflammation Program, University of Iowa, 431 Newton Road, Iowa City, IA 52242, United States
- Department of Internal Medicine, University of Iowa, 431 Newton Road, Iowa City, IA 52442, United States
- Immunology Graduate Program, University of Iowa, 431 Newton Road, Iowa City, IA 52242, United States
- Interdisciplinary Graduate Program in Human Toxicology, University of Iowa, 431 Newton Road, Iowa City, IA 52242, United States
- Center for Immunology and Immune Based Disease, University of Iowa, 431 Newton Road, Iowa City, IA 52242, United States
- Iowa City Veterans Affairs (VA) Medical Center, 601 US-6, Iowa City, IA 52246, United States
| |
Collapse
|
3
|
Barthels DA, House RV, Gelhaus HC. The immune response to Francisella tularensis. Front Microbiol 2025; 16:1549343. [PMID: 40351308 PMCID: PMC12062900 DOI: 10.3389/fmicb.2025.1549343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 03/24/2025] [Indexed: 05/14/2025] Open
Abstract
Francisella tularensis (Ft) is a Gram negative intracellular bacterial pathogen, commonly transmitted via arthropod bites, but is most lethal when contracted via inhalation. The nature of a Gram-negative intracellular pathogen presents unique challenges to the mammalian immune response, unlike more common viral pathogens and extracellular bacterial pathogens. The current literature on Ft involves numerous variables, including the use of differing research strains and variation in animal models. This review aims to consolidate much of the recent literature on Ft to suggest promising research to better understand the complex immune response to this bacterium.
Collapse
Affiliation(s)
- Derek A. Barthels
- Department of Biology, Life Sciences Research Center, United States Air Force Academy, Colorado Springs, CO, United States
- National Research Council Research Associateships Program, Washington, DC, United States
| | - Robert V. House
- Dr. RV House LLC, Harpers Ferry, WV, United States
- Appili Therapeutics, Halifax, NS, Canada
| | | |
Collapse
|
4
|
Lim YJ, Duckworth AD, Clarke K, Kennedy P, Karpha I, Oates M, Gornall M, Kalakonda N, Slupsky JR, Pettitt AR. Influence of polyfunctional Tbet + T cells on specific clinical events in chronic lymphocytic leukaemia. Front Immunol 2025; 16:1528405. [PMID: 40313965 PMCID: PMC12043603 DOI: 10.3389/fimmu.2025.1528405] [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: 11/14/2024] [Accepted: 03/18/2025] [Indexed: 05/03/2025] Open
Abstract
Introduction T-cell dysfunction is a hallmark of chronic lymphocytic leukemia (CLL), but the extent to which individual CD4+ or CD8+ T-cell subpopulations influence specific clinical events remains unclear. To address this knowledge gap, we utilised high-dimensional mass cytometry to profile circulating CD4+ and CD8+ T-cells in pre-treatment samples from a well-defined cohort of CLL patients undergoing initial therapy as part of a clinical trial. Methods Pre-treatment blood samples from 138 CLL patients receiving initial chemoimmunotherapy containing bendamustine or chlorambucil in the NCRI RIAltO trial (NCT01678430; EudraCT 2011-000919-22) were subjected to deep immunophenotyping by mass cytometry using a bespoke panel of 37 antibodies. T-cell clusters were identified through unsupervised clustering and related to treatment outcomes. Additionally, a randomly selected cohort of 30 CLL patients underwent T-cell stimulation with anti-CD3/CD28 microbeads, followed by cytokine analysis using a separate 36-antibody panel, which included seven cytokines. Results Seventeen CD4+ and 22 CD8+ T-cell clusters were identified in a discovery cohort of 79 patients. Three of these clusters, measured as a proportion of their parental CD4+ or CD8+ populations, correlated with a reduced risk of grade ≥3 infection, grade ≥3 second primary malignancy (SPM) and death, respectively. Three corresponding T-cell subpopulations prospectively defined by non-redundant markers and Boolean gating (ICOS+HLA-DR+PD1+TIGIT+Tbet+CD4+ T-helper cells; CD27+CD28-PD1+Tbet+Eomes+CD8+ cells; and CD27+CD28-GrymB+Tbet+Eomes+CD8+ terminal effector cells) showed the same clinical correlations as the clusters on which they were based. With the exception of SPM for which there were insufficient events, these correlations were confirmed in a separate validation cohort of 59 patients. In-vitro stimulation of a subset of CLL patients in the discovery cohort showed an enrichment of primed and polyfunctional cells in all three Tbet+ T-cell subpopulations of interest. Conclusion Our study provides new insights into the potential for Tbet+ T-cell subpopulations to influence and predict specific clinical events in CLL. This, in turn, raises the possibility that these respective subpopulations could play an important role in controlling infection, solid tumours and CLL itself. Clinical Trial Registration https://www.clinicaltrials.gov/, identifier NCT01678430; https://www.isrctn.com/ISRCTN09988575, identifier EudraCT 2011-000919-22.
Collapse
Affiliation(s)
- Yeong Jer Lim
- Department of Molecular & Clinical Cancer Medicine, University of Liverpool, Liverpool, United Kingdom
- Haemato-oncology Department, The Clatterbridge Cancer Centre National Health Service (NHS) Foundation Trust, Liverpool, United Kingdom
| | - Andrew D. Duckworth
- Department of Molecular & Clinical Cancer Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Kim Clarke
- Computational Biology Facility, University of Liverpool, Liverpool, United Kingdom
| | - Paul Kennedy
- Department of Pharmacology & Therapeutics, University of Liverpool, Liverpool, United Kingdom
| | - Indrani Karpha
- Department of Molecular & Clinical Cancer Medicine, University of Liverpool, Liverpool, United Kingdom
- Haemato-oncology Department, The Clatterbridge Cancer Centre National Health Service (NHS) Foundation Trust, Liverpool, United Kingdom
| | - Melanie Oates
- Department of Molecular & Clinical Cancer Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Matthew Gornall
- Liverpool Clinical Trials Centre, University of Liverpool, Liverpool, United Kingdom
| | - Nagesh Kalakonda
- Department of Molecular & Clinical Cancer Medicine, University of Liverpool, Liverpool, United Kingdom
- Haemato-oncology Department, The Clatterbridge Cancer Centre National Health Service (NHS) Foundation Trust, Liverpool, United Kingdom
| | - Joseph R. Slupsky
- Department of Molecular & Clinical Cancer Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Andrew R. Pettitt
- Department of Molecular & Clinical Cancer Medicine, University of Liverpool, Liverpool, United Kingdom
- Haemato-oncology Department, The Clatterbridge Cancer Centre National Health Service (NHS) Foundation Trust, Liverpool, United Kingdom
| |
Collapse
|
5
|
Lee S, Dohlman TH, Dana R. Immunology in corneal transplantation-From homeostasis to graft rejection. Transplant Rev (Orlando) 2025; 39:100909. [PMID: 39798206 PMCID: PMC11975484 DOI: 10.1016/j.trre.2025.100909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Revised: 12/19/2024] [Accepted: 01/08/2025] [Indexed: 01/15/2025]
Abstract
Immunology depends on maintaining a delicate balance within the human body, and disruptions can result in conditions such as autoimmune diseases, immunodeficiencies, and hypersensitivity reactions. This balance is especially crucial in transplantation immunology, where one of the primary challenges is preventing graft rejection. Such rejection can lead to organ failure, increased patient mortality, and higher healthcare costs due to the limited availability of donor tissues relative to patient needs. Xenotransplantation, like using porcine corneas for human transplants, offers a potential solution to the donor tissue shortage but faces substantial immunological rejection issues. To prevent rejection in both allo- and xenotransplantation, a deep understanding of how the body maintains immunological balance is essential, particularly since achieving tolerance to non-self tissues is considered the "holy grail" of the field. The cornea, the most frequently transplanted solid organ, has a high acceptance rate due to its immune-privileged status and serves as an ideal model for studying graft rejection mechanisms that disrupt tolerance. However, multiple immune pathways complicate our understanding of these mechanisms. This review examines the rejection mechanisms in corneal transplantation, identifying key cells involved and potential therapeutic strategies to induce and maintain immunological tolerance in both allo- and xenografts across various transplants.
Collapse
Affiliation(s)
- Seokjoo Lee
- Laboratory of Ocular Immunology, Transplantation, and Regeneration, Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Thomas H Dohlman
- Laboratory of Ocular Immunology, Transplantation, and Regeneration, Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Reza Dana
- Laboratory of Ocular Immunology, Transplantation, and Regeneration, Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
6
|
Jablonka-Shariff A, Broberg C, Snyder-Warwick AK. Absence of T-box transcription factor 21 limits neuromuscular junction recovery after nerve injury in T-bet-knockout mice. Front Cell Dev Biol 2025; 13:1535323. [PMID: 40162097 PMCID: PMC11949913 DOI: 10.3389/fcell.2025.1535323] [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: 11/27/2024] [Accepted: 02/20/2025] [Indexed: 04/02/2025] Open
Abstract
Introduction Terminal Schwann cells (tSCs), at the neuromuscular junction (NMJ), play critical roles in the repair of motor axon terminals at muscle, and rebuild neuronal signaling following nerve injury. Knowledge of mediators impacting tSCs post-nerve injury and in disease may guide beneficial therapies to improve motor outcomes. We previously found T-box transcription factor 21 (TBX21/TBET), classically associated with T-helper1 cells and immune cell recruitment, is expressed in tSCs at the mouse NMJ. The purpose of this study was to examine effects of Tbx21 absence during NMJ regeneration following peripheral nerve injury. Methods Wildtype (WT) and Tbet-knockout (Tbet-KO) mice underwent sciatic nerve transection and immediate repair. Functional muscle recovery assessment was performed with muscle force testing on mice at 2-, 3-, 4-, and 6-week (wks) and 6 months after nerve injury repair. Morphometric analyses of NMJ reinnervation, tSC number, and tSC processes were evaluated. Full NMJ reinnervation was defined as ≥75% coverage of endplates by axons. A minimum of three mice were evaluated in each group, and 50-100 NMJs were evaluated per mouse. Results Tbet-KO mice had significantly diminished muscle function compared to WT mice at every time point beyond 3 weeks. Tbet-KO mice showed just over half of the muscle force generated by WT mice at 4 weeks and 6 weeks post-injury and repair. By 6 months, Tbet-KO mice generated only 84.1% the muscle force of WT mice. Tbet-KO mice showed significantly decreased levels of fully reinnervated NMJs compared to WT mice at each time point tested. Tbet-KO mice also showed a lower number of tSCs with reduced cytoplasmic processes beyond NMJ area and lower number of immune cells during process of NMJ regeneration. Discussion Our findings show that the Tbx21 transcription factor promotes NMJ reinnervation to regain muscle function following nerve injury.
Collapse
Affiliation(s)
- Albina Jablonka-Shariff
- Research Scientist, Division of Plastic Surgery, Department of Surgery, Washington University School of Medicine, St. Louis, MO, United States
| | - Curtis Broberg
- Research Student, Washington University School of Medicine, St. Louis, MO, United States
| | - Alison K. Snyder-Warwick
- Division of Plastic Surgery, Department of Surgery, Washington University School of Medicine, St. Louis, MO, United States
| |
Collapse
|
7
|
Deng F, Xiao G, Tanzhu G, Chu X, Ning J, Lu R, Chen L, Zhang Z, Zhou R. Predicting Survival Rates in Brain Metastases Patients from Non-Small Cell Lung Cancer Using Radiomic Signatures Associated with Tumor Immune Heterogeneity. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2412590. [PMID: 39840456 PMCID: PMC11904944 DOI: 10.1002/advs.202412590] [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: 10/08/2024] [Revised: 12/17/2024] [Indexed: 01/23/2025]
Abstract
Non-small cell lung cancer (NSCLC) frequently metastasizes to the brain, significantly worsened prognoses. This study aimed to develop an interpretable model for predicting survival in NSCLC patients with brain metastases (BM) integrating radiomic features and RNA sequencing data. 292 samples are collected and analyzed utilizing T1/T2 MRIs. Bidirectional stepwise logistic regression is employed to identify significant variables, facilitating the construction of a prognostic model, which is benchmarked against four machine learning algorithms. BM tissue samples are processed for RNA extraction and sequencing. The optimal model achieved an AUC of 0.96 and a C-index of 0.89 in the train set and an AUC of 0.84 with a C-index of 0.78 in the test set, indicating strong predictive performance and generalizability. Patients from Xiangya Hospital are stratified into high-risk (n = 11) and low-risk (n = 30) groups. RNA sequencing revealed an enrichment of immune-related pathways, particularly the interferon (IFN) pathway in the low-risk group. Immune cell infiltration analysis identified a significant presence of CD8+-T cells, IFNγ-6/-18 in the low-risk group, suggesting an immunologically favorable tumor microenvironment. These findings highlight the potential of combining radiomic and RNA sequencing data for improved survival predictions and personalized treatment strategies in BM patients from NSCLC.
Collapse
Affiliation(s)
- Fuxing Deng
- The department of oncologyXiangya HospitalCentral South UniversityChangsha410008China
| | - Gang Xiao
- The department of oncologyXiangya HospitalCentral South UniversityChangsha410008China
| | - Guilong Tanzhu
- The department of oncologyXiangya HospitalCentral South UniversityChangsha410008China
| | - Xianjing Chu
- The department of oncologyXiangya HospitalCentral South UniversityChangsha410008China
| | - Jiaoyang Ning
- The department of oncologyXiangya HospitalCentral South UniversityChangsha410008China
| | - Ruoyu Lu
- The department of oncologyXiangya HospitalCentral South UniversityChangsha410008China
| | - Liu Chen
- The department of oncologyXiangya HospitalCentral South UniversityChangsha410008China
| | - Zijian Zhang
- The department of oncologyXiangya HospitalCentral South UniversityChangsha410008China
| | - Rongrong Zhou
- The department of oncologyXiangya HospitalCentral South UniversityChangsha410008China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangsha410008China
- Xiangya Lung Cancer CenterXiangya HospitalCentral South UniversityChangsha410008China
| |
Collapse
|
8
|
Xin L, Xu HS, Fan LJ, Liu C, Zou YH, Zhou Q, Yue ZQ, Gan JH, Liu J. Methionine Restriction Exerts Anti-Tumor Immunity via Joint Intervention of T-Bet Palmitoylation in Gastric Cancer. Biotechnol J 2025; 20:e202400574. [PMID: 39989253 DOI: 10.1002/biot.202400574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Revised: 12/01/2024] [Accepted: 02/04/2025] [Indexed: 02/25/2025]
Abstract
BACKGROUND Methionine restriction (MR) exerts an anti-tumor immunomodulatory role. Th1 cells facilitate CD8+ cytotoxic T cell activation and targeted tumor cell killing. Our previous work shows that MR enhances the immunotherapy effect of PD-L1/PD-1 blockade on gastric cancer, MR can simultaneously inhibit Th1 cell differentiation, which may affect their synergistic therapeutic outcome. We aim to elucidate the molecular mechanism of MR regulating Th1 cell activation in gastric cancer. METHODS Murine Foregastric Carcinoma (MFC) cells were injected into 615 mice to establish transplanted tumor models, which were then treated with an MR diet or combined with 2-bromopalmitate (2-BP). CD4+T cells were cultured with deficient methionine. The acyl-biotinyl exchange (ABE) method was to detect T-bet palmitoylation and cycloheximide experiments to detect protein stability. GPS-Palm tool was employed to screen palmitoyltransferases. The impact of T-bet palmitoylation on the pro-tumor-killing effect of Th1 cells was examined. RESULTS MR enhanced anti-PD-1's inhibition of tumor growth, while concurrently suppressing the increased Th1 cells. Combined with 2-BP further inhibited tumor and increased Th1 cells. Suppressing Th1 activity attenuated 2-BP's synergistic therapeutic effect and reduced CD8+ GZMB+ T cells. MR inhibited Th1 differentiation by reducing T-bet expression, 2-BP treatment restored, while T-bet interference reversed 2-BP's effect. MR increased palmitoylation and T-bet underwent palmitoylation modification. ZDHHC23 mediated T-bet palmitoylation and promoted T-bet degradation. MR promoted T-bet degradation, thereby decreasing T-bet content, inhibiting Th1 cell polarization and CD8+ T cell killing effect. CONCLUSIONS MR combined with T-bet palmitoylation intervention promotes Th1 polarization and CD8+ T cell toxicity, thereby enhancing anti-tumor immunity in gastric cancer.
Collapse
Affiliation(s)
- Lin Xin
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
| | - He-Song Xu
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
| | - Luo-Jun Fan
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
| | - Chuan Liu
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
| | - Yong-Hui Zou
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
| | - Qi Zhou
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
| | - Zhen-Qi Yue
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
| | - Jin-Heng Gan
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
| | - Jiang Liu
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
| |
Collapse
|
9
|
Mi R, Li X, Li Y, Du X. Anti-oxidation enhancement, inflammation alleviation, and microbial composition optimization of using tussah (Antheraea pernyi) silk fibroin peptides for hyperglycaemia remission. PLoS One 2025; 20:e0317891. [PMID: 39847549 PMCID: PMC11756761 DOI: 10.1371/journal.pone.0317891] [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: 07/07/2024] [Accepted: 12/17/2024] [Indexed: 01/25/2025] Open
Abstract
OBJECTIVE This study aimed to evaluate the positive effects on anti-oxidation, anti-inflammation, and microbial composition optimization of diabetic mice using tussah (Antheraea pernyi) silk fibroin peptides (TSFP), providing the theoretical foundation for making the use of silk resources of A. pernyi and incorporating as a supplement into the hypoglycemic foods. METHOD The animal model of diabetes was established successfully. Alloxan-induced diabetic mice were orally administered using TSFP, and the hypoglycaemic effects in vivo were systematically investigated. RESULTS The results indicated that TSFP could significantly reduce the fasting blood glucose (FBG) levels and suppress the mRNA expression of glycometabolism genes of diabetic mice. In addition, the TSFP could ameliorate the lipid dysbolism and contribute to a higher anti-oxidation capacity. Moreover, TSFP could alleviate pathological damages and hinder inflammatory processes of diabetic mice. Besides, the supplementation of TSFP presented a greater ability to shape and optimize the gut microbial composition by enriching the profitable bacteria and inhibiting the pathogenic microorganisms. Correlation analysis also revealed that the abundances of functional bacteria in the TSFP-treated groups exhibited better correlations with serum parameters, which would be of positive significance for blood glucose regulation and inflammation remission. CONCLUSIONS These results collectively corroborated the feasibility and superiority of using TSFP for hyperglycaemia remission via anti-oxidation enhancement, inflammation alleviation, and microbial composition optimization, contributing to a safely feasible and biologically efficient strategy for improving anti-diabetic effects.
Collapse
Affiliation(s)
- Rui Mi
- Liaoning Ocean and Fisheries Science Research Institute, Liaoning Academy of Agricultural Sciences, Dalian, PR China
| | - Xuejun Li
- Liaoning Ocean and Fisheries Science Research Institute, Liaoning Academy of Agricultural Sciences, Dalian, PR China
| | - Yajie Li
- Liaoning Ocean and Fisheries Science Research Institute, Liaoning Academy of Agricultural Sciences, Dalian, PR China
| | - Xingfan Du
- Liaoning Ocean and Fisheries Science Research Institute, Liaoning Academy of Agricultural Sciences, Dalian, PR China
| |
Collapse
|
10
|
Li Q, Marcoux G, Hu Y, Rebetz J, Guo L, Semple E, Provan D, Xu S, Hou M, Peng J, Semple JW. Autoimmune effector mechanisms associated with a defective immunosuppressive axis in immune thrombocytopenia (ITP). Autoimmun Rev 2024; 23:103677. [PMID: 39515406 DOI: 10.1016/j.autrev.2024.103677] [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: 10/01/2024] [Revised: 10/29/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024]
Abstract
Immune thrombocytopenia (ITP) is an autoimmune disease characterized by an isolated thrombocytopenia and variable phenotype as some patients suffer no bleeding whilst others have bleeding from mild to severe, which may be fatal. This variability probably reflects the disease's complex pathophysiology; a dysregulated hyperreactive immune effector cell response involving the entire adaptive immune system (e.g. B and T cell subsets) that leads to platelet and megakaryocyte (MK) destruction. It appears that these effector responses are due to a breakdown in immune tolerance, and this is characterized by defects in several immunosuppressive cell types. These include defective T regulatory cells (Tregs), B regulatory cells (Bregs) and Myeloid-derived suppressor cells (MDSC), all of which are all intimately associated with antigen presenting cells (APC) such as dendritic cells (DC). The loss of this immunosuppressive axis allows for the activation of unchecked autoreactive T cells and B cells, leading to the development of autoantibodies and cytotoxic T cells (CTL), which can directly destroy platelets in the periphery and inhibit MK platelet production in the bone marrow (BM). This review will focus on the effector cell mechanisms in ITP and highlight the defective immunosuppressive axis that appears responsible for this platelet-specific immune hyperreactivity.
Collapse
Affiliation(s)
- Qizhao Li
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Geneviève Marcoux
- Division of Hematology and Transfusion Medicine, Lund University, Lund, Sweden
| | - Yuefen Hu
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Johan Rebetz
- Division of Hematology and Transfusion Medicine, Lund University, Lund, Sweden
| | - Li Guo
- Bloodworks Northwest Research Institute, Seattle, USA; Division of Hematology and Oncology, University of Washington; Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, USA
| | | | - Drew Provan
- Department of Haematology, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, UK
| | - Shuqian Xu
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, Shandong, China.
| | - Ming Hou
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Jun Peng
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - John W Semple
- Division of Hematology and Transfusion Medicine, Lund University, Lund, Sweden; Clinical Immunology and Transfusion Medicine, Office of Medical Services, Region Skåne, Lund, Sweden; Departments of Pharmacology, Medicine and Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada.
| |
Collapse
|
11
|
Wang M, Deng B, Jiang T, Duolikun A, Li Y, Ainiwaer A, Kang X, Zheng X, Rousu Z, Yu Q, Li J, Wang H, Zhang C, Aji T, Shao Y. Upregulation of CD244 promotes CD8 + T cell exhaustion in patients with alveolar echinococcosis and a murine model. Parasit Vectors 2024; 17:483. [PMID: 39578914 PMCID: PMC11585139 DOI: 10.1186/s13071-024-06573-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 11/07/2024] [Indexed: 11/24/2024] Open
Abstract
BACKGROUND In patients with alveolar echinococcosis (AE), CD8+ T cells undergo functional exhaustion, which accelerates the malignant progression of AE. However, the role of inhibitory receptor CD244 in mediating CD8+ T cell exhaustion remains elusive. METHODS CD244 expression on exhausted CD8+ T cells in the close liver tissue (CLT) of AE patients was analyzed using single-cell RNA sequencing data. Immunohistochemistry and immunofluorescence were employed to detect CD244 expression. Flow cytometry was used to assess the impact of CD244 on differentiation and effector function of CD8+ T cells in patients with AE, in vitro and in vivo models. Reactive oxygen species (ROS) and oxygen consumption rate (OCR) were measured to evaluate the influence of CD244 on mitochondrial function of CD8+ T cells. RESULTS CD244+CD8+ T cells in the CLT of AE patients exhibit a more terminal differentiation phenotype, with reduced secretion of IFN-γ and TNF-α. In vitro studies revealed that CD8+ T cells from CD244-deficient mice produced higher levels of IFN-γ, TNF-α and Granzyme B. In vivo studies revealed that CD244 deficiency enhanced the secretion capacity of IFN-γ and TNF-α by CD8+ T cells, inhibiting the growth of metacestodes. Moreover, CD244 deficiency leads to a decrease in ROS levels in liver CD8+ T cells, while significantly increasing their adenosine triphosphate (ATP)-linked oxygen consumption rate. CONCLUSIONS CD244 facilitates AE disease progression by mediating immune exhaustion in CD8+ T cells.
Collapse
Affiliation(s)
- Maolin Wang
- Department of Hepatobiliary and Echinococcosis Surgery, Digestive and Vascular Surgery Center, First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, China
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Xinjiang Medical University, Clinical Medicine Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, China
- Clinical Medical Research Center of Echinococcosis and Hepatobiliary Disease of Xinjiang Uygur Autonomous Region, Urumqi, 830054, China
| | - Bingqing Deng
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Xinjiang Medical University, Clinical Medicine Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, China
- Basic Medical College, Xinjiang Medical University, Urumqi, 830017, China
| | - Tiemin Jiang
- Department of Hepatobiliary and Echinococcosis Surgery, Digestive and Vascular Surgery Center, First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, China
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Xinjiang Medical University, Clinical Medicine Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, China
- Clinical Medical Research Center of Echinococcosis and Hepatobiliary Disease of Xinjiang Uygur Autonomous Region, Urumqi, 830054, China
| | - Adilai Duolikun
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Xinjiang Medical University, Clinical Medicine Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, China
- Basic Medical College, Xinjiang Medical University, Urumqi, 830017, China
| | - Yinshi Li
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Xinjiang Medical University, Clinical Medicine Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, China
- Basic Medical College, Xinjiang Medical University, Urumqi, 830017, China
| | - Abidan Ainiwaer
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Xinjiang Medical University, Clinical Medicine Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, China
- Basic Medical College, Xinjiang Medical University, Urumqi, 830017, China
| | - Xuejiao Kang
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Xinjiang Medical University, Clinical Medicine Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, China
- Basic Medical College, Xinjiang Medical University, Urumqi, 830017, China
| | - Xuran Zheng
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Xinjiang Medical University, Clinical Medicine Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, China
- Basic Medical College, Xinjiang Medical University, Urumqi, 830017, China
| | - Zibigu Rousu
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Xinjiang Medical University, Clinical Medicine Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, China
- Basic Medical College, Xinjiang Medical University, Urumqi, 830017, China
| | - Qian Yu
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Xinjiang Medical University, Clinical Medicine Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, China
- Basic Medical College, Xinjiang Medical University, Urumqi, 830017, China
| | - Jing Li
- Basic Medical College, Xinjiang Medical University, Urumqi, 830017, China
| | - Hui Wang
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Xinjiang Medical University, Clinical Medicine Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, China
- Basic Medical College, Xinjiang Medical University, Urumqi, 830017, China
| | - Chuanshan Zhang
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Xinjiang Medical University, Clinical Medicine Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, China.
- Basic Medical College, Xinjiang Medical University, Urumqi, 830017, China.
| | - Tuerganaili Aji
- Department of Hepatobiliary and Echinococcosis Surgery, Digestive and Vascular Surgery Center, First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, China.
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Xinjiang Medical University, Clinical Medicine Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, China.
- Clinical Medical Research Center of Echinococcosis and Hepatobiliary Disease of Xinjiang Uygur Autonomous Region, Urumqi, 830054, China.
| | - Yingmei Shao
- Department of Hepatobiliary and Echinococcosis Surgery, Digestive and Vascular Surgery Center, First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, China.
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Xinjiang Medical University, Clinical Medicine Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, China.
- Clinical Medical Research Center of Echinococcosis and Hepatobiliary Disease of Xinjiang Uygur Autonomous Region, Urumqi, 830054, China.
| |
Collapse
|
12
|
Hwang AS, Kechter JA, Do TH, Hughes AN, Zhang N, Li X, Bogle R, Brumfiel CM, Patel MH, Boudreaux B, Bhullar P, Nassir S, Yousif ML, Stockard AL, Leibovit-Reiben Z, Ogbaudu E, DiCaudo DJ, Fox J, Gharaee-Kermani M, Xing X, Zunich S, Branch E, Kahlenberg JM, Billi AC, Plazyo O, Tsoi LC, Pittelkow MR, Gudjonsson JE, Mangold AR. Rapid response of lichen planus to baricitinib associated with suppression of cytotoxic CXCL13+CD8+ T cells. J Clin Invest 2024; 135:e179436. [PMID: 39541169 PMCID: PMC11735091 DOI: 10.1172/jci179436] [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: 01/22/2024] [Accepted: 11/11/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUNDCutaneous lichen planus (LP) is a recalcitrant, difficult-to-treat, inflammatory skin disease characterized by pruritic, flat-topped, violaceous papules on the skin. Baricitinib is an oral Janus kinase (JAK) 1/2 inhibitor that interrupts the signaling pathway of IFN-γ, a cytokine implicated in the pathogenesis of LP.METHODSIn this phase II trial, 12 patients with cutaneous LP received 2 mg daily baricitinib for 16 weeks, accompanied by in-depth spatial, single-cell, and bulk transcriptomic profiling of pre- and posttreatment samples.RESULTSAn early and sustained clinical response was seen, with 83.3% of patients responsive at week 16. Our molecular data identified a unique, oligoclonal IFN-γ, CD8+, and CXCL13+ cytotoxic T cell population in LP skin and demonstrated a rapid decrease in IFN signature within 2 weeks of treatment, most prominently in the basal layer of the epidermis.CONCLUSIONThis study demonstrates the efficacy and molecular mechanisms of JAK inhibition in LP.TRIAL REGISTRATIONNCT05188521FUNDINGEli Lilly, Appignani Benefactor Funds, 5P30AR075043, Mayo Clinic Clinical Trials Stimulus Funds.
Collapse
Affiliation(s)
- Angelina S. Hwang
- Mayo Clinic Arizona Department of Dermatology, Scottsdale, Arizona, USA
| | - Jacob A. Kechter
- Mayo Clinic Arizona Department of Dermatology, Scottsdale, Arizona, USA
| | - Tran H. Do
- University of Michigan Department of Dermatology, Ann Arbor, Michigan, USA
| | - Alysia N. Hughes
- Mayo Clinic Arizona Department of Dermatology, Scottsdale, Arizona, USA
| | - Nan Zhang
- Mayo Clinic Department of Quantitative Health Sciences, Scottsdale, Arizona, USA
| | - Xing Li
- Mayo Clinic Arizona Department of Dermatology, Scottsdale, Arizona, USA
| | - Rachael Bogle
- University of Michigan Department of Dermatology, Ann Arbor, Michigan, USA
| | | | - Meera H. Patel
- Mayo Clinic Arizona Department of Dermatology, Scottsdale, Arizona, USA
| | - Blake Boudreaux
- Mayo Clinic Arizona Department of Dermatology, Scottsdale, Arizona, USA
| | - Puneet Bhullar
- Mayo Clinic Arizona Department of Dermatology, Scottsdale, Arizona, USA
| | - Shams Nassir
- Mayo Clinic Arizona Department of Dermatology, Scottsdale, Arizona, USA
| | - Miranda L. Yousif
- Mayo Clinic Arizona Department of Dermatology, Scottsdale, Arizona, USA
| | | | | | - Ewoma Ogbaudu
- Mayo Clinic Arizona Department of Dermatology, Scottsdale, Arizona, USA
| | - David J. DiCaudo
- Mayo Clinic Arizona Department of Dermatology, Scottsdale, Arizona, USA
- Mayo Clinic Department of Laboratory Medicine and Pathology, Scottsdale, Arizona, USA
| | - Jennifer Fox
- University of Michigan Department of Dermatology, Ann Arbor, Michigan, USA
| | - Mehrnaz Gharaee-Kermani
- University of Michigan Department of Dermatology, Ann Arbor, Michigan, USA
- University of Michigan Department of Internal Medicine, Rheumatology, Ann Arbor, Michigan, USA
| | - Xianying Xing
- University of Michigan Department of Dermatology, Ann Arbor, Michigan, USA
| | - Samantha Zunich
- Mayo Clinic Arizona Department of Dermatology, Scottsdale, Arizona, USA
| | - Emily Branch
- Mayo Clinic Arizona Department of Dermatology, Scottsdale, Arizona, USA
| | - J. Michelle Kahlenberg
- University of Michigan Department of Dermatology, Ann Arbor, Michigan, USA
- University of Michigan Department of Internal Medicine, Rheumatology, Ann Arbor, Michigan, USA
| | - Allison C. Billi
- University of Michigan Department of Dermatology, Ann Arbor, Michigan, USA
| | - Olesya Plazyo
- University of Michigan Department of Dermatology, Ann Arbor, Michigan, USA
| | - Lam C. Tsoi
- University of Michigan Department of Dermatology, Ann Arbor, Michigan, USA
| | - Mark R. Pittelkow
- Mayo Clinic Arizona Department of Dermatology, Scottsdale, Arizona, USA
| | | | - Aaron R. Mangold
- Mayo Clinic Arizona Department of Dermatology, Scottsdale, Arizona, USA
| |
Collapse
|
13
|
Zhang Q, Su C, Luo Y, Zheng F, Liang CL, Chen Y, Liu H, Qiu F, Liu Y, Feng W, Dai Z. Astragalus polysaccharide enhances antitumoral effects of chimeric antigen receptor- engineered (CAR) T cells by increasing CD122 +CXCR3 +PD-1 - memory T cells. Biomed Pharmacother 2024; 179:117401. [PMID: 39243425 DOI: 10.1016/j.biopha.2024.117401] [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: 06/27/2024] [Revised: 08/21/2024] [Accepted: 09/02/2024] [Indexed: 09/09/2024] Open
Abstract
Chimeric antigen receptor-engineered T (CAR-T) cell therapy of cancer has been a hotspot and promising. However, due to rapid exhaustion, CAR-T cells are less effective in solid tumors than in hematological ones. CD122+CXCR3+ memory T cells are characterized with longevity, self-renewal and great antitumoral capacity. Thus, it's compelling to induce memory CAR-T cells to enhance their efficacy on solid tumors. Astragalus polysaccharide (APS) has reportedly exhibited antitumoral effects. However, it's unclear if APS has an impact on CD8+ memory T cell generation or persistence. Using two human cancer cell lines, here we found that APS significantly improved the persistence of GPC3-targeted CAR-T cells and enhanced their suppression of tumor growth in both Huh7 and HepG2 xenograft models of hepatocellular carcinoma. APS increased CD122+/CXCR3+ memory T cells, but decreased their PD-1+ subset within CD8+ CAR-T cells in tumor-bearing mice, while these effects of APS were also confirmed with in vitro experiments. Moreover, APS augmented the expression of chemokines CXCL9/CXCL10 by the tumor in vivo and in vitro. It also enhanced the proliferation and chemotaxis/migration of CAR-T cells in vitro. Finally, APS promoted the phosphorylation of STAT5 in CD8+ CAR-T cells, whereas inhibition of STAT5 activation reversed these in vitro effects of APS. Therefore, APS enhanced the antitumoral effects of CD8+ CAR-T cells by promoting formation/persistence of CD122+/CXCR3+/PD-1- memory T cells and their migration to the tumor.
Collapse
Affiliation(s)
- Qunfang Zhang
- Immunology Program, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China; Section of Immunology, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, Guangdong 510006, China
| | - Chunzhao Su
- Immunology Program, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China
| | - Yini Luo
- Immunology Program, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China
| | - Fang Zheng
- Immunology Program, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China
| | - Chun-Ling Liang
- Immunology Program, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China; Section of Immunology, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, Guangdong 510006, China
| | - Yuchao Chen
- Immunology Program, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China; Section of Immunology, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, Guangdong 510006, China
| | - Huazhen Liu
- Immunology Program, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China; Section of Immunology, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, Guangdong 510006, China
| | - Feifei Qiu
- Immunology Program, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China; Section of Immunology, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, Guangdong 510006, China
| | - Yunshan Liu
- Department of Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Wenxuan Feng
- Immunology Program, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China.
| | - Zhenhua Dai
- Immunology Program, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China; Section of Immunology, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, Guangdong 510006, China.
| |
Collapse
|
14
|
Murayama M, Chow SK, Lee ML, Young B, Ergul YS, Shinohara I, Susuki Y, Toya M, Gao Q, Goodman SB. The interactions of macrophages, lymphocytes, and mesenchymal stem cells during bone regeneration. Bone Joint Res 2024; 13:462-473. [PMID: 39237112 PMCID: PMC11377107 DOI: 10.1302/2046-3758.139.bjr-2024-0122.r1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/07/2024] Open
Abstract
Bone regeneration and repair are crucial to ambulation and quality of life. Factors such as poor general health, serious medical comorbidities, chronic inflammation, and ageing can lead to delayed healing and nonunion of fractures, and persistent bone defects. Bioengineering strategies to heal bone often involve grafting of autologous bone marrow aspirate concentrate (BMAC) or mesenchymal stem cells (MSCs) with biocompatible scaffolds. While BMAC shows promise, variability in its efficacy exists due to discrepancies in MSC concentration and robustness, and immune cell composition. Understanding the mechanisms by which macrophages and lymphocytes - the main cellular components in BMAC - interact with MSCs could suggest novel strategies to enhance bone healing. Macrophages are polarized into pro-inflammatory (M1) or anti-inflammatory (M2) phenotypes, and influence cell metabolism and tissue regeneration via the secretion of cytokines and other factors. T cells, especially helper T1 (Th1) and Th17, promote inflammation and osteoclastogenesis, whereas Th2 and regulatory T (Treg) cells have anti-inflammatory pro-reconstructive effects, thereby supporting osteogenesis. Crosstalk among macrophages, T cells, and MSCs affects the bone microenvironment and regulates the local immune response. Manipulating the proportion and interactions of these cells presents an opportunity to alter the local regenerative capacity of bone, which potentially could enhance clinical outcomes.
Collapse
Affiliation(s)
- Masatoshi Murayama
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Simon K. Chow
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Max L. Lee
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Bill Young
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Yasemin S. Ergul
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Issei Shinohara
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Yosuke Susuki
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Masakazu Toya
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Qi Gao
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Stuart B. Goodman
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California, USA
- Department of Bioengineering, Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
15
|
Xu Z, Ma W, Wang J, Chen H, Li H, Yin Z, Hao J, Chen K. Nuclear HMGB1 is critical for CD8 T cell IFN-γ production and anti-tumor immunity. Cell Rep 2024; 43:114591. [PMID: 39116204 DOI: 10.1016/j.celrep.2024.114591] [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: 03/05/2024] [Revised: 06/24/2024] [Accepted: 07/22/2024] [Indexed: 08/10/2024] Open
Abstract
HMGB1 (high-mobility group box-1) has been extensively studied as a damage-associated molecular pattern, with secreted cytokine function. However, its regulation on T cells, especially the function in the nucleus, has not been elucidated. Here, we use conditional knockout (HMGB1-f/f; CD2-cre) mice and find that HMGB1 potentiates the proliferation and interferon gamma (IFN-γ) expression of CD8 T cells rather than CD4 T cells. Notably, nuclear, but not secreted, HMGB1 supports the expression of IFN-γ in CD8 T cells via directly regulating the activity of Eomes, the transcription factor for IFN-γ. Functional study shows that HMGB1 promotes the anti-tumor ability of CD8 T cells in vitro and in vivo. Finally, tumor environmental interleukin-7 promotes HMGB1 and IFN-γ production via fatty acid oxidation in CD8 T cells. Overall, we identify the role of nuclear HMGB1 in CD8 T cell differentiation and demonstrate that it plays an important role in the anti-tumor programs of CD8 T cells.
Collapse
Affiliation(s)
- Zhiguang Xu
- Department of Spine Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, P.R. China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, P.R. China
| | - Weiying Ma
- Department of Anesthesiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, P.R. China
| | - Ji Wang
- Department of Spine Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, P.R. China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, P.R. China
| | - Haofan Chen
- Department of Spine Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, P.R. China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, P.R. China
| | - Hui Li
- School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, P.R. China
| | - Zhinan Yin
- Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital Affiliated with Jinan University, Jinan University, Zhuhai, Guangdong, P.R. China; The Biomedical Translational Research Institute, Faculty of Medical Science, Jinan University, Guangzhou, Guangdong, P.R. China.
| | - Jianlei Hao
- Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital Affiliated with Jinan University, Jinan University, Zhuhai, Guangdong, P.R. China; The Biomedical Translational Research Institute, Faculty of Medical Science, Jinan University, Guangzhou, Guangdong, P.R. China.
| | - Kebing Chen
- Department of Spine Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, P.R. China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, P.R. China.
| |
Collapse
|
16
|
Kitelinger LE, Thim EA, Zipkowitz SY, Price RJ, Bullock TNJ. Tissue- and Temporal-Dependent Dynamics of Myeloablation in Response to Gemcitabine Chemotherapy. Cells 2024; 13:1317. [PMID: 39195207 PMCID: PMC11352862 DOI: 10.3390/cells13161317] [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: 06/15/2024] [Revised: 07/26/2024] [Accepted: 07/28/2024] [Indexed: 08/29/2024] Open
Abstract
For triple-negative breast cancer (TNBC), the most aggressive subset of breast cancer, immune cell infiltrates have prognostic implications. The presence of myeloid-derived suppressor cells supports tumor progression, while tumor-infiltrating lymphocytes (TILs) correlate with improved survival and responsiveness to immunotherapy. Manipulating the abundance of these populations may enhance tumor immunity. Gemcitabine (GEM), a clinically employed chemotherapeutic, is reported to be systemically myeloablative, and thus it is a potentially useful adjunct therapy for promoting anti-tumor immunity. However, knowledge about the immunological effects of GEM intratumorally is limited. Thus, we directly compared the impact of systemic GEM on immune cell presence and functionality in the tumor microenvironment (TME) to its effects in the periphery. We found that GEM is not myeloablative in the TME; rather, we observed sustained, significant reductions in TILs and dendritic cells-crucial components in initiating an adaptive immune response. We also performed bulk-RNA sequencing to identify immunological alterations transcriptionally induced by GEM. While we found evidence of upregulation in the interferon-gamma (IFN-γ) response pathway, we determined that GEM-mediated growth control is not dependent on IFN-γ. Overall, our findings yield new insights into the tissue- and temporal-dependent immune ablative effects of GEM, contrasting the paradigm that this therapy is specifically myeloablative.
Collapse
Affiliation(s)
- Lydia E. Kitelinger
- Department of Pathology, University of Virginia, Charlottesville, VA 22908, USA; (L.E.K.); (S.Y.Z.)
| | - Eric A. Thim
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA;
| | - Sarah Y. Zipkowitz
- Department of Pathology, University of Virginia, Charlottesville, VA 22908, USA; (L.E.K.); (S.Y.Z.)
| | - Richard J. Price
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA;
| | - Timothy N. J. Bullock
- Department of Pathology, University of Virginia, Charlottesville, VA 22908, USA; (L.E.K.); (S.Y.Z.)
| |
Collapse
|
17
|
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
|
18
|
Hegazy AN, Peine C, Niesen D, Panse I, Vainshtein Y, Kommer C, Zhang Q, Brunner TM, Peine M, Fröhlich A, Ishaque N, Marek RM, Zhu J, Höfer T, Löhning M. Plasticity and lineage commitment of individual T H1 cells are determined by stable T-bet expression quantities. SCIENCE ADVANCES 2024; 10:eadk2693. [PMID: 38838155 PMCID: PMC11152138 DOI: 10.1126/sciadv.adk2693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 05/01/2024] [Indexed: 06/07/2024]
Abstract
T helper 1 (TH1) cell identity is defined by the expression of the lineage-specifying transcription factor T-bet. Here, we examine the influence of T-bet expression heterogeneity on subset plasticity by leveraging cell sorting of distinct in vivo-differentiated TH1 cells based on their quantitative expression of T-bet and interferon-γ. Heterogeneous T-bet expression states were regulated by virus-induced type I interferons and were stably maintained even after secondary viral infection. Exposed to alternative differentiation signals, the sorted subpopulations exhibited graded levels of plasticity, particularly toward the TH2 lineage: T-bet quantities were inversely correlated with the ability to express the TH2 lineage-specifying transcription factor GATA-3 and TH2 cytokines. Reprogramed TH1 cells acquired graded mixed TH1 + TH2 phenotypes with a hybrid epigenetic landscape. Continuous presence of T-bet in differentiated TH1 cells was essential to ensure TH1 cell stability. Thus, innate cytokine signals regulate TH1 cell plasticity via an individual cell-intrinsic rheostat to enable T cell subset adaptation to subsequent challenges.
Collapse
Affiliation(s)
- Ahmed N. Hegazy
- Charité—Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Medical Department of Gastroenterology, Infectious Diseases and Rheumatology, 12203 Berlin, Germany
- German Rheumatism Research Center (DRFZ), a Leibniz Institute, Inflammatory Mechanisms, 10117 Berlin, Germany
- Berlin Institute of Health (BIH) at Charité—Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Caroline Peine
- Charité—Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Experimental Immunology and Osteoarthritis Research, Department of Rheumatology and Clinical Immunology, 10117 Berlin, Germany
- German Rheumatism Research Center (DRFZ), a Leibniz Institute, Pitzer Laboratory of Osteoarthritis Research, 10117 Berlin, Germany
| | - Dominik Niesen
- Charité—Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Experimental Immunology and Osteoarthritis Research, Department of Rheumatology and Clinical Immunology, 10117 Berlin, Germany
- German Rheumatism Research Center (DRFZ), a Leibniz Institute, Pitzer Laboratory of Osteoarthritis Research, 10117 Berlin, Germany
| | - Isabel Panse
- Charité—Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Experimental Immunology and Osteoarthritis Research, Department of Rheumatology and Clinical Immunology, 10117 Berlin, Germany
- German Rheumatism Research Center (DRFZ), a Leibniz Institute, Pitzer Laboratory of Osteoarthritis Research, 10117 Berlin, Germany
| | - Yevhen Vainshtein
- German Cancer Research Center (DKFZ), Division of Theoretical Systems Biology, 69120 Heidelberg, Germany
- University of Heidelberg, Bioquant Center, 69120 Heidelberg, Germany
| | - Christoph Kommer
- German Cancer Research Center (DKFZ), Division of Theoretical Systems Biology, 69120 Heidelberg, Germany
- University of Heidelberg, Bioquant Center, 69120 Heidelberg, Germany
| | - Qin Zhang
- German Cancer Research Center (DKFZ), Division of Theoretical Systems Biology, 69120 Heidelberg, Germany
- University of Heidelberg, Bioquant Center, 69120 Heidelberg, Germany
| | - Tobias M. Brunner
- Charité—Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Experimental Immunology and Osteoarthritis Research, Department of Rheumatology and Clinical Immunology, 10117 Berlin, Germany
- German Rheumatism Research Center (DRFZ), a Leibniz Institute, Pitzer Laboratory of Osteoarthritis Research, 10117 Berlin, Germany
| | - Michael Peine
- Charité—Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Experimental Immunology and Osteoarthritis Research, Department of Rheumatology and Clinical Immunology, 10117 Berlin, Germany
- German Rheumatism Research Center (DRFZ), a Leibniz Institute, Pitzer Laboratory of Osteoarthritis Research, 10117 Berlin, Germany
| | - Anja Fröhlich
- Charité—Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Experimental Immunology and Osteoarthritis Research, Department of Rheumatology and Clinical Immunology, 10117 Berlin, Germany
- German Rheumatism Research Center (DRFZ), a Leibniz Institute, Pitzer Laboratory of Osteoarthritis Research, 10117 Berlin, Germany
| | - Naveed Ishaque
- German Cancer Research Center (DKFZ), Division of Theoretical Systems Biology, 69120 Heidelberg, Germany
- University of Heidelberg, Bioquant Center, 69120 Heidelberg, Germany
| | - Roman M. Marek
- Charité—Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Experimental Immunology and Osteoarthritis Research, Department of Rheumatology and Clinical Immunology, 10117 Berlin, Germany
- German Rheumatism Research Center (DRFZ), a Leibniz Institute, Pitzer Laboratory of Osteoarthritis Research, 10117 Berlin, Germany
| | - Jinfang Zhu
- National Institute of Allergy and Infectious Diseases, Laboratory of Immune System Biology, National Institutes of Health, Bethesda, MD 20892, USA
| | - Thomas Höfer
- German Cancer Research Center (DKFZ), Division of Theoretical Systems Biology, 69120 Heidelberg, Germany
- University of Heidelberg, Bioquant Center, 69120 Heidelberg, Germany
| | - Max Löhning
- Charité—Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Experimental Immunology and Osteoarthritis Research, Department of Rheumatology and Clinical Immunology, 10117 Berlin, Germany
- German Rheumatism Research Center (DRFZ), a Leibniz Institute, Pitzer Laboratory of Osteoarthritis Research, 10117 Berlin, Germany
| |
Collapse
|
19
|
Kotliar D, Curtis M, Agnew R, Weinand K, Nathan A, Baglaenko Y, Zhao Y, Sabeti PC, Rao DA, Raychaudhuri S. Reproducible single cell annotation of programs underlying T-cell subsets, activation states, and functions. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.03.592310. [PMID: 38746317 PMCID: PMC11092745 DOI: 10.1101/2024.05.03.592310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
T-cells recognize antigens and induce specialized gene expression programs (GEPs) enabling functions including proliferation, cytotoxicity, and cytokine production. Traditionally, different classes of helper T-cells express mutually exclusive responses - for example, Th1, Th2, and Th17 programs. However, new single-cell RNA sequencing (scRNA-Seq) experiments have revealed a continuum of T-cell states without discrete clusters corresponding to these subsets, implying the need for new analytical frameworks. Here, we advance the characterization of T-cells with T-CellAnnoTator (TCAT), a pipeline that simultaneously quantifies pre-defined GEPs capturing activation states and cellular subsets. From 1,700,000 T-cells from 700 individuals across 38 tissues and five diverse disease contexts, we discover 46 reproducible GEPs reflecting the known core functions of T-cells including proliferation, cytotoxicity, exhaustion, and T helper effector states. We experimentally characterize several novel activation programs and apply TCAT to describe T-cell activation and exhaustion in Covid-19 and cancer, providing insight into T-cell function in these diseases.
Collapse
Affiliation(s)
- Dylan Kotliar
- Center for Data Sciences, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
- Division of Rheumatology, Inflammation, and Immunity, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Harvard-MIT Division of Health Sciences and Technology, Harvard Medical School, Boston, MA 02115, USA
| | - Michelle Curtis
- Center for Data Sciences, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
- Division of Rheumatology, Inflammation, and Immunity, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Ryan Agnew
- Center for Data Sciences, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
- Division of Rheumatology, Inflammation, and Immunity, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Kathryn Weinand
- Center for Data Sciences, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
- Division of Rheumatology, Inflammation, and Immunity, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA 02115, USA
| | - Aparna Nathan
- Center for Data Sciences, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
- Division of Rheumatology, Inflammation, and Immunity, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA 02115, USA
| | - Yuriy Baglaenko
- Center for Data Sciences, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
- Center for Autoimmune Genetics and Etiology and Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
- Department of Pediatrics, University of Cincinnati, College of Medicine, Cincinnati, OH 45219, USA
| | - Yu Zhao
- Center for Data Sciences, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
- Division of Rheumatology, Inflammation, and Immunity, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Pardis C. Sabeti
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Department of Organismic and Evolutionary Biology, FAS Center for Systems Biology, Harvard University, Cambridge, MA 02138, USA
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
| | - Deepak A. Rao
- Division of Rheumatology, Inflammation, and Immunity, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Soumya Raychaudhuri
- Center for Data Sciences, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
- Division of Rheumatology, Inflammation, and Immunity, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
20
|
Kandel A, Li L, Wang Y, Tuo W, Xiao Z. Differentiation and Regulation of Bovine Th2 Cells In Vitro. Cells 2024; 13:738. [PMID: 38727273 PMCID: PMC11083891 DOI: 10.3390/cells13090738] [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: 03/19/2024] [Revised: 04/19/2024] [Accepted: 04/22/2024] [Indexed: 05/13/2024] Open
Abstract
Bovine Th2 cells have usually been characterized by IL4 mRNA expression, but it is unclear whether their IL4 protein expression corresponds to transcription. We found that grass-fed healthy beef cattle, which had been regularly exposed to parasites on the grass, had a low frequency of IL4+ Th2 cells during flow cytometry, similar to animals grown in feedlots. To assess the distribution of IL4+ CD4+ T cells across tissues, samples from the blood, spleen, abomasal (draining), and inguinal lymph nodes were examined, which revealed limited IL4 protein detection in the CD4+ T cells across the examined tissues. To determine if bovine CD4+ T cells may develop into Th2 cells, naïve cells were stimulated with anti-bovine CD3 under a Th2 differentiation kit in vitro. The cells produced primarily IFNγ proteins, with only a small fraction (<10%) co-expressing IL4 proteins. Quantitative PCR confirmed elevated IFNγ transcription but no significant change in IL4 transcription. Surprisingly, GATA3, the master regulator of IL4, was highest in naïve CD4+ T cells but was considerably reduced following differentiation. To determine if the differentiated cells were true Th2 cells, an unbiased proteomic assay was carried out. The assay identified 4212 proteins, 422 of which were differently expressed compared to those in naïve cells. Based on these differential proteins, Th2-related upstream components were predicted, including CD3, CD28, IL4, and IL33, demonstrating typical Th2 differentiation. To boost IL4 expression, T cell receptor (TCR) stimulation strength was reduced by lowering anti-CD3 concentrations. Consequently, weak TCR stimulation essentially abolished Th2 expansion and survival. In addition, extra recombinant bovine IL4 (rbIL4) was added during Th2 differentiation, but, despite enhanced expansion, the IL4 level remained unaltered. These findings suggest that, while bovine CD4+ T cells can respond to Th2 differentiation stimuli, the bovine IL4 pathway is not regulated in the same way as in mice and humans. Furthermore, Ostertagia ostertagi (OO) extract, a gastrointestinal nematode in cattle, inhibited signaling via CD3, CD28, IL4, and TLRs/MYD88, indicating that external pathogens can influence bovine Th2 differentiation. In conclusion, though bovine CD4+ T cells can respond to IL4-driven differentiation, IL4 expression is not a defining feature of differentiated bovine Th2 cells.
Collapse
Affiliation(s)
- Anmol Kandel
- Department of Animal and Avian Sciences, University of Maryland, College Park, MD 20742, USA; (A.K.); (L.L.)
| | - Lei Li
- Department of Animal and Avian Sciences, University of Maryland, College Park, MD 20742, USA; (A.K.); (L.L.)
| | - Yan Wang
- Mass Spectrometry Facility, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - Wenbin Tuo
- Animal Parasitic Diseases Laboratory, U.S. Department of Agriculture, Agricultural Research Service, Beltsville, MD 20705, USA;
| | - Zhengguo Xiao
- Department of Animal and Avian Sciences, University of Maryland, College Park, MD 20742, USA; (A.K.); (L.L.)
| |
Collapse
|
21
|
Schanz ML, Bitters AM, Zadeii KE, Joulani D, Chamberlain AK, López-Yglesias AH. IL-12 Mediates T-bet-Expressing Myeloid Cell-Dependent Host Resistance against Toxoplasma gondii. Immunohorizons 2024; 8:355-362. [PMID: 38687282 PMCID: PMC11066714 DOI: 10.4049/immunohorizons.2400029] [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: 04/05/2024] [Accepted: 04/15/2024] [Indexed: 05/02/2024] Open
Abstract
To defend against intracellular pathogens such as Toxoplasma gondii, the host generates a robust type 1 immune response. Specifically, host defense against T. gondii is defined by an IL-12-dependent IFN-γ response that is critical for host resistance. Previously, we demonstrated that host resistance is mediated by T-bet-dependent ILC-derived IFN-γ by maintaining IRF8+ conventional type 1 dendritic cells during parasitic infection. Therefore, we hypothesized that innate lymphoid cells are indispensable for host survival. Surprisingly, we observed that T-bet-deficient mice succumb to infection quicker than do mice lacking lymphocytes, suggesting an unknown T-bet-dependent-mediated host defense pathway. Analysis of parasite-mediated inflammatory myeloid cells revealed a novel subpopulation of T-bet+ myeloid cells (TMCs). Our results reveal that TMCs have the largest intracellular parasite burden compared with other professional phagocytes, suggesting they are associated with active killing of T. gondii. Mechanistically, we established that IL-12 is necessary for the induction of inflammatory TMCs during infection and these cells are linked to a role in host survival.
Collapse
Affiliation(s)
- Madison L. Schanz
- Department of Microbiology and Immunology, Indiana University School of Medicine–Terre Haute, Terre Haute, IN
| | - Abigail M. Bitters
- Department of Microbiology and Immunology, Indiana University School of Medicine–Terre Haute, Terre Haute, IN
| | - Kamryn E. Zadeii
- Department of Biology, Indiana State University, Terre Haute, IN
| | - Dana Joulani
- Department of Microbiology and Immunology, Indiana University School of Medicine–Terre Haute, Terre Haute, IN
| | - Angela K. Chamberlain
- Department of Microbiology and Immunology, Indiana University School of Medicine–Terre Haute, Terre Haute, IN
| | - Américo H. López-Yglesias
- Department of Microbiology and Immunology, Indiana University School of Medicine–Terre Haute, Terre Haute, IN
| |
Collapse
|
22
|
Zhu X, Hong S, Bu J, Liu Y, Liu C, Li R, Zhang T, Zhang Z, Li L, Zhou X, Hua Z, Zhu B, Hou B. Antiviral memory B cells exhibit enhanced innate immune response facilitated by epigenetic memory. SCIENCE ADVANCES 2024; 10:eadk0858. [PMID: 38552009 PMCID: PMC10980274 DOI: 10.1126/sciadv.adk0858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 02/26/2024] [Indexed: 04/01/2024]
Abstract
The long-lasting humoral immunity induced by viral infections or vaccinations depends on memory B cells with greatly increased affinity to viral antigens, which are evolved from germinal center (GC) responses. However, it is unclear whether antiviral memory B cells represent a distinct subset among the highly heterogeneous memory B cell population. Here, we examined memory B cells induced by a virus-mimicking antigen at both transcriptome and epigenetic levels and found unexpectedly that antiviral memory B cells exhibit an enhanced innate immune response, which appeared to be facilitated by the epigenetic memory that is established through the memory B cell development. In addition, T-bet is associated with the altered chromatin architecture and is required for the formation of the antiviral memory B cells. Thus, antiviral memory B cells are distinct from other GC-derived memory B cells in both physiological functions and epigenetic landmarks.
Collapse
Affiliation(s)
- Xiping Zhu
- Key Laboratory of Epigenetic Regulation and Intervention, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Sheng Hong
- Key Laboratory of Epigenetic Regulation and Intervention, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Jiachen Bu
- Key Laboratory of Epigenetic Regulation and Intervention, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Yingping Liu
- Key Laboratory of Epigenetic Regulation and Intervention, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Can Liu
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Runhan Li
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Tiantian Zhang
- Key Laboratory of Epigenetic Regulation and Intervention, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Zhuqiang Zhang
- Key Laboratory of Epigenetic Regulation and Intervention, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Liping Li
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Xuyu Zhou
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Zhaolin Hua
- Key Laboratory of Epigenetic Regulation and Intervention, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Bing Zhu
- Key Laboratory of Epigenetic Regulation and Intervention, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
- New Cornerstone Science Laboratory, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Baidong Hou
- Key Laboratory of Epigenetic Regulation and Intervention, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
23
|
Kim HW, Ko MK, Shin S, Park SH, Park JH, Kim SM, Lee MJ. Isoprinosine as a foot-and-mouth disease vaccine adjuvant elicits robust host defense against viral infection through immunomodulation. Front Cell Infect Microbiol 2024; 14:1331779. [PMID: 38510965 PMCID: PMC10951065 DOI: 10.3389/fcimb.2024.1331779] [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: 11/01/2023] [Accepted: 02/22/2024] [Indexed: 03/22/2024] Open
Abstract
Background Commercial foot-and-mouth disease (FMD) vaccines have limitations, such as local side effects, periodic vaccinations, and weak host defenses. To overcome these limitations, we developed a novel FMD vaccine by combining an inactivated FMD viral antigen with the small molecule isoprinosine, which served as an adjuvant (immunomodulator). Method We evaluated the innate and adaptive immune responses elicited by the novel FMD vaccine involved both in vitro and in vivo using mice and pigs. Results We demonstrated isoprinosine-mediated early, mid-term, and long-term immunity through in vitro and in vivo studies and complete host defense against FMD virus (FMDV) infection through challenge experiments in mice and pigs. We also elucidated that isoprinosine induces innate and adaptive (cellular and humoral) immunity via promoting the expression of immunoregulatory gene such as pattern recognition receptors [PRRs; retinoic acid-inducible gene (RIG)-I and toll like receptor (TLR)9], transcription factors [T-box transcription factor (TBX)21, eomesodermin (EOMES), and nuclear factor kappa B (NF-kB)], cytokines [interleukin (IL)-12p40, IL-23p19, IL-23R, and IL-17A)], and immune cell core receptors [cluster of differentiation (CD)80, CD86, CD28, CD19, CD21, and CD81] in pigs. Conclusion These findings present an attractive strategy for constructing novel FMD vaccines and other difficult-to-control livestock virus vaccine formulations based on isoprinosine induced immunomodulatory functions.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Min Ja Lee
- Center for Foot-and-Mouth Disease Vaccine Research, Animal and Plant Quarantine Agency, Gimcheon-si, Gyeongsangbuk-do, Republic of Korea
| |
Collapse
|
24
|
Shimasaki Y, Miyoshi H, Kawamoto K, Yoshida N, Mishina T, Nakashima K, Imamoto T, Sugio T, Yanagida E, Kato T, Yamada K, Takeuchi M, Suzuki T, Moritsubo M, Furuta T, Imaizumi Y, Takizawa J, Kato K, Suzumiya J, Suzuki R, Ohshima K. Clinicopathological comparison between PTCL-TBX21 and PTCL-GATA3 in Japanese patients. Cancer Med 2024; 13:e6793. [PMID: 38234210 PMCID: PMC10905534 DOI: 10.1002/cam4.6793] [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: 08/16/2023] [Revised: 11/17/2023] [Accepted: 11/23/2023] [Indexed: 01/19/2024] Open
Abstract
AIM Peripheral T-cell lymphoma not otherwise specified (PTCL-NOS) is a heterogeneous disease that can be classified into the PTCL-TBX21 and PTCL-GATA3 subtypes. METHODS In this study, we compared the clinicopathological features of PTCL-NOS in a Japanese cohort, classified using an IHC algorithm. RESULTS One hundred patients with PTCL-NOS were categorized as having PTCL-TBX21 (n = 55), PTCL-GATA3 (n = 24), or PTCL-unclassified (n = 21). When comparing PTCL-TBX21 and PTCL-GATA3, PTCL-TBX21 showed significantly lower CD4 positivity (p = 0.047), lower counts of high endothelial venules (p = 0.032), and a tendency for a better response to initial treatment (p = 0.088). Gene expression analysis using the nCounter system showed higher expression of tumor immunity-related genes, such as PD-L1, LAG3, and IDO1, in PTCL-TBX21 than in PTCL-GATA3. PTCL-GATA3 had significantly worse overall survival (OS) than those with PTCL-TBX21 (p = 0.047), although a similar tendency was observed for progression-free survival (PFS) (p = 0.064). PTCL-GATA3 was a prognostic factor for OS in univariate analysis (HR 2.02; 95% CI, 1.09-3.77; p = 0.027), although multivariate analysis did not show significance (HR 2.07; 95% CI, 0.93-4.61; p = 0.074). In the PFS analysis, PTCL-GATA3 was an independent prognostic factor by univariate analysis (HR 1.96; 95% CI, 1.08-3.56; p = 0.027) and multivariate analysis (HR 2.34; 95% CI, 1.07-5.11; p = 0.032). CONCLUSION The classification of PTCL-NOS into PTCL-TBX21 and PTCL-GATA3 is useful for predicting the prognosis of Japanese patients and stratifying the administration of tumor immune checkpoint inhibitors in clinical practice.
Collapse
Affiliation(s)
- Yasumasa Shimasaki
- Department of Pathology, School of MedicineKurume UniversityKurumeJapan
- Department of HematologyShimane University HospitalIzumoJapan
| | - Hiroaki Miyoshi
- Department of Pathology, School of MedicineKurume UniversityKurumeJapan
| | - Keisuke Kawamoto
- Department of Pathology, School of MedicineKurume UniversityKurumeJapan
- Department of Hematology, Endocrinology, and Metabolism, Faculty of MedicineNiigata UniversityNiigataJapan
| | - Noriaki Yoshida
- Department of Pathology, School of MedicineKurume UniversityKurumeJapan
- Department of Clinical StudiesRadiation Effects Research FoundationHiroshimaJapan
| | - Tatsuzo Mishina
- Department of Pathology, School of MedicineKurume UniversityKurumeJapan
- Department of HematologyChiba University HospitalChibaJapan
| | | | - Teppei Imamoto
- Department of Pathology, School of MedicineKurume UniversityKurumeJapan
- Department of Surgical PathologyHokkaido University HospitalSapporoJapan
| | - Takeshi Sugio
- Department of Medicine and Biosystemic Science, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | - Eriko Yanagida
- Department of Pathology, School of MedicineKurume UniversityKurumeJapan
| | - Takeharu Kato
- Department of Hematology, Atomic Bomb Disease and Hibakusha Medicine UnitNagasaki University Graduate School of Biomedical SciencesNagasakiJapan
| | - Kyohei Yamada
- Department of Pathology, School of MedicineKurume UniversityKurumeJapan
| | - Mai Takeuchi
- Department of Pathology, School of MedicineKurume UniversityKurumeJapan
| | - Takaharu Suzuki
- Department of Hematology, Endocrinology, and Metabolism, Faculty of MedicineNiigata UniversityNiigataJapan
| | - Mayuko Moritsubo
- Department of Pathology, School of MedicineKurume UniversityKurumeJapan
| | - Takuya Furuta
- Department of Pathology, School of MedicineKurume UniversityKurumeJapan
| | - Yoshitaka Imaizumi
- Department of Hematology, Atomic Bomb Disease and Hibakusha Medicine UnitNagasaki University Graduate School of Biomedical SciencesNagasakiJapan
| | - Jun Takizawa
- Department of Hematology, Endocrinology, and Metabolism, Faculty of MedicineNiigata UniversityNiigataJapan
| | - Koji Kato
- Department of Medicine and Biosystemic Science, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | | | - Ritsuro Suzuki
- Department of HematologyShimane University HospitalIzumoJapan
| | - Koichi Ohshima
- Department of Pathology, School of MedicineKurume UniversityKurumeJapan
| |
Collapse
|
25
|
Brunner TM, Serve S, Marx AF, Fadejeva J, Saikali P, Dzamukova M, Durán-Hernández N, Kommer C, Heinrich F, Durek P, Heinz GA, Höfer T, Mashreghi MF, Kühn R, Pinschewer DD, Löhning M. A type 1 immunity-restricted promoter of the IL-33 receptor gene directs antiviral T-cell responses. Nat Immunol 2024; 25:256-267. [PMID: 38172258 PMCID: PMC10834369 DOI: 10.1038/s41590-023-01697-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 11/02/2023] [Indexed: 01/05/2024]
Abstract
The pleiotropic alarmin interleukin-33 (IL-33) drives type 1, type 2 and regulatory T-cell responses via its receptor ST2. Subset-specific differences in ST2 expression intensity and dynamics suggest that transcriptional regulation is key in orchestrating the context-dependent activity of IL-33-ST2 signaling in T-cell immunity. Here, we identify a previously unrecognized alternative promoter in mice and humans that is located far upstream of the curated ST2-coding gene and drives ST2 expression in type 1 immunity. Mice lacking this promoter exhibit a selective loss of ST2 expression in type 1- but not type 2-biased T cells, resulting in impaired expansion of cytotoxic T cells (CTLs) and T-helper 1 cells upon viral infection. T-cell-intrinsic IL-33 signaling via type 1 promoter-driven ST2 is critical to generate a clonally diverse population of antiviral short-lived effector CTLs. Thus, lineage-specific alternative promoter usage directs alarmin responsiveness in T-cell subsets and offers opportunities for immune cell-specific targeting of the IL-33-ST2 axis in infections and inflammatory diseases.
Collapse
Affiliation(s)
- Tobias M Brunner
- Experimental Immunology and Osteoarthritis Research, Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.
- Pitzer Laboratory of Osteoarthritis Research, German Rheumatism Research Center (DRFZ), a Leibniz Institute, Berlin, Germany.
| | - Sebastian Serve
- Experimental Immunology and Osteoarthritis Research, Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Pitzer Laboratory of Osteoarthritis Research, German Rheumatism Research Center (DRFZ), a Leibniz Institute, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BIH Biomedical Innovation Academy, Berlin, Germany
| | - Anna-Friederike Marx
- Division of Experimental Virology, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Jelizaveta Fadejeva
- Experimental Immunology and Osteoarthritis Research, Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Pitzer Laboratory of Osteoarthritis Research, German Rheumatism Research Center (DRFZ), a Leibniz Institute, Berlin, Germany
| | - Philippe Saikali
- Experimental Immunology and Osteoarthritis Research, Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Pitzer Laboratory of Osteoarthritis Research, German Rheumatism Research Center (DRFZ), a Leibniz Institute, Berlin, Germany
| | - Maria Dzamukova
- Experimental Immunology and Osteoarthritis Research, Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Pitzer Laboratory of Osteoarthritis Research, German Rheumatism Research Center (DRFZ), a Leibniz Institute, Berlin, Germany
| | - Nayar Durán-Hernández
- Experimental Immunology and Osteoarthritis Research, Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Pitzer Laboratory of Osteoarthritis Research, German Rheumatism Research Center (DRFZ), a Leibniz Institute, Berlin, Germany
| | - Christoph Kommer
- Division of Theoretical Systems Biology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- BioQuant Center, University of Heidelberg, Heidelberg, Germany
| | - Frederik Heinrich
- Therapeutic Gene Regulation, German Rheumatism Research Center (DRFZ), a Leibniz Institute, Berlin, Germany
| | - Pawel Durek
- Therapeutic Gene Regulation, German Rheumatism Research Center (DRFZ), a Leibniz Institute, Berlin, Germany
| | - Gitta A Heinz
- Therapeutic Gene Regulation, German Rheumatism Research Center (DRFZ), a Leibniz Institute, Berlin, Germany
| | - Thomas Höfer
- Division of Theoretical Systems Biology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- BioQuant Center, University of Heidelberg, Heidelberg, Germany
| | - Mir-Farzin Mashreghi
- Therapeutic Gene Regulation, German Rheumatism Research Center (DRFZ), a Leibniz Institute, Berlin, Germany
| | - Ralf Kühn
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Daniel D Pinschewer
- Division of Experimental Virology, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Max Löhning
- Experimental Immunology and Osteoarthritis Research, Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.
- Pitzer Laboratory of Osteoarthritis Research, German Rheumatism Research Center (DRFZ), a Leibniz Institute, Berlin, Germany.
| |
Collapse
|
26
|
Hwang A, Kechter J, Do T, Hughes A, Zhang N, Li X, Wasikowski R, Brumfiel C, Patel M, Boudreaux B, Bhullar P, Nassir S, Yousif M, DiCaudo DJ, Fox J, Gharaee-Kermani M, Xing X, Zunich S, Branch E, Kahlenberg JM, Billi AC, Plazyo O, Tsoi LC, Pittelkow MR, Gudjonsson JE, Mangold AR. Oral Baricitinib in the Treatment of Cutaneous Lichen Planus. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.01.09.24300946. [PMID: 38260663 PMCID: PMC10802654 DOI: 10.1101/2024.01.09.24300946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Background Cutaneous lichen planus (LP) is a recalcitrant, difficult-to-treat, inflammatory skin disease characterized by pruritic, flat-topped, violaceous papules on the skin. Baricitinib is an oral Janus kinase (JAK) 1/2 inhibitor that interrupts the signaling pathway of interferon (IFN)-γ, a cytokine implicated in the pathogenesis of LP. Methods In this phase II trial, twelve patients with cutaneous LP received baricitinib 2 mg daily for 16 weeks, accompanied by in-depth spatial, single-cell, and bulk transcriptomic profiling of pre-and post-treatment samples. Results An early and sustained clinical response was seen with 83.3% of patients responsive at week 16. Our molecular data identified a unique, oligoclonal IFN-γ, CD8+, CXCL13+ cytotoxic T-cell population in LP skin and demonstrate a rapid decrease in interferon signature within 2 weeks of treatment, most prominent in the basal layer of the epidermis. Conclusion This study demonstrates the efficacy and molecular mechanisms of JAK inhibition in LP. Trial Registration Number : NCT05188521.
Collapse
|
27
|
Mehta V, Dwivedi AR, Ludhiadch A, Rana V, Goel KK, Uniyal P, Joshi G, Kumar A, Kumar B. A decade of USFDA-approved small molecules as anti-inflammatory agents: Recent trends and Commentaries on the "industrial" perspective. Eur J Med Chem 2024; 263:115942. [PMID: 38000212 DOI: 10.1016/j.ejmech.2023.115942] [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: 10/08/2023] [Revised: 11/05/2023] [Accepted: 11/05/2023] [Indexed: 11/26/2023]
Abstract
Inflammation is the human body's defence process against various pathogens, toxic substances, irradiation, and physically injured cells that have been damaged. Inflammation is characterized by swelling, pain, redness, heat, as well as diminished tissue function. Multiple important inflammatory markers determine the prognosis of inflammatory processes, which include likes of pro-inflammatory cytokines which are controlled by nuclear factor kappa-B (NF-kB), mitogen-activated protein kinase (MAPK), Janus kinase signal transducer and activator of transcription (JAK-STAT) pathway, all of which are activated in response to the stimulation of specific receptors. Besides these, the cyclooxygenase (COX) enzyme family also plays a significant role in inflammation. The current review is kept forth to compile a summary of small molecules-based drugs approved by the USFDA during the study period of 2013-2023. A thorough discussion has also been made to focus on biologics, macromolecules, and small chemical entities approved during this study period and their greener synthetic routes with a brief discussion on the chemical spacing parameters of anti-inflammatory drugs. The compilation is expected to assist the medicinal chemist and the scientist actively engaged in drug discovery and development of anti-inflammatory agents from newer perspectives during the current years.
Collapse
Affiliation(s)
- Vikrant Mehta
- Department of Cell Systems & Anatomy, University of Texas Health Science Center at San Antonio, Texas, 78229, USA
| | | | - Abhilash Ludhiadch
- Department of Genetics and Development, Columbia University Irving Medical Center, New York, 10032, USA
| | - Vikas Rana
- School of Pharmacy, Graphic Era Hill University, Clement town, Dehradun, 248002, Uttarakhand, India
| | - Kapil Kumar Goel
- Department of Pharmaceutical Sciences, Gurukul Kangri (Deemed to Be University), Haridwar, 249404, Uttarakhand, India
| | - Prerna Uniyal
- School of Pharmacy, Graphic Era Hill University, Clement town, Dehradun, 248002, Uttarakhand, India
| | - Gaurav Joshi
- Department of Pharmaceutical Sciences, HNB Garhwal University, Chauras Campus, Garhwal, Srinagar, Uttarakhand, 246174, India; Department of Biotechnology, Graphic Era (Deemed to be University), Dehradun, 248002, Uttarakhand, India.
| | - Asim Kumar
- Amity Institute of Pharmacy (AIP), Amity University Haryana, Panchgaon, Manesar, 122413, India.
| | - Bhupinder Kumar
- Department of Pharmaceutical Sciences, HNB Garhwal University, Chauras Campus, Garhwal, Srinagar, Uttarakhand, 246174, India.
| |
Collapse
|
28
|
Peng C, Jiang X, Jaeger M, van Houten P, van Herwaarden AE, Koeken VACM, Moorlag SJCFM, Mourits VP, Lemmers H, Dijkstra H, Koenen HJPM, Joosten I, van Cranenbroek B, Li Y, Joosten LAB, Netea MG, Netea-Maier RT, Xu CJ. 11-deoxycortisol positively correlates with T cell immune traits in physiological conditions. EBioMedicine 2024; 99:104935. [PMID: 38134621 PMCID: PMC10776925 DOI: 10.1016/j.ebiom.2023.104935] [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: 07/31/2023] [Revised: 12/07/2023] [Accepted: 12/07/2023] [Indexed: 12/24/2023] Open
Abstract
BACKGROUND Endogenous steroid hormones have significant effects on inflammatory and immune processes, but the immunological activities of steroidogenesis precursors remain largely unexplored. METHODS We conducted a systematic approach to examine the association between steroid hormones profile and immune traits in a cohort of 534 healthy volunteers. Serum concentrations of steroid hormones and their precursors (cortisol, progesterone, testosterone, androstenedione, 11-deoxycortisol and 17-OH progesterone) were determined by liquid chromatography-tandem mass spectrometry. Immune traits were evaluated by quantifying cellular composition of the circulating immune system and ex vivo cytokine responses elicited by major human pathogens and microbial ligands. An independent cohort of 321 individuals was used for validation, followed by in vitro validation experiments. FINDINGS We observed a positive association between 11-deoxycortisol and lymphoid cellular subsets numbers and function (especially IL-17 response). The association with lymphoid cellularity was validated in an independent validation cohort. In vitro experiments showed that, as compared to androstenedione and 17-OH progesterone, 11-deoxycortisol promoted T cell proliferation and Candida-induced Th17 polarization at physiologically relevant concentrations. Functionally, 11-deoxycortisol-treated T cells displayed a more activated phenotype (PD-L1high CD25high CD62Llow CD127low) in response to CD3/CD28 co-stimulation, and downregulated expression of T-bet nuclear transcription factor. INTERPRETATION Our findings suggest a positive association between 11-deoxycortisol and T-cell function under physiological conditions. Further investigation is needed to explore the potential mechanisms and clinical implications. FUNDING Found in acknowledgements.
Collapse
Affiliation(s)
- Chunying Peng
- Division of Endocrinology, Department of Internal Medicine, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Xun Jiang
- Centre for Individualised Infection Medicine (CiiM), A Joint Venture Between the Helmholtz-Centre for Infection Research (HZI) and Hannover Medical School (MHH), Hannover, Germany; TWINCORE, Centre for Experimental and Clinical Infection Research, A Joint Venture Between the Helmholtz-Centre for Infection Research (HZI) and Hannover Medical School (MHH), Hannover, Germany
| | - Martin Jaeger
- Division of Endocrinology, Department of Internal Medicine, Radboud University Medical Center, Nijmegen, the Netherlands; Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Pepijn van Houten
- Division of Endocrinology, Department of Internal Medicine, Radboud University Medical Center, Nijmegen, the Netherlands
| | | | - Valerie A C M Koeken
- Centre for Individualised Infection Medicine (CiiM), A Joint Venture Between the Helmholtz-Centre for Infection Research (HZI) and Hannover Medical School (MHH), Hannover, Germany; TWINCORE, Centre for Experimental and Clinical Infection Research, A Joint Venture Between the Helmholtz-Centre for Infection Research (HZI) and Hannover Medical School (MHH), Hannover, Germany; Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands; Research Centre Innovations in Care, Rotterdam University of Applied Science, Rotterdam, the Netherlands
| | - Simone J C F M Moorlag
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Vera P Mourits
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Heidi Lemmers
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Helga Dijkstra
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Hans J P M Koenen
- Laboratory Medical Immunology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Irma Joosten
- Laboratory Medical Immunology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Bram van Cranenbroek
- Laboratory Medical Immunology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Yang Li
- Centre for Individualised Infection Medicine (CiiM), A Joint Venture Between the Helmholtz-Centre for Infection Research (HZI) and Hannover Medical School (MHH), Hannover, Germany; TWINCORE, Centre for Experimental and Clinical Infection Research, A Joint Venture Between the Helmholtz-Centre for Infection Research (HZI) and Hannover Medical School (MHH), Hannover, Germany; Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Leo A B Joosten
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands; Department of Medical Genetics, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Mihai G Netea
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Romana T Netea-Maier
- Division of Endocrinology, Department of Internal Medicine, Radboud University Medical Center, Nijmegen, the Netherlands.
| | - Cheng-Jian Xu
- Centre for Individualised Infection Medicine (CiiM), A Joint Venture Between the Helmholtz-Centre for Infection Research (HZI) and Hannover Medical School (MHH), Hannover, Germany; TWINCORE, Centre for Experimental and Clinical Infection Research, A Joint Venture Between the Helmholtz-Centre for Infection Research (HZI) and Hannover Medical School (MHH), Hannover, Germany; Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands.
| |
Collapse
|
29
|
Rixon JA, Fong KD, Morris C, Nguyen AT, Depew CE, McSorley SJ. Elimination of Chlamydia muridarum from the female reproductive tract is IL-12p40 dependent, but independent of Th1 and Th2 cells. PLoS Pathog 2024; 20:e1011914. [PMID: 38166152 PMCID: PMC10786385 DOI: 10.1371/journal.ppat.1011914] [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: 09/28/2023] [Revised: 01/12/2024] [Accepted: 12/19/2023] [Indexed: 01/04/2024] Open
Abstract
Chlamydia vaccine approaches aspire to induce Th1 cells for optimal protection, despite the fact that there is no direct evidence demonstrating Th1-mediated Chlamydia clearance from the female reproductive tract (FRT). We recently reported that T-bet-deficient mice can resolve primary Chlamydia infection normally, undermining the potentially protective role of Th1 cells in Chlamydia immunity. Here, we show that T-bet-deficient mice develop robust Th17 responses and that mice deficient in Th17 cells exhibit delayed bacterial clearance, demonstrating that Chlamydia-specific Th17 cells represent an underappreciated protective population. Additionally, Th2-deficient mice competently clear cervicovaginal infection. Furthermore, we show that sensing of IFN-γ by non-hematopoietic cells is essential for Chlamydia immunity, yet bacterial clearance in the FRT does not require IFN-γ secretion by CD4 T cells. Despite the fact that Th1 cells are not necessary for Chlamydia clearance, protective immunity to Chlamydia is still dependent on MHC class-II-restricted CD4 T cells and IL-12p40. Together, these data point to IL-12p40-dependent CD4 effector maturation as essential for Chlamydia immunity, and Th17 cells to a lesser extent, yet neither Th1 nor Th2 cell development is critical. Future Chlamydia vaccination efforts will be more effective if they focus on induction of this protective CD4 T cell population.
Collapse
Affiliation(s)
- Jordan A. Rixon
- Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, University of California Davis, Davis, California, United States of America
| | - Kevin D. Fong
- Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, University of California Davis, Davis, California, United States of America
| | - Claire Morris
- Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, University of California Davis, Davis, California, United States of America
| | - Alana T. Nguyen
- Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, University of California Davis, Davis, California, United States of America
| | - Claire E. Depew
- Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, University of California Davis, Davis, California, United States of America
| | - Stephen J. McSorley
- Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, University of California Davis, Davis, California, United States of America
| |
Collapse
|
30
|
Zimmermann J, van Haren SD, Diray-Arce J, Adriawan IR, Wørzner K, Krog RT, Guleed S, Hu T, Mortensen R, Dietrich J, Solbak SMØ, Levy O, Christensen D, Pedersen GK. Co-adjuvanting DDA/TDB liposomes with a TLR7 agonist allows for IgG2a/c class-switching in the absence of Th1 cells. NPJ Vaccines 2023; 8:189. [PMID: 38135685 PMCID: PMC10746746 DOI: 10.1038/s41541-023-00781-0] [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: 06/16/2023] [Accepted: 11/22/2023] [Indexed: 12/24/2023] Open
Abstract
Class-switching to IgG2a/c in mice is a hallmark response to intracellular pathogens. T cells can promote class-switching and the predominant pathway for induction of IgG2a/c antibody responses has been suggested to be via stimulation from Th1 cells. We previously formulated CAF®01 (cationic liposomes containing dimethyldioctadecylammonium bromide (DDA) and Trehalose-6,6-dibehenate (TDB)) with the lipidated TLR7/8 agonist 3M-052 (DDA/TDB/3M-052), which promoted robust Th1 immunity in newborn mice. When testing this adjuvant in adult mice using the recombinant Chlamydia trachomatis (C.t.) vaccine antigen CTH522, it similarly enhanced IgG2a/c responses compared to DDA/TDB, but surprisingly reduced the magnitude of the IFN-γ+Th1 response in a TLR7 agonist dose-dependent manner. Single-cell RNA-sequencing revealed that DDA/TDB/3M-052 liposomes initiated early transcription of class-switch regulating genes directly in pre-germinal center B cells. Mixed bone marrow chimeras further demonstrated that this adjuvant did not require Th1 cells for IgG2a/c switching, but rather facilitated TLR7-dependent T-bet programming directly in B cells. This study underlines that adjuvant-directed IgG2a/c class-switching in vivo can occur in the absence of T-cell help, via direct activation of TLR7 on B cells and positions DDA/TDB/3M-052 as a powerful adjuvant capable of eliciting type I-like immunity in B cells without strong induction of Th1 responses.
Collapse
Affiliation(s)
- Julie Zimmermann
- Center for Vaccine Research, Statens Serum Institut, Copenhagen, Denmark
| | - Simon D van Haren
- Precision Vaccines Program, Boston Children's Hospital, Boston, MA, 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Joann Diray-Arce
- Precision Vaccines Program, Boston Children's Hospital, Boston, MA, 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | | | - Katharina Wørzner
- Center for Vaccine Research, Statens Serum Institut, Copenhagen, Denmark
| | - Ricki T Krog
- Center for Vaccine Research, Statens Serum Institut, Copenhagen, Denmark
| | - Safia Guleed
- Center for Vaccine Research, Statens Serum Institut, Copenhagen, Denmark
| | - Tu Hu
- Center for Vaccine Research, Statens Serum Institut, Copenhagen, Denmark
| | - Rasmus Mortensen
- Center for Vaccine Research, Statens Serum Institut, Copenhagen, Denmark
| | - Jes Dietrich
- Center for Vaccine Research, Statens Serum Institut, Copenhagen, Denmark
| | - Sara M Ø Solbak
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Ofer Levy
- Precision Vaccines Program, Boston Children's Hospital, Boston, MA, 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Dennis Christensen
- Center for Vaccine Research, Statens Serum Institut, Copenhagen, Denmark
| | - Gabriel K Pedersen
- Center for Vaccine Research, Statens Serum Institut, Copenhagen, Denmark.
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
31
|
Okuma HS, Watanabe K, Tsuchihashi K, Machida R, Sadachi R, Hirakawa A, Ariyama H, Kanai M, Kamikura M, Anjo K, Hiramitsu A, Sekine S, Okita N, Mano H, Nishikawa H, Nakamura K, Yonemori K. Phase II Trial of Nivolumab in Metastatic Rare Cancer with dMMR or MSI-H and Relation with Immune Phenotypic Analysis (the ROCK Trial). Clin Cancer Res 2023; 29:5079-5086. [PMID: 37819940 PMCID: PMC10722134 DOI: 10.1158/1078-0432.ccr-23-1807] [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: 06/15/2023] [Revised: 08/09/2023] [Accepted: 10/09/2023] [Indexed: 10/13/2023]
Abstract
PURPOSE Mismatch repair deficiency (dMMR)/microsatellite instability-high (MSI-H) are positive predictive markers for immune checkpoint inhibitors. However, data on the activity of nivolumab in advanced dMMR/MSI-H rare cancers and more accurate biomarkers are worth exploring. PATIENTS AND METHODS We conducted a multicenter phase II, open-label, single-arm clinical trial to explore the effectiveness and safety of nivolumab monotherapy in patients with advanced rare cancers with dMMR/MSI-H, in parallel with immune phenotype analysis, to explore new biomarkers. A Bayesian adaptive design was applied. Characterization of peripheral blood mononuclear cells (PBMC) was characterized by multicolor flow cytometric analysis and CyTOF using samples collected before and after the intervention. The dMMR was identified by the complete loss of MLH1/MSH2/MSH6/PMS2. RESULTS From May 2018 to March 2021, 242 patients were screened, and 11 patients were enrolled, of whom 10 were included in the full analysis. Median follow-up was 24.7 months (interquartile range, 12.4-31.5). Objective response rate was 60% [95% confidence interval (CI), 26.2-87.8] by central assessment and 70% (95% CI, 34.8-93.3) by local investigators. Median progression-free survival was 10.1 months (95% CI, 0.9-11.1). No treatment-related adverse events of grade 3 or higher were observed. Patients with a tumor mutation burden of ≥10/Mb showed a 100% response rate (95% CI, 47.8-100). Responders had increased T-bet+ PD-1+ CD4+ T cells in PBMC compared with nonresponders (P < 0.05). CONCLUSIONS The trial met its primary endpoint with nivolumab, demonstrating clinical benefit in advanced dMMR/MSI-H rare solid cancers. Besides, the proportion of T-bet+ PD-1+ CD4+ T-cells may serve as a novel predictive biomarker.
Collapse
Affiliation(s)
- Hitomi S. Okuma
- Department of Medical Oncology, National Cancer Center Hospital, Tsukiji, Chuo-ku, Tokyo, Japan
- Clinical Research Support Office, National Cancer Center Hospital, Tsukiji, Chuo-ku, Tokyo, Japan
| | - Keisuke Watanabe
- Division of Cancer Immunology, National Cancer Center Research Institute, Tsukiji, Chuo-ku, Tokyo, Japan
| | - Kenji Tsuchihashi
- Department of Hematology, Oncology & Cardiovascular Medicine, Kyushu University Hospital, Maidashi Higashi-ku, Fukuoka, Japan
| | - Ryunosuke Machida
- Clinical Research Support Office, National Cancer Center Hospital, Tsukiji, Chuo-ku, Tokyo, Japan
| | - Ryo Sadachi
- Clinical Research Support Office, National Cancer Center Hospital, Tsukiji, Chuo-ku, Tokyo, Japan
| | - Akihiro Hirakawa
- Department of Clinical Biostatistics, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Yushima, Bunkyo-ku, Tokyo, Japan
| | - Hiroshi Ariyama
- Department of Hematology, Oncology & Cardiovascular Medicine, Kyushu University Hospital, Maidashi Higashi-ku, Fukuoka, Japan
| | - Masashi Kanai
- Department of Therapeutic Oncology, Graduate School of Medicine, Kyoto University, Shogoin-kawahara-cho, Sakyo-ku, Kyoto, Japan
| | - Masahisa Kamikura
- Clinical Research Support Office, National Cancer Center Hospital, Tsukiji, Chuo-ku, Tokyo, Japan
| | - Kenta Anjo
- Clinical Research Support Office, National Cancer Center Hospital, Tsukiji, Chuo-ku, Tokyo, Japan
| | - Akari Hiramitsu
- Clinical Research Support Office, National Cancer Center Hospital, Tsukiji, Chuo-ku, Tokyo, Japan
| | - Shigeki Sekine
- Department of Diagnostic Pathology, National Cancer Center Hospital, Tsukiji, Chuo-ku, Tokyo, Japan
| | - Natsuko Okita
- Clinical Research Support Office, National Cancer Center Hospital, Tsukiji, Chuo-ku, Tokyo, Japan
| | - Hiroyuki Mano
- Division of Cellular Signaling, National Cancer Center Research Institute, Tsukiji, Chuo-ku, Tokyo, Japan
| | - Hiroyoshi Nishikawa
- Division of Cancer Immunology, Research Institute/Exploratory Oncology Research and Clinical Trial Center (EPOC), National Cancer Center, Tsukiji, Chuo-ku, Tokyo, Japan
| | - Kenichi Nakamura
- Clinical Research Support Office, National Cancer Center Hospital, Tsukiji, Chuo-ku, Tokyo, Japan
| | - Kan Yonemori
- Department of Medical Oncology, National Cancer Center Hospital, Tsukiji, Chuo-ku, Tokyo, Japan
| |
Collapse
|
32
|
Wu J, Lu Z, Zhao H, Lu M, Gao Q, Che N, Wang J, Ma T. The expanding Pandora's toolbox of CD8 +T cell: from transcriptional control to metabolic firing. J Transl Med 2023; 21:905. [PMID: 38082437 PMCID: PMC10714647 DOI: 10.1186/s12967-023-04775-3] [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: 08/17/2023] [Accepted: 11/28/2023] [Indexed: 12/18/2023] Open
Abstract
CD8+ T cells are the executor in adaptive immune response, especially in anti-tumor immunity. They are the subset immune cells that are of high plasticity and multifunction. Their development, differentiation, activation and metabolism are delicately regulated by multiple factors. Stimuli from the internal and external environment could remodel CD8+ T cells, and correspondingly they will also make adjustments to the microenvironmental changes. Here we describe the most updated progresses in CD8+ T biology from transcriptional regulation to metabolism mechanisms, and also their interactions with the microenvironment, especially in cancer and immunotherapy. The expanding landscape of CD8+ T cell biology and discovery of potential targets to regulate CD8+ T cells will provide new viewpoints for clinical immunotherapy.
Collapse
Affiliation(s)
- Jinghong Wu
- Cancer Research Center, Beijing Chest Hospital, Beijing Tuberculosis and Thoracic Tumor Research Institute, Capital Medical University, Beijing, 101149, China
| | - Zhendong Lu
- Cancer Research Center, Beijing Chest Hospital, Beijing Tuberculosis and Thoracic Tumor Research Institute, Capital Medical University, Beijing, 101149, China
| | - Hong Zhao
- Department of Pathology, Beijing Tuberculosis & Thoracic Tumor Research Institute, Beijing Chest Hospital, Capital Medical University, Beijing, 101149, China
| | - Mingjun Lu
- Cancer Research Center, Beijing Chest Hospital, Beijing Tuberculosis and Thoracic Tumor Research Institute, Capital Medical University, Beijing, 101149, China
| | - Qing Gao
- Cancer Research Center, Beijing Chest Hospital, Beijing Tuberculosis and Thoracic Tumor Research Institute, Capital Medical University, Beijing, 101149, China
| | - Nanying Che
- Department of Pathology, Beijing Tuberculosis & Thoracic Tumor Research Institute, Beijing Chest Hospital, Capital Medical University, Beijing, 101149, China
| | - Jinghui Wang
- Cancer Research Center, Beijing Chest Hospital, Beijing Tuberculosis and Thoracic Tumor Research Institute, Capital Medical University, Beijing, 101149, China.
| | - Teng Ma
- Cancer Research Center, Beijing Chest Hospital, Beijing Tuberculosis and Thoracic Tumor Research Institute, Capital Medical University, Beijing, 101149, China.
| |
Collapse
|
33
|
Narsale A, Almanza F, Tran T, Lam B, Seo D, Vu A, Long SA, Cooney L, Serti E, Davies JD. Th2 cell clonal expansion at diagnosis in human type 1 diabetes. Clin Immunol 2023; 257:109829. [PMID: 37907122 DOI: 10.1016/j.clim.2023.109829] [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: 08/27/2023] [Revised: 10/26/2023] [Accepted: 10/27/2023] [Indexed: 11/02/2023]
Abstract
Soon after diagnosis with type 1 diabetes (T1D), many patients experience a period of partial remission. A longer partial remission is associated with a better response to treatment, but the mechanism is not known. The frequency of CD4+CD25+CD127hi (127-hi) cells, a cell subset with an anti-inflammatory Th2 bias, correlates positively with length of partial remission. The purpose of this study was to further characterize the nature of the Th2 bias in 127-hi cells. Single cell RNA sequencing paired with TCR sequencing of sorted 127-hi memory cells identifies clonally expanded Th2 clusters in 127-hi cells from T1D, but not from healthy donors. The Th2 clusters express GATA3, GATA3-AS1, PTGDR2, IL17RB, IL4R and IL9R. The existence of 127-hi Th2 cell clonal expansion in T1D suggests that disease factors may induce clonal expansion of 127-hi Th2 cells that prolong partial remission and delay disease progression.
Collapse
Affiliation(s)
- Aditi Narsale
- San Diego Biomedical Research Institute, 3525 John Hopkins Court, San Diego, CA 92121, USA.
| | - Francisco Almanza
- San Diego Biomedical Research Institute, 3525 John Hopkins Court, San Diego, CA 92121, USA.
| | - Theo Tran
- San Diego Biomedical Research Institute, 3525 John Hopkins Court, San Diego, CA 92121, USA
| | - Breanna Lam
- San Diego Biomedical Research Institute, 3525 John Hopkins Court, San Diego, CA 92121, USA.
| | - David Seo
- San Diego Biomedical Research Institute, 3525 John Hopkins Court, San Diego, CA 92121, USA
| | - Alisa Vu
- San Diego Biomedical Research Institute, 3525 John Hopkins Court, San Diego, CA 92121, USA.
| | - S Alice Long
- Benaroya Research Institute, 1201 9(th) Ave, Seattle, WA 98101, USA.
| | | | | | - Joanna D Davies
- San Diego Biomedical Research Institute, 3525 John Hopkins Court, San Diego, CA 92121, USA.
| |
Collapse
|
34
|
Trujillo-Ochoa JL, Kazemian M, Afzali B. The role of transcription factors in shaping regulatory T cell identity. Nat Rev Immunol 2023; 23:842-856. [PMID: 37336954 PMCID: PMC10893967 DOI: 10.1038/s41577-023-00893-7] [Citation(s) in RCA: 48] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/16/2023] [Indexed: 06/21/2023]
Abstract
Forkhead box protein 3-expressing (FOXP3+) regulatory T cells (Treg cells) suppress conventional T cells and are essential for immunological tolerance. FOXP3, the master transcription factor of Treg cells, controls the expression of multiples genes to guide Treg cell differentiation and function. However, only a small fraction (<10%) of Treg cell-associated genes are directly bound by FOXP3, and FOXP3 alone is insufficient to fully specify the Treg cell programme, indicating a role for other accessory transcription factors operating upstream, downstream and/or concurrently with FOXP3 to direct Treg cell specification and specialized functions. Indeed, the heterogeneity of Treg cells can be at least partially attributed to differential expression of transcription factors that fine-tune their trafficking, survival and functional properties, some of which are niche-specific. In this Review, we discuss the emerging roles of accessory transcription factors in controlling Treg cell identity. We specifically focus on members of the basic helix-loop-helix family (AHR), basic leucine zipper family (BACH2, NFIL3 and BATF), CUT homeobox family (SATB1), zinc-finger domain family (BLIMP1, Ikaros and BCL-11B) and interferon regulatory factor family (IRF4), as well as lineage-defining transcription factors (T-bet, GATA3, RORγt and BCL-6). Understanding the imprinting of Treg cell identity and specialized function will be key to unravelling basic mechanisms of autoimmunity and identifying novel targets for drug development.
Collapse
Affiliation(s)
- Jorge L Trujillo-Ochoa
- Immunoregulation Section, Kidney Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), NIH, Bethesda, MD, USA
| | - Majid Kazemian
- Departments of Biochemistry and Computer Science, Purdue University, West Lafayette, IN, USA
| | - Behdad Afzali
- Immunoregulation Section, Kidney Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), NIH, Bethesda, MD, USA.
| |
Collapse
|
35
|
Hou W, Ji Z, Chen Z, Wherry EJ, Hicks SC, Ji H. A statistical framework for differential pseudotime analysis with multiple single-cell RNA-seq samples. Nat Commun 2023; 14:7286. [PMID: 37949861 PMCID: PMC10638410 DOI: 10.1038/s41467-023-42841-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Accepted: 10/24/2023] [Indexed: 11/12/2023] Open
Abstract
Pseudotime analysis with single-cell RNA-sequencing (scRNA-seq) data has been widely used to study dynamic gene regulatory programs along continuous biological processes. While many methods have been developed to infer the pseudotemporal trajectories of cells within a biological sample, it remains a challenge to compare pseudotemporal patterns with multiple samples (or replicates) across different experimental conditions. Here, we introduce Lamian, a comprehensive and statistically-rigorous computational framework for differential multi-sample pseudotime analysis. Lamian can be used to identify changes in a biological process associated with sample covariates, such as different biological conditions while adjusting for batch effects, and to detect changes in gene expression, cell density, and topology of a pseudotemporal trajectory. Unlike existing methods that ignore sample variability, Lamian draws statistical inference after accounting for cross-sample variability and hence substantially reduces sample-specific false discoveries that are not generalizable to new samples. Using both real scRNA-seq and simulation data, including an analysis of differential immune response programs between COVID-19 patients with different disease severity levels, we demonstrate the advantages of Lamian in decoding cellular gene expression programs in continuous biological processes.
Collapse
Affiliation(s)
- Wenpin Hou
- Department of Biostatistics, The Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, 21205, USA
- Department of Biostatistics, Mailman School of Public Health, Columbia University, New York, NY, 10032, USA
| | - Zhicheng Ji
- Department of Biostatistics, The Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, 21205, USA
- Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Zeyu Chen
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Parker Institute for Cancer Immunotherapy at University of Pennsylvania, Philadelphia, PA, 19104, USA
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
| | - E John Wherry
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Parker Institute for Cancer Immunotherapy at University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Stephanie C Hicks
- Department of Biostatistics, The Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, 21205, USA.
| | - Hongkai Ji
- Department of Biostatistics, The Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, 21205, USA.
| |
Collapse
|
36
|
Hoces D, Miguens Blanco J, Hernández-López RA. A synthetic biology approach to engineering circuits in immune cells. Immunol Rev 2023; 320:120-137. [PMID: 37464881 DOI: 10.1111/imr.13244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 06/08/2023] [Indexed: 07/20/2023]
Abstract
A synthetic circuit in a biological system involves the designed assembly of genetic elements, biomolecules, or cells to create a defined function. These circuits are central in synthetic biology, enabling the reprogramming of cellular behavior and the engineering of cells with customized responses. In cancer therapeutics, engineering T cells with circuits have the potential to overcome the challenges of current approaches, for example, by allowing specific recognition and killing of cancer cells. Recent advances also facilitate engineering integrated circuits for the controlled release of therapeutic molecules at specified locations, for example, in a solid tumor. In this review, we discuss recent strategies and applications of synthetic receptor circuits aimed at enhancing immune cell functions for cancer immunotherapy. We begin by introducing the concept of circuits in networks at the molecular and cellular scales and provide an analysis of the development and implementation of several synthetic circuits in T cells that have the goal to overcome current challenges in cancer immunotherapy. These include specific targeting of cancer cells, increased T-cell proliferation, and persistence in the tumor microenvironment. By harnessing the power of synthetic biology, and the characteristics of certain circuit architectures, it is now possible to engineer a new generation of immune cells that recognize cancer cells, while minimizing off-target toxicities. We specifically discuss T-cell circuits for antigen density sensing. These circuits allow targeting of solid tumors that share antigens with normal tissues. Additionally, we explore designs for synthetic circuits that could control T-cell differentiation or T-cell fate as well as the concept of synthetic multicellular circuits that leverage cellular communication and division of labor to achieve improved therapeutic efficacy. As our understanding of cell biology expands and novel tools for genome, protein, and cell engineering are developed, we anticipate further innovative approaches to emerge in the design and engineering of circuits in immune cells.
Collapse
Affiliation(s)
- Daniel Hoces
- Department of Bioengineering, Stanford University, Stanford, California, USA
- Department of Genetics, Stanford University, Stanford, California, USA
| | - Jesús Miguens Blanco
- Department of Bioengineering, Stanford University, Stanford, California, USA
- Department of Genetics, Stanford University, Stanford, California, USA
| | - Rogelio A Hernández-López
- Department of Bioengineering, Stanford University, Stanford, California, USA
- Department of Genetics, Stanford University, Stanford, California, USA
- Stanford Cancer Institute, Stanford, California, USA
- Chan-Zuckerberg Biohub-San Francisco, San Francisco, California, USA
| |
Collapse
|
37
|
Ding Y, Chen Y, Feng W, Huang G, Dong M, Zhao T, Chen N, Yang L, Mao G, Wu X. Persistent immune injury induced by short-term decabromodiphenyl ether (BDE-209) exposure to female middle-aged Balb/c mice. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2023; 30:111325-111343. [PMID: 37814044 DOI: 10.1007/s11356-023-30148-w] [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: 04/27/2023] [Accepted: 09/25/2023] [Indexed: 10/11/2023]
Abstract
Decabromodiphenyl ether (BDE-209), widely used in various industries for its excellent flame-retardant performance, could be enriched in humans and is closely associated with immune impairment. In addition, immune system is gradually declined and becoming more sensitive to environmental pollutants in the ageing process. Therefore, the immunotoxicity of BDE-209 (4, 40, and 400 mg/kg/day) to middle-aged mice and its recovery and susceptibility was first to be comprehensively investigated in this study. The results showed that BDE-209 exposure could lead to oxidative injury to immune organs (spleen, thymus, and liver), impair humoral (immunoglobulins), cellular (lymphopoiesis), and non-specific immunity, and disturb the expressions of the genes related to Th1/Th2 balance (T helper cells) in the middle-aged mice. In addition, Integrated Biomarker Response (IBR) indicated that BDE-209-induced immune impairment was challenging to self-regulated, and even exacerbated after 21 days of recovery and oxidative injury in immune organs could be the main reason. Furthermore, factorial analysis showed that middle-aged mice exposed to BDE-209 suffered from greater immune impairment than adult mice, and the immune impairment in aged mice is more difficult to be self-repaired than that in adult mice. It can be seen that the aged tend to suffer from BDE-209-induced persistent immune impairment and health threats.
Collapse
Affiliation(s)
- Yangyang Ding
- School of the Environment and Safety Engineering, Jiangsu University, Zhenjiang, Jiangsu, China
- The Laboratory Animal Research Center of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Yao Chen
- School of the Environment and Safety Engineering, Jiangsu University, Zhenjiang, Jiangsu, China
- Institute of Environmental Health and Ecological Security, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Weiwei Feng
- School of the Environment and Safety Engineering, Jiangsu University, Zhenjiang, Jiangsu, China
- Institute of Environmental Health and Ecological Security, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Guijuan Huang
- School of the Environment and Safety Engineering, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Mingyue Dong
- School of the Environment and Safety Engineering, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Ting Zhao
- School of Chemistry and Chemical Engineering, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Na Chen
- Zhenjiang Food and Drug Supervision and Inspection Center, Zhenjiang, Jiangsu, China
| | - Liuqing Yang
- School of Chemistry and Chemical Engineering, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Guanghua Mao
- School of the Environment and Safety Engineering, Jiangsu University, Zhenjiang, Jiangsu, China.
| | - Xiangyang Wu
- School of the Environment and Safety Engineering, Jiangsu University, Zhenjiang, Jiangsu, China
| |
Collapse
|
38
|
Yan K, Zhang F, Ren J, Huang Q, Yawalkar N, Han L. MicroRNA-125a-5p regulates the effect of Tregs on Th1 and Th17 through targeting ETS-1/STAT3 in psoriasis. J Transl Med 2023; 21:678. [PMID: 37773129 PMCID: PMC10543306 DOI: 10.1186/s12967-023-04427-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Accepted: 08/07/2023] [Indexed: 09/30/2023] Open
Abstract
BACKGROUND Psoriasis is an inflammatory disease mediated by helper T (Th)17 and Th1 cells. MicroRNA-125a (miR-125a) is reduced in the lesional skin of psoriatic patients. However, the mechanism by which miR-125a participates in psoriasis remains unclear. METHODS The levels of miR-125a-5p and its downstream targets (ETS-1, IFN-γ, and STAT3) were detected in CD4+ T cells of healthy controls and psoriatic patients by quantitative real-time PCR (qRT-PCR). In vitro, transfection of miR-125a-5p mimics was used to analyze the effect of miR-125a-5p on the differentiation of Th17 cells by flow cytometry. Imiquimod (IMQ)-induced mouse model was used to evaluate the role of upregulating miR-125a-5p by intradermal injection of agomir-125a-5p in vivo. RESULTS miR-125a-5p was downregulated in peripheral blood CD4+ T cells of psoriatic patients, which was positively associated with the proportion of regulatory T cells (Tregs) and negatively correlated with the Psoriasis Area and Severity Index (PASI) score. Moreover, the miR-125a-5p mimics promoted the differentiation of Tregs and downregulated the messenger RNA (mRNA) levels of ETS-1, IFN-γ, and STAT3 in murine CD4+ T cells. Furthermore, agomir-125a-5p alleviated psoriasis-like inflammation in an IMQ-induced mouse model by downregulating the proportion of Th17 cells. CONCLUSIONS miR-125a-5p may have therapeutic potential in psoriasis by restoring the suppressive function of Tregs on Th17 cells through targeting STAT3, and on Th1 cells indirectly through targeting ETS-1 and IFN-γ.
Collapse
Affiliation(s)
- Kexiang Yan
- Department of Dermatology, Huashan Hospital, Shanghai Institute of Dermatology, Fudan University, Shanghai, 200040, China
| | - Fuxin Zhang
- Department of Dermatology, Huashan Hospital, Shanghai Institute of Dermatology, Fudan University, Shanghai, 200040, China
| | - Jie Ren
- Department of Dermatology, Huashan Hospital, Shanghai Institute of Dermatology, Fudan University, Shanghai, 200040, China
| | - Qiong Huang
- Department of Dermatology, Huashan Hospital, Shanghai Institute of Dermatology, Fudan University, Shanghai, 200040, China
| | - Nikhil Yawalkar
- Department of Dermatology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Ling Han
- Department of Dermatology, Huashan Hospital, Shanghai Institute of Dermatology, Fudan University, Shanghai, 200040, China.
| |
Collapse
|
39
|
Tremain AC, Wallace RP, Lorentz KM, Thornley TB, Antane JT, Raczy MR, Reda JW, Alpar AT, Slezak AJ, Watkins EA, Maulloo CD, Budina E, Solanki A, Nguyen M, Bischoff DJ, Harrington JL, Mishra R, Conley GP, Marlin R, Dereuddre-Bosquet N, Gallouët AS, LeGrand R, Wilson DS, Kontos S, Hubbell JA. Synthetically glycosylated antigens for the antigen-specific suppression of established immune responses. Nat Biomed Eng 2023; 7:1142-1155. [PMID: 37679570 DOI: 10.1038/s41551-023-01086-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 08/02/2023] [Indexed: 09/09/2023]
Abstract
Inducing antigen-specific tolerance during an established immune response typically requires non-specific immunosuppressive signalling molecules. Hence, standard treatments for autoimmunity trigger global immunosuppression. Here we show that established antigen-specific responses in effector T cells and memory T cells can be suppressed by a polymer glycosylated with N-acetylgalactosamine (pGal) and conjugated to the antigen via a self-immolative linker that allows for the dissociation of the antigen on endocytosis and its presentation in the immunoregulatory environment. We show that pGal-antigen therapy induces antigen-specific tolerance in a mouse model of experimental autoimmune encephalomyelitis (with programmed cell-death-1 and the co-inhibitory ligand CD276 driving the tolerogenic responses), as well as the suppression of antigen-specific responses to vaccination against a DNA-based simian immunodeficiency virus in non-human primates. Our findings show that pGal-antigen therapy invokes mechanisms of immune tolerance to resolve antigen-specific inflammatory T-cell responses and suggest that the therapy may be applicable across autoimmune diseases.
Collapse
Affiliation(s)
- Andrew C Tremain
- Committee on Immunology, University of Chicago, Chicago, IL, USA
| | - Rachel P Wallace
- Pritzker School for Molecular Engineering, University of Chicago, Chicago, IL, USA
| | | | | | - Jennifer T Antane
- Pritzker School for Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Michal R Raczy
- Pritzker School for Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Joseph W Reda
- Pritzker School for Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Aaron T Alpar
- Pritzker School for Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Anna J Slezak
- Pritzker School for Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Elyse A Watkins
- Pritzker School for Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Chitavi D Maulloo
- Pritzker School for Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Erica Budina
- Pritzker School for Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Ani Solanki
- Animal Resources Center, University of Chicago, Chicago, IL, USA
| | - Mindy Nguyen
- Animal Resources Center, University of Chicago, Chicago, IL, USA
| | | | | | | | | | - Romain Marlin
- Center for Immunology of Viral, Auto-immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Université Paris-Saclay, INSERM, CEA, Fontenay-aux-Roses, France
| | - Nathalie Dereuddre-Bosquet
- Center for Immunology of Viral, Auto-immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Université Paris-Saclay, INSERM, CEA, Fontenay-aux-Roses, France
| | - Anne-Sophie Gallouët
- Center for Immunology of Viral, Auto-immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Université Paris-Saclay, INSERM, CEA, Fontenay-aux-Roses, France
| | - Roger LeGrand
- Center for Immunology of Viral, Auto-immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Université Paris-Saclay, INSERM, CEA, Fontenay-aux-Roses, France
| | - D Scott Wilson
- Pritzker School for Molecular Engineering, University of Chicago, Chicago, IL, USA.
- Biomedical Engineering Department, Johns Hopkins University, Baltimore, MD, USA.
| | | | - Jeffrey A Hubbell
- Committee on Immunology, University of Chicago, Chicago, IL, USA.
- Pritzker School for Molecular Engineering, University of Chicago, Chicago, IL, USA.
- Committee on Cancer Biology, University of Chicago, Chicago, IL, USA.
| |
Collapse
|
40
|
Bergeron HC, Hansen MR, Tripp RA. Interferons-Implications in the Immune Response to Respiratory Viruses. Microorganisms 2023; 11:2179. [PMID: 37764023 PMCID: PMC10535750 DOI: 10.3390/microorganisms11092179] [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: 07/17/2023] [Revised: 08/15/2023] [Accepted: 08/28/2023] [Indexed: 09/29/2023] Open
Abstract
Interferons (IFN) are an assemblage of signaling proteins made and released by various host cells in response to stimuli, including viruses. Respiratory syncytial virus (RSV), influenza virus, and SARS-CoV-2 are major causes of respiratory disease that induce or antagonize IFN responses depending on various factors. In this review, the role and function of type I, II, and III IFN responses to respiratory virus infections are considered. In addition, the role of the viral proteins in modifying anti-viral immunity is noted, as are the specific IFN responses that underly the correlates of immunity and protection from disease.
Collapse
Affiliation(s)
| | | | - Ralph A. Tripp
- Department of Infectious Diseases, University of Georgia College of Veterinary Medicine, Athens, GA 30605, USA; (H.C.B.); (M.R.H.)
| |
Collapse
|
41
|
Jacobsen LM, Diggins K, Blanchfield L, McNichols J, Perry DJ, Brant J, Dong X, Bacher R, Gersuk VH, Schatz DA, Atkinson MA, Mathews CE, Haller MJ, Long SA, Linsley PS, Brusko TM. Responders to low-dose ATG induce CD4+ T cell exhaustion in type 1 diabetes. JCI Insight 2023; 8:e161812. [PMID: 37432736 PMCID: PMC10543726 DOI: 10.1172/jci.insight.161812] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 07/06/2023] [Indexed: 07/12/2023] Open
Abstract
BACKGROUNDLow-dose anti-thymocyte globulin (ATG) transiently preserves C-peptide and lowers HbA1c in individuals with recent-onset type 1 diabetes (T1D); however, the mechanisms of action and features of the response remain unclear. Here, we characterized the post hoc immunological outcomes of ATG administration and their potential use as biomarkers of metabolic response to therapy (i.e., improved preservation of endogenous insulin production).METHODSWe assessed gene and protein expression, targeted gene methylation, and cytokine concentrations in peripheral blood following treatment with ATG (n = 29), ATG plus granulocyte colony-stimulating factor (ATG/G-CSF, n = 28), or placebo (n = 31).RESULTSTreatment with low-dose ATG preserved regulatory T cells (Tregs), as measured by stable methylation of FOXP3 Treg-specific demethylation region (TSDR) and increased proportions of CD4+FOXP3+ Tregs (P < 0.001) identified by flow cytometry. While treatment effects were consistent across participants, not all maintained C-peptide. Responders exhibited a transient rise in IL-6, IP-10, and TNF-α (P < 0.05 for all) 2 weeks after treatment and a durable CD4+ exhaustion phenotype (increased PD-1+KLRG1+CD57- on CD4+ T cells [P = 0.011] and PD1+CD4+ Temra MFI [P < 0.001] at 12 weeks, following ATG and ATG/G-CSF, respectively). ATG nonresponders displayed higher proportions of senescent T cells (at baseline and after treatment) and increased methylation of EOMES (i.e., less expression of this exhaustion marker).CONCLUSIONAltogether in these exploratory analyses, Th1 inflammation-associated serum and CD4+ exhaustion transcript and cellular phenotyping profiles may be useful for identifying signatures of clinical response to ATG in T1D.TRIAL REGISTRATIONClinicalTrials.gov NCT02215200.FUNDINGThe Leona M. and Harry B. Helmsley Charitable Trust (2019PG-T1D011), the NIH (R01 DK106191 Supplement, K08 DK128628), NIH TrialNet (U01 DK085461), and the NIH NIAID (P01 AI042288).
Collapse
Affiliation(s)
- Laura M. Jacobsen
- Department of Pediatrics, College of Medicine, University of Florida, Gainesville, Florida, USA
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, Florida, USA
| | - Kirsten Diggins
- Benaroya Research Institute at Virginia Mason, Seattle, Washington, USA
| | - Lori Blanchfield
- Benaroya Research Institute at Virginia Mason, Seattle, Washington, USA
| | - James McNichols
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, Florida, USA
| | - Daniel J. Perry
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, Florida, USA
| | - Jason Brant
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, Florida, USA
| | - Xiaoru Dong
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, Florida, USA
- Department of Biostatistics, University of Florida, Gainesville, Florida, USA
| | - Rhonda Bacher
- Department of Biostatistics, University of Florida, Gainesville, Florida, USA
| | - Vivian H. Gersuk
- Benaroya Research Institute at Virginia Mason, Seattle, Washington, USA
| | - Desmond A. Schatz
- Department of Pediatrics, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Mark A. Atkinson
- Department of Pediatrics, College of Medicine, University of Florida, Gainesville, Florida, USA
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, Florida, USA
| | - Clayton E. Mathews
- Department of Pediatrics, College of Medicine, University of Florida, Gainesville, Florida, USA
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, Florida, USA
| | - Michael J. Haller
- Department of Pediatrics, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - S. Alice Long
- Benaroya Research Institute at Virginia Mason, Seattle, Washington, USA
| | - Peter S. Linsley
- Benaroya Research Institute at Virginia Mason, Seattle, Washington, USA
| | - Todd M. Brusko
- Department of Pediatrics, College of Medicine, University of Florida, Gainesville, Florida, USA
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, Florida, USA
| |
Collapse
|
42
|
McDonald B, Chick BY, Ahmed NS, Burns M, Ma S, Casillas E, Chen D, Mann TH, O'Connor C, Hah N, Hargreaves DC, Kaech SM. Canonical BAF complex activity shapes the enhancer landscape that licenses CD8 + T cell effector and memory fates. Immunity 2023; 56:1303-1319.e5. [PMID: 37315534 PMCID: PMC10281564 DOI: 10.1016/j.immuni.2023.05.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 02/08/2023] [Accepted: 05/10/2023] [Indexed: 06/16/2023]
Abstract
CD8+ T cells provide host protection against pathogens by differentiating into distinct effector and memory cell subsets, but how chromatin is site-specifically remodeled during their differentiation is unclear. Due to its critical role in regulating chromatin and enhancer accessibility through its nucleosome remodeling activities, we investigated the role of the canonical BAF (cBAF) chromatin remodeling complex in antiviral CD8+ T cells during infection. ARID1A, a subunit of cBAF, was recruited early after activation and established de novo open chromatin regions (OCRs) at enhancers. Arid1a deficiency impaired the opening of thousands of activation-induced enhancers, leading to loss of TF binding, dysregulated proliferation and gene expression, and failure to undergo terminal effector differentiation. Although Arid1a was dispensable for circulating memory cell formation, tissue-resident memory (Trm) formation was strongly impaired. Thus, cBAF governs the enhancer landscape of activated CD8+ T cells that orchestrates TF recruitment and activity and the acquisition of specific effector and memory differentiation states.
Collapse
Affiliation(s)
- Bryan McDonald
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA 92037, USA; Biomedical Sciences Graduate Program, University of California at San Diego, La Jolla, CA 92093, USA
| | - Brent Y Chick
- Molecular and Cell Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA; Biological Sciences Graduate Program, University of California at San Diego, La Jolla, CA 92093, USA
| | - Nasiha S Ahmed
- Molecular and Cell Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Mannix Burns
- Molecular and Cell Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Shixin Ma
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Eduardo Casillas
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Dan Chen
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Thomas H Mann
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Carolyn O'Connor
- Flow Cytometry Core, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Nasun Hah
- Chapman Charitable Foundations Genomic Sequencing Core, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Diana C Hargreaves
- Molecular and Cell Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA.
| | - Susan M Kaech
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA 92037, USA.
| |
Collapse
|
43
|
Fevang B. Treatment of inflammatory complications in common variable immunodeficiency (CVID): current concepts and future perspectives. Expert Rev Clin Immunol 2023; 19:627-638. [PMID: 36996348 DOI: 10.1080/1744666x.2023.2198208] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/01/2023]
Abstract
INTRODUCTION Patients with Common variable immunodeficiency (CVID) have a high frequency of inflammatory complications like autoimmune cytopenias, interstitial lung disease and enteropathy. These patients have poor prognosis and effective, timely and safe treatment of inflammatory complications in CVID are essential, but guidelines and consensus on therapy are often lacking. AREAS COVERED This review will focus on current medical treatment of inflammatory complications in CVID and point out some future perspectives based on literature indexed in PubMed. There are a number of good observational studies and case reports on treatment of specific complications but randomized controlled trials are scarce. EXPERT OPINION In clinical practice, the most urgent issues that need to be addressed are the preferred treatment of GLILD, enteropathy and liver disease. Treating the underlying immune dysregulation and immune exhaustion in CVID is an alternative approach that potentially could alleviate these and other organ-specific inflammatory complications. Therapies of potential interest and wider use in CVID include mTOR-inhibitors like sirolimus, JAK-inhibitors like tofacitinib, the monoclonal IL-12/23 antibody ustekinumab, the anti-BAFF antibody belimumab and abatacept. For all inflammatory complications, there is a need for prospective therapeutic trials, preferably randomized controlled trials, and multi-center collaborations with larger cohorts of patients will be essential.
Collapse
Affiliation(s)
- Børre Fevang
- Centre for Rare Disorders, Oslo University Hospital, Oslo, Norway
- Section of Clinical Immunology and Infectious Diseases, Oslo University Hospital, Oslo, Norway
- Research Institute for Internal Medicine, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
44
|
Han SJ, Jain P, Gilad Y, Xia Y, Sung N, Park MJ, Dean AM, Lanz RB, Xu J, Dacso CC, Lonard DM, O'Malley BW. Steroid Receptor Coactivator-3 is a Key Modulator of Regulatory T Cell-Mediated Tumor Evasion. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.28.534575. [PMID: 37034717 PMCID: PMC10081245 DOI: 10.1101/2023.03.28.534575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Steroid receptor coactivator 3 (SRC-3) is most strongly expressed in regulatory T cells (Tregs) and B cells, suggesting that it plays an important role in the regulation of Treg function. Using an aggressive E0771 mouse breast cell line syngeneic immune-intact murine model, we observed that breast tumors were 'permanently eradicated' in a genetically engineered tamoxifen-inducible Treg-cell specific SRC-3 knockout (KO) female mouse that does not possess a systemic autoimmune pathological phenotype. A similar eradication of tumor was noted in a syngeneic model of prostate cancer. A subsequent injection of additional E0771 cancer cells into these mice showed continued resistance to tumor development without the need for tamoxifen induction to produce additional SRC-3 KO Tregs. SRC-3 KO Tregs were highly proliferative and preferentially infiltrated into breast tumors by activating the Chemokine (C-C motif) ligand (Ccl) 19/Ccl21/ Chemokine (C-C motif) Receptor (Ccr)7 signaling axis, generating antitumor immunity by enhancing the interferon-γ/C-X-C Motif Chemokine Ligand (Cxcl) 9 signaling axis to facilitate the entrance and function of effector T cells and Natural Killer cells. SRC-3 KO Tregs also show a dominant effect by blocking the immune suppressive function of WT Tregs. Importantly, a single adoptive transfer of SRC-3 KO Tregs into wild-type E0771 tumor-bearing mice can completely abolish pre-established breast tumors by generating potent antitumor immunity with a durable effect that prevents tumor reoccurrence. Therefore, treatment with SRC-3 deleted Tregs represents a novel approach to completely block tumor growth and recurrence without the autoimmune side-effects that typically accompany immune checkpoint modulators. Significance statement Tregs are essential in restraining immune responses for immune homeostasis. SRC-3 is a pleiotropic coactivator, the second-most highly expressed transcriptional coactivator in Tregs, and a suspect in Treg function. The disruption of SRC-3 expression in Tregs leads to a 'complete lifetime eradication' of tumors in aggressive syngeneic breast cancer mouse models because deletion of SRC-3 alters the expression of a wide range of key genes involved in efferent and afferent Treg signaling. SRC-3KO Tregs confer this long-lasting protection against cancer recurrence in mice without an apparent systemic autoimmune pathological phenotype. Therefore, treatment with SRC-3 deleted Tregs could represent a novel and efficient future target for eliminating tumor growth and recurrence without the autoimmune side-effects that typically accompany immune checkpoint modulators.
Collapse
|
45
|
Pritchard GH, Phan AT, Christian DA, Blain TJ, Fang Q, Johnson J, Roy NH, Shallberg L, Kedl RM, Hunter CA. Early T-bet promotes LFA1 upregulation required for CD8+ effector and memory T cell development. J Exp Med 2023; 220:e20191287. [PMID: 36445307 PMCID: PMC9712775 DOI: 10.1084/jem.20191287] [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: 07/12/2019] [Revised: 08/29/2022] [Accepted: 11/10/2022] [Indexed: 12/03/2022] Open
Abstract
The T-box transcription factor T-bet is regarded as a "master regulator" of CD4+ Th1 differentiation and IFN-γ production. However, in multiple models of infection, T-bet appears less critical for CD8+ T cell expansion and effector function. Here, we show that following vaccination with a replication-deficient strain of Toxoplasma gondii, CD8+ T cell expression of T-bet is required for optimal expansion of parasite-specific effector CD8+ T cells. Analysis of the early events associated with T cell activation reveals that the α chain of LFA1, CD11a, is a target of T-bet, and T-bet is necessary for CD8+ T cell upregulation of this integrin, which influences the initial priming of CD8+ effector T cells. We propose that the early expression of T-bet represents a T cell-intrinsic factor that optimizes T-DC interactions necessary to generate effector responses.
Collapse
Affiliation(s)
- Gretchen Harms Pritchard
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA
| | - Anthony T. Phan
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA
| | - David A. Christian
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA
| | - Trevor J. Blain
- Department of Immunology and Microbiology, School of Medicine, University of Colorado Denver, Aurora, CO
| | - Qun Fang
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA
| | - John Johnson
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA
| | - Nathan H. Roy
- Department of Pathology and Laboratory Medicine, Children’s Hospital of Philadelphia Research Institute and Perelman School of Medicine of the University of Pennsylvania, Philadelphia, PA
| | - Lindsey Shallberg
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA
| | - Ross M. Kedl
- Department of Immunology and Microbiology, School of Medicine, University of Colorado Denver, Aurora, CO
| | - Christopher A. Hunter
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
46
|
Rebech GT, Bragato JP, Costa SF, de Freitas JH, dos Santos MO, Soares MF, Eugênio FDR, dos Santos PSP, de Lima VMF. miR-148a regulation interferes in inflammatory cytokine and parasitic load in canine leishmaniasis. PLoS Negl Trop Dis 2023; 17:e0011039. [PMID: 36719867 PMCID: PMC9888699 DOI: 10.1371/journal.pntd.0011039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 12/19/2022] [Indexed: 02/01/2023] Open
Abstract
Canine leishmaniasis (CanL) is a severe public health threat. Infected animals mediate transmission of the Leishmania protozoan to humans via the sandfly's bite during a blood meal. CanL progression depends on the degree of suppression of the immune response, possibly associated with microRNAs (miR), which can modulate mRNA translation into proteins and (consequently) regulate cell function. Increased miR-148a in splenic leukocytes (SL) of dogs with CanL was observed in previous studies, and in silico analysis, identified possible pathways involved in immune response regulation that are affected by this miR. Therefore, we evaluated the involvement of miR-148a in the regulation of TNF-α, IL-6, IL-12, IL-1β, iNOS, MHCII, CD80, CD3, T-bet, and GATA-3 transcription factors and their relationship with parasite load in SL of dogs with CanL. Splenic leukocytes obtained from healthy and diseased dogs were transfected with miR-148a mimic and inhibitor oligonucleotides. After 48 hours, expression levels of MHCII, CD80, iNOS, CD3, T-bet, and GATA-3 were evaluated by flow cytometry, and concentrations of TNF-α, IL-12, IL-6, and IL-1β were measured in culture supernatants by capture enzyme-linked immunosorbent assays. Transfection of SL with miR-148a mimics decreased iNOS levels in cells and TNF-α, IL-6, and IL-12 in the supernatants of cultured SL from CanL dogs. Interestingly, transfection with miR-148a inhibitor decreased parasite load in SL cells. These results suggest a direct or not regulatory role of this miR in the immune response to Leishmania infantum infection. We conclude that miR-148a can modulate immune responses by regulating inflammatory cytokines during CanL. Our results contribute to understanding the complex host/parasite interaction in CanL and could assist the development of treatments.
Collapse
Affiliation(s)
- Gabriela Torres Rebech
- Department of Clinical Medicine, Surgery and Animal Reproduction, São Paulo State University (UNESP), School of Veterinary Medicine, Araçatuba, Brazil
- * E-mail: (GTR); (VMFDL)
| | - Jaqueline Poleto Bragato
- Department of Clinical Medicine, Surgery and Animal Reproduction, São Paulo State University (UNESP), School of Veterinary Medicine, Araçatuba, Brazil
| | - Sidnei Ferro Costa
- Department of Clinical Medicine, Surgery and Animal Reproduction, São Paulo State University (UNESP), School of Veterinary Medicine, Araçatuba, Brazil
| | - Jéssica Henrique de Freitas
- Department of Clinical Medicine, Surgery and Animal Reproduction, São Paulo State University (UNESP), School of Veterinary Medicine, Araçatuba, Brazil
| | - Marilene Oliveira dos Santos
- Department of Clinical Medicine, Surgery and Animal Reproduction, São Paulo State University (UNESP), School of Veterinary Medicine, Araçatuba, Brazil
| | - Matheus Fujimura Soares
- Department of Clinical Medicine, Surgery and Animal Reproduction, São Paulo State University (UNESP), School of Veterinary Medicine, Araçatuba, Brazil
| | - Flávia de Rezende Eugênio
- Department of Clinical Medicine, Surgery and Animal Reproduction, São Paulo State University (UNESP), School of Veterinary Medicine, Araçatuba, Brazil
| | - Paulo Sérgio Patto dos Santos
- Department of Clinical Medicine, Surgery and Animal Reproduction, São Paulo State University (UNESP), School of Veterinary Medicine, Araçatuba, Brazil
| | - Valéria Marçal Felix de Lima
- Department of Clinical Medicine, Surgery and Animal Reproduction, São Paulo State University (UNESP), School of Veterinary Medicine, Araçatuba, Brazil
- * E-mail: (GTR); (VMFDL)
| |
Collapse
|
47
|
Gaghan C, Browning M, Cortes AL, Gimeno IM, Kulkarni RR. Effect of CpG-Oligonucleotide in Enhancing Recombinant Herpes Virus of Turkey-Laryngotracheitis Vaccine-Induced Immune Responses in One-Day-Old Broiler Chickens. Vaccines (Basel) 2023; 11:vaccines11020294. [PMID: 36851171 PMCID: PMC9965839 DOI: 10.3390/vaccines11020294] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 01/25/2023] [Accepted: 01/25/2023] [Indexed: 02/03/2023] Open
Abstract
Infectious laryngotracheitis (ILT) is an economically important disease of chickens. While the recombinant vaccines can reduce clinical disease severity, the associated drawbacks are poor immunogenicity and delayed onset of immunity. Here, we used CpG-oligonucleotides (ODN) as an in ovo adjuvant in boosting recombinant herpesvirus of turkey-laryngotracheitis (rHVT-LT) vaccine-induced responses in one-day-old broiler chickens. Two CpG-ODN doses (5 and 10 μg/egg) with no adverse effect on the vaccine-virus replication or chick hatchability were selected for immune-response evaluation. Results showed that while CpG-ODN adjuvantation induced an increased transcription of splenic IFNγ and IL-1β, and lung IFNγ genes, the IL-1β gene expression in the lung was significantly downregulated compared to the control. Additionally, the transcription of toll-like receptor (TLR)21 in the spleen and lung and inducible nitric oxide synthase (iNOS) in the spleen of all vaccinated groups was significantly reduced. Furthermore, splenic cellular immunophenotyping showed that the CpG-ODN-10μg adjuvanted vaccination induced a significantly higher number of macrophages, TCRγδ+, and CD4+ T cells as well as a higher frequency of activated T cells (CD4+CD44+) when compared to the control. Collectively, the findings suggested that CpG-ODN can boost rHVT-LT-induced immune responses in day-old chicks, which may help in anti-ILT defense during their later stages of life.
Collapse
Affiliation(s)
| | | | | | - Isabel M. Gimeno
- Correspondence: (I.M.G.); (R.R.K.); Tel.: +1-919-513-6852 (I.M.G.); +1-919-513-6277 (R.R.K.)
| | - Raveendra R. Kulkarni
- Correspondence: (I.M.G.); (R.R.K.); Tel.: +1-919-513-6852 (I.M.G.); +1-919-513-6277 (R.R.K.)
| |
Collapse
|
48
|
León B. Understanding the development of Th2 cell-driven allergic airway disease in early life. FRONTIERS IN ALLERGY 2023; 3:1080153. [PMID: 36704753 PMCID: PMC9872036 DOI: 10.3389/falgy.2022.1080153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 12/21/2022] [Indexed: 01/12/2023] Open
Abstract
Allergic diseases, including atopic dermatitis, allergic rhinitis, asthma, and food allergy, are caused by abnormal responses to relatively harmless foreign proteins called allergens found in pollen, fungal spores, house dust mites (HDM), animal dander, or certain foods. In particular, the activation of allergen-specific helper T cells towards a type 2 (Th2) phenotype during the first encounters with the allergen, also known as the sensitization phase, is the leading cause of the subsequent development of allergic disease. Infants and children are especially prone to developing Th2 cell responses after initial contact with allergens. But in addition, the rates of allergic sensitization and the development of allergic diseases among children are increasing in the industrialized world and have been associated with living in urban settings. Particularly for respiratory allergies, greater susceptibility to developing allergic Th2 cell responses has been shown in children living in urban environments containing low levels of microbial contaminants, principally bacterial endotoxins [lipopolysaccharide (LPS)], in the causative aeroallergens. This review highlights the current understanding of the factors that balance Th2 cell immunity to environmental allergens, with a particular focus on the determinants that program conventional dendritic cells (cDCs) toward or away from a Th2 stimulatory function. In this context, it discusses transcription factor-guided functional specialization of type-2 cDCs (cDC2s) and how the integration of signals derived from the environment drives this process. In addition, it analyzes observational and mechanistic studies supporting an essential role for innate sensing of microbial-derived products contained in aeroallergens in modulating allergic Th2 cell immune responses. Finally, this review examines whether hyporesponsiveness to microbial stimulation, particularly to LPS, is a risk factor for the induction of Th2 cell responses and allergic sensitization during infancy and early childhood and the potential factors that may affect early-age response to LPS and other environmental microbial components.
Collapse
Affiliation(s)
- Beatriz León
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
49
|
Fang D, Healy A, Zhu J. Differential regulation of lineage-determining transcription factor expression in innate lymphoid cell and adaptive T helper cell subsets. Front Immunol 2023; 13:1081153. [PMID: 36685550 PMCID: PMC9846361 DOI: 10.3389/fimmu.2022.1081153] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 12/14/2022] [Indexed: 01/05/2023] Open
Abstract
CD4 T helper (Th) cell subsets, including Th1, Th2 and Th17 cells, and their innate counterparts innate lymphoid cell (ILC) subsets consisting of ILC1s, ILC2s and ILC3s, display similar effector cytokine-producing capabilities during pro-inflammatory immune responses. These lymphoid cell subsets utilize the same set of lineage-determining transcription factors (LDTFs) for their differentiation, development and functions. The distinct ontogeny and developmental niches between Th cells and ILCs indicate that they may adopt different external signals for the induction of LDTF during lineage commitment. Increasing evidence demonstrates that many conserved cis-regulatory elements at the gene loci of LDTFs are often preferentially utilized for the induction of LDTF expression during Th cell differentiation and ILC development at different stages. In this review, we discuss the functions of lineage-related cis-regulatory elements in inducing T-bet, GATA3 or RORγt expression based on the genetic evidence provided in recent publications. We also review and compare the upstream signals involved in LDTF induction in Th cells and ILCs both in vitro and in vivo. Finally, we discuss the possible mechanisms and physiological importance of regulating LDTF dynamic expression during ILC development and activation.
Collapse
Affiliation(s)
- Difeng Fang
- *Correspondence: Difeng Fang, ; Jinfang Zhu,
| | | | - Jinfang Zhu
- *Correspondence: Difeng Fang, ; Jinfang Zhu,
| |
Collapse
|
50
|
Tan TCJ, Kelly V, Zou X, Wright D, Ly T, Zamoyska R. Translation factor eIF5a is essential for IFNγ production and cell cycle regulation in primary CD8 + T lymphocytes. Nat Commun 2022; 13:7796. [PMID: 36528626 PMCID: PMC9759561 DOI: 10.1038/s41467-022-35252-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 11/24/2022] [Indexed: 12/23/2022] Open
Abstract
Control of mRNA translation adjusts protein production rapidly and facilitates local cellular responses to environmental conditions. Traditionally initiation of translation is considered to be a major translational control point, however, control of peptide elongation is also important. Here we show that the function of the elongation factor, eIF5a, is regulated dynamically in naïve CD8+ T cells upon activation by post-translational modification, whereupon it facilitates translation of specific subsets of proteins. eIF5a is essential for long-term survival of effector CD8+ T cells and sequencing of nascent polypeptides indicates that the production of proteins which regulate proliferation and key effector functions, particularly the production of IFNγ and less acutely TNF production and cytotoxicity, is dependent on the presence of functional eIF5a. Control of translation in multiple immune cell lineages is required to co-ordinate immune responses and these data illustrate that translational elongation contributes to post-transcriptional regulons important for the control of inflammation.
Collapse
Affiliation(s)
- Thomas C J Tan
- Institute of Immunology and Infection Research, University of Edinburgh, Ashworth Laboratories, Charlotte Auerbach Road, Edinburgh, EH9 3FL, UK
| | - Van Kelly
- Wellcome Trust Centre for Cell Biology, University of Edinburgh, Michael Swann Building, Max Born Crescent, Edinburgh, EH9 3BF, UK
| | - Xiaoyan Zou
- Institute of Immunology and Infection Research, University of Edinburgh, Ashworth Laboratories, Charlotte Auerbach Road, Edinburgh, EH9 3FL, UK
| | - David Wright
- Institute of Immunology and Infection Research, University of Edinburgh, Ashworth Laboratories, Charlotte Auerbach Road, Edinburgh, EH9 3FL, UK
| | - Tony Ly
- Wellcome Trust Centre for Cell Biology, University of Edinburgh, Michael Swann Building, Max Born Crescent, Edinburgh, EH9 3BF, UK
- Centre for Gene Regulation and Expression, Life Sciences Research Complex, University of Dundee, Dundee, DD1 5EH, UK
| | - Rose Zamoyska
- Institute of Immunology and Infection Research, University of Edinburgh, Ashworth Laboratories, Charlotte Auerbach Road, Edinburgh, EH9 3FL, UK.
| |
Collapse
|