1
|
Huang H, Li M, Song S, Feng S, Feng X, Liu Y, Yang P, Zheng P. Galectin 9 rescues the inducibility of IL-10 expression in regulatory B cells of patients with food allergy. Sci Rep 2025; 15:196. [PMID: 39747510 PMCID: PMC11696059 DOI: 10.1038/s41598-024-84079-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Accepted: 12/19/2024] [Indexed: 01/04/2025] Open
Abstract
The deregulation of immune responses is what causes food allergy (FA) to occur. FA's cause is still unknown. The goal of this study is to investigate the mechanism how the impaired production of IL-10 occurs in peripheral naive B cells of patients with FA. Samples from patients with FA and healthy controls (HC) were used to isolate CD19+ CD45R+ naive B cells from peripheral blood mononuclear cells (PBMC). Lipopolysaccharide (LPS) exposure was used to assess the expression of interleukin-10 (IL-10) in B cells. Although the FA and HC groups had similar total B cell counts, the FA patients had fewer IL-10+ B cell counts than the HC group. In peripheral B cells, the concentrations of IL-10 were inversely related to the concentrations of specific IgE, Th2 and Th1 cytokines in the serum. In patients with FA, peripheral B cells experienced impaired immune-suppressive functions. Galectin-9 could restore the defective induction of IL-10 expression in naive B cells of FA patients. In conclusion, FA patients with naive B cells experience impaired IL-10 induction. The induction of IL-10 in naive B cells of FA patients can be restored by galectin-9 treatment, which triggers B cells to differentiate into immune regulatory B cells.
Collapse
Affiliation(s)
- Huang Huang
- Department of Gastroenterology, Fifth Affiliated Hospital, Zhengzhou University, Zhengzhou, China
| | - Minyao Li
- Department of General Practice Medicine, Third Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Shuo Song
- Department of General Practice Medicine, Third Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Shiyu Feng
- Department of Traditional Chinese Medicine, Affiliated Shenzhen Maternity & Child Healthcare Hospital, Southern Medical University, Shenzhen, China
| | - Xiaoyang Feng
- Department of Traditional Chinese Medicine, Affiliated Shenzhen Maternity & Child Healthcare Hospital, Southern Medical University, Shenzhen, China
| | - Yu Liu
- Department of General Practice Medicine, Third Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Pingchang Yang
- Institute of Allergy & Immunology of Shenzhen University, State Key Laboratory of Respiratory Diseases Allergy Division, Shenzhen Key Laboratory of Allergy & Immunology, Shenzhen University, A7-509 at Lihu Campus. 1066 Xueyuan Blvd, Shenzhen, 518055, China.
| | - Pengyuan Zheng
- Department of Gastroenterology, Fifth Affiliated Hospital, Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
2
|
Wang Y, Chen X, Huws SA, Xu G, Li J, Ren J, Xu J, Guan LL, Yao J, Wu S. Ileal microbial microbiome and its secondary bile acids modulate susceptibility to nonalcoholic steatohepatitis in dairy goats. MICROBIOME 2024; 12:247. [PMID: 39578870 PMCID: PMC11585128 DOI: 10.1186/s40168-024-01964-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 11/02/2024] [Indexed: 11/24/2024]
Abstract
BACKGROUND Liver damage from nonalcoholic steatohepatitis (NASH) presents a significant challenge to the health and productivity of ruminants. However, the regulatory mechanisms behind variations in NASH susceptibility remain unclear. The gut‒liver axis, particularly the enterohepatic circulation of bile acids (BAs), plays a crucial role in regulating the liver diseases. Since the ileum is the primary site for BAs reabsorption and return to the liver, we analysed the ileal metagenome and metabolome, liver and serum metabolome, and liver single-nuclei transcriptome of NASH-resistant and susceptible goats together with a mice validation model to explore how ileal microbial BAs metabolism affects liver metabolism and immunity, uncovering the key mechanisms behind varied NASH pathogenesis in dairy goats. RESULTS In NASH goats, increased total cholesterol (TC), triglyceride (TG), and primary BAs and decreased secondary BAs in the liver and serum promoted hepatic fat accumulation. Increased ileal Escherichia coli, Erysipelotrichaceae bacterium and Streptococcus pneumoniae as well as proinflammatory compounds damaged ileal histological morphology, and increased ileal permeability contributes to liver inflammation. In NASH-tolerance (NASH-T) goats, increased ursodeoxycholic acid (UDCA), isodeoxycholic acid (isoDCA) and isolithocholic acid (isoLCA) in the liver, serum and ileal contents were attributed to ileal secondary BAs-producing bacteria (Clostridium, Bifidobacterium and Lactobacillus) and key microbial genes encoding enzymes. Meanwhile, decreased T-helper 17 (TH17) cells and increased regulatory T (Treg) cells proportion were identified in both liver and ileum of NASH-T goats. To further validate whether these key BAs affected the progression of NASH by regulating the proliferation of TH17 and Treg cells, the oral administration of bacterial UDCA, isoDCA and isoLCA to a high-fat diet-induced NASH mouse model confirmed the amelioration of NASH through the TH17 cell differentiation/IL-17 signalling/PPAR signalling pathway by these bacterial secondary BAs. CONCLUSION This study revealed the roles of ileal microbiome and its secondary BAs in resilience and susceptibility to NASH by affecting the hepatic Treg and TH17 cells proportion in dairy goats. Bacterial UDCA, isoDCA and isoLCA were demonstrated to alleviate NASH and could be novel postbiotics to modulate and improve the liver health in ruminants. Video Abstract.
Collapse
Affiliation(s)
- Yue Wang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
- Faculty of Land and Food Systems, the University of British Columbia, Vancouver, BC, V6T 1Z4, Canada
- Key Laboratory of Livestock Biology, Northwest A&F University, Yangling, Shaanxi, China
| | - Xiaodong Chen
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
- College of Animal Science and Technology, Ningxia University, Yinchuan, China
- Key Laboratory of Livestock Biology, Northwest A&F University, Yangling, Shaanxi, China
| | - Sharon A Huws
- Institute of Global Food Security, School of Biological Sciences, Queen's University Belfast, 19 Chlorine Gardens, Belfast, Northern Ireland, BT9 5DL, UK
| | - Guanghao Xu
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
- Key Laboratory of Livestock Biology, Northwest A&F University, Yangling, Shaanxi, China
| | - Jing Li
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
- Key Laboratory of Livestock Biology, Northwest A&F University, Yangling, Shaanxi, China
| | - Jianrong Ren
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
- Key Laboratory of Livestock Biology, Northwest A&F University, Yangling, Shaanxi, China
| | - Jingyi Xu
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
- Key Laboratory of Livestock Biology, Northwest A&F University, Yangling, Shaanxi, China
| | - Le Luo Guan
- Faculty of Land and Food Systems, the University of British Columbia, Vancouver, BC, V6T 1Z4, Canada.
| | - Junhu Yao
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China.
- Key Laboratory of Livestock Biology, Northwest A&F University, Yangling, Shaanxi, China.
| | - Shengru Wu
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China.
- Key Laboratory of Livestock Biology, Northwest A&F University, Yangling, Shaanxi, China.
| |
Collapse
|
3
|
Jiang S, Zhu L, Xu Y, Liu Z, Cai J, Zhu T, Fan Q, Zhao Z. Subcutaneously transplanted xenogeneic protein recruits treg cells and M2 macrophages to induce browning of inguinal white adipose tissue. Endocrine 2024; 86:631-643. [PMID: 38900356 DOI: 10.1007/s12020-024-03932-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 06/13/2024] [Indexed: 06/21/2024]
Abstract
OBJECTIVE To study whether subcutaneously embedding xenogeneic protein threads or synthetic polymer absorbable threads can improve obesity phenotypes and metabolic conditions, and to further explore its underlying mechanism. METHODS Thirty-six 8-week-old ob/ob mice were randomly allocated to three groups, respectively, receiving catgut embedding, PGA thread embedding or sham treatment bilaterally to the groin. Individual parameters including weight, food intake, and core temperature are recorded and metabolism assessment, energy expenditure analysis, and PET/CT scanning are also performed at fixed timepoints. After surgical incision, the inguinal white adipose tissue was histologically examined and its expression profile was tested and compared among groups 4 weeks and 12 weeks after operation. RESULTS Catgut embedding reduced weight gain and improved metabolic status in ob/ob mice. Browning of bilateral inguinal WAT (white adipose tissue) was induced after catgut embedding, with massive infiltration of Treg cells and M2 macrophages in the tissue slices of fat pads. IL-10 and TGF-β released by Treg cells targeted macrophages and the induced M2 macrophages increased the expression of thermogenic and anti-inflammatory genes in fat. The secretion of catecholamines by polarized M2 macrophages led to the activation of β3-AR-related pathways in adipocytes and the browning of adipose tissue. CONCLUSIONS Abdominal subcutaneous catgut embedding has the potential to combat obesity through the induction of WAT browning mediated by infiltrated Treg cells and macrophages.
Collapse
Affiliation(s)
- Shenglu Jiang
- Taizhou Central Hospital (Taizhou University Hospital), Taizhou, China
| | - Lili Zhu
- Taizhou Enze Hospital, Taizhou, China
| | - Yukun Xu
- Taizhou Central Hospital (Taizhou University Hospital), Taizhou, China
| | - Zhao Liu
- Taizhou Central Hospital (Taizhou University Hospital), Taizhou, China
| | - Jialin Cai
- Taizhou Central Hospital (Taizhou University Hospital), Taizhou, China
| | - Tao Zhu
- Taizhou Central Hospital (Taizhou University Hospital), Taizhou, China
| | - Qing Fan
- Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China.
| | - Zhenxiong Zhao
- Taizhou Central Hospital (Taizhou University Hospital), Taizhou, China.
| |
Collapse
|
4
|
Chmiel J, Stasiak M, Skrzypkowska M, Samson L, Łuczkiewicz P, Trzonkowski P. Regulatory T lymphocytes as a treatment method for rheumatoid arthritis - Superiority of allogeneic to autologous cells. Heliyon 2024; 10:e36512. [PMID: 39319132 PMCID: PMC11419861 DOI: 10.1016/j.heliyon.2024.e36512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 08/15/2024] [Accepted: 08/16/2024] [Indexed: 09/26/2024] Open
Abstract
Cellular therapies utilizing regulatory T cells (Tregs) have flourished in the autoimmunity space as a new pillar of medicine. These cells have shown a great promise in the treatment of such devastating conditions as type 1 diabetes mellitus (T1DM), systemic lupus erythematosus (SLE) and graft versus host disease (GVHD). Novel treatment protocols, which utilize Tregs-mediated suppressive mechanisms, are based on the two main strategies: administration of immunomodulatory factors affecting Tregs or adoptive cell transfer (ACT). ACT involves extraction, in vitro expansion and subsequent administration of Tregs that could be either of autologous or allogeneic origin. Rheumatoid arthritis (RA) is another autoimmune candidate where this treatment approach is being considered. RA remains an especially challenging adversary since it is one of the most frequent and debilitating conditions among all autoaggressive disorders. Noteworthy, Tregs circulating in RA patients' blood have been proven defective and unable to suppress inflammation and joint destruction. With this knowledge, adoptive transfer of compromised autologous Tregs in the fledgling clinical trials involving RA patients should be reconsidered. In this article we hypothesize that incorporation of healthy donor allogeneic Tregs may provide more lucid and beneficial results.
Collapse
Affiliation(s)
- Joanna Chmiel
- University Clinical Centre in Gdańsk, Second Clinic of Orthopaedics and Kinetic Organ Traumatology, Poland
- Faculty of Medicine, Medical University of Gdańsk, Poland
| | - Mariusz Stasiak
- University Clinical Centre in Gdańsk, Second Clinic of Orthopaedics and Kinetic Organ Traumatology, Poland
- Faculty of Medicine, Medical University of Gdańsk, Poland
| | - Maria Skrzypkowska
- Department of Medical Immunology, Faculty of Medicine, Medical University of Gdańsk, Poland
| | - Lucjan Samson
- University Clinical Centre in Gdańsk, Second Clinic of Orthopaedics and Kinetic Organ Traumatology, Poland
- Faculty of Medicine, Medical University of Gdańsk, Poland
| | | | - Piotr Trzonkowski
- Department of Medical Immunology, Faculty of Medicine, Medical University of Gdańsk, Poland
| |
Collapse
|
5
|
Wang Z, Tang R, Wang H, Li X, Liu Z, Li W, Peng G, Zhou H. Bioinformatics analysis of the role of lysosome-related genes in breast cancer. Comput Methods Biomech Biomed Engin 2024:1-20. [PMID: 39054687 DOI: 10.1080/10255842.2024.2379936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 07/05/2024] [Accepted: 07/09/2024] [Indexed: 07/27/2024]
Abstract
This study aimed to investigate the roles of lysosome-related genes in BC prognosis and immunity. Transcriptome data from TCGA and MSigDB, along with lysosome-related gene sets, underwent NMF cluster analysis, resulting in two subtypes. Using lasso regression and univariate/multivariate Cox regression analysis, an 11-gene signature was successfully identified and verified. High- and low-risk populations were dominated by HR+ sample types. There were differences in pathway enrichment, immune cell infiltration, and immune scores. Sensitive drugs targeting model genes were screened using GDSC and CCLE. This study constructed a reliable prognostic model with lysosome-related genes, providing valuable insights for BC clinical immunotherapy.
Collapse
Affiliation(s)
- Zhongming Wang
- Department of Breast Oncology, The Third People's Hospital of Yongzhou, Yongzhou City, Hunan Province, China
| | - Ruiyao Tang
- Department of Breast Oncology, The Third People's Hospital of Yongzhou, Yongzhou City, Hunan Province, China
| | - Huazhong Wang
- Department of Breast Oncology, The Third People's Hospital of Yongzhou, Yongzhou City, Hunan Province, China
| | - Xizhang Li
- Department of Breast Oncology, The Third People's Hospital of Yongzhou, Yongzhou City, Hunan Province, China
| | - Zhenbang Liu
- Department of Breast Oncology, The Third People's Hospital of Yongzhou, Yongzhou City, Hunan Province, China
| | - Wenjie Li
- Department of Breast Oncology, The Third People's Hospital of Yongzhou, Yongzhou City, Hunan Province, China
| | - Gui Peng
- Department of Breast Oncology, The Third People's Hospital of Yongzhou, Yongzhou City, Hunan Province, China
| | - Huaiying Zhou
- Department of Breast Oncology, The Third People's Hospital of Yongzhou, Yongzhou City, Hunan Province, China
| |
Collapse
|
6
|
Huang J, Li X, Zhu Q, Wang M, Xie Z, Zhao T. Imbalance of Th17 cells, Treg cells and associated cytokines in patients with systemic lupus erythematosus: a meta-analysis. Front Immunol 2024; 15:1425847. [PMID: 39086480 PMCID: PMC11288813 DOI: 10.3389/fimmu.2024.1425847] [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: 04/30/2024] [Accepted: 07/01/2024] [Indexed: 08/02/2024] Open
Abstract
Objective This article aims to investigate the changes of T helper 17 (Th17) cells, regulatory T (Treg) cells and their associated cytokines in patients with systemic lupus erythematosus (SLE). Methods Multiple databases were investigated to identify articles that explored Th17 cells, Treg cells and relevant cytokines in SLE patients. A random effects model was used for calculating pooled standardized mean differences. Stata version 15.0 was utilized to conduct the meta-analysis. Results The levels of Th17 cells, IL-17, IL-6, IL-21 and IL-10 were higher in SLE patients than in healthy controls (HCs), but the TGF-β levels were lower. The percentage of Treg cells was lower than HCs in SLE individuals older than 33. Among studies that had 93% or lower females, the percentage of Th17 cells was greater in patients than in HCs. However, the percentage of Treg cells was lower when the proportion of females was less than 90%. Patients with lupus nephritis or active SLE had an increased proportion of Th17 cells and a decreased proportion of Treg cells. Conclusions The increased level of Th17 cells and related cytokines could be the main reason for the elevated Th17/Treg ratio in SLE. The percentages of Th17 and Treg cells were associated with gender, age, disease activity and kidney function. Furthermore, the reduced proportions of Treg cells may primarily result in a rise in the Th17/Treg ratio in older or active SLE patients. Systematic Review Registration https://www.crd.york.ac.uk/prospero, identifier CRD42023454937.
Collapse
Affiliation(s)
- Jinge Huang
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
| | - Xiaolong Li
- College of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Qingmiao Zhu
- College of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Meijiao Wang
- College of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Zhijun Xie
- Key Laboratory of Chinese Medicine Rheumatology of Zhejiang Province, Research Institute of Chinese Medical Clinical Foundation and Immunology, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Ting Zhao
- Key Laboratory of Chinese Medicine Rheumatology of Zhejiang Province, Research Institute of Chinese Medical Clinical Foundation and Immunology, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
7
|
Contreras-Castillo E, García-Rasilla VY, García-Patiño MG, Licona-Limón P. Stability and plasticity of regulatory T cells in health and disease. J Leukoc Biol 2024; 116:33-53. [PMID: 38428948 DOI: 10.1093/jleuko/qiae049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 02/09/2024] [Accepted: 02/19/2024] [Indexed: 03/03/2024] Open
Abstract
The mechanisms that negatively regulate inflammation upon a pathogenic stimulus are crucial for the maintenance of tissue integrity and organ function. T regulatory cells are one of the main drivers in controlling inflammation. The ability of T regulatory cells to adapt to different inflammatory cues and suppress inflammation is one of the relevant features of T regulatory cells. During this process, T regulatory cells express different transcription factors associated with their counterparts, Th helper cells, including Tbx21, GATA-3, Bcl6, and Rorc. The acquisition of this transcription factor helps the T regulatory cells to suppress and migrate to the different inflamed tissues. Additionally, the T regulatory cells have different mechanisms that preserve stability while acquiring a particular T regulatory cell subtype. This review focuses on describing T regulatory cell subtypes and the mechanisms that maintain their identity in health and diseases.
Collapse
Affiliation(s)
- Eugenio Contreras-Castillo
- Departamento de Biología Celular y del Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Circuito exterior s/n, CU Coyoacán, México City 04510, Mexico
| | - Verónica Yutsil García-Rasilla
- Departamento de Biología Celular y del Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Circuito exterior s/n, CU Coyoacán, México City 04510, Mexico
| | - María Guadalupe García-Patiño
- Departamento de Biología Celular y del Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Circuito exterior s/n, CU Coyoacán, México City 04510, Mexico
| | - Paula Licona-Limón
- Departamento de Biología Celular y del Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Circuito exterior s/n, CU Coyoacán, México City 04510, Mexico
| |
Collapse
|
8
|
Chai WH, Ma Y, Li JJ, Guo F, Wu YZ, Liu JW. Immune cell signatures and causal association with irritable bowel syndrome: A mendelian randomization study. World J Clin Cases 2024; 12:3094-3104. [PMID: 38898868 PMCID: PMC11185378 DOI: 10.12998/wjcc.v12.i17.3094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 02/10/2024] [Accepted: 04/29/2024] [Indexed: 06/04/2024] Open
Abstract
BACKGROUND The mucosal barrier's immune-brain interactions, pivotal for neural development and function, are increasingly recognized for their potential causal and therapeutic relevance to irritable bowel syndrome (IBS). Prior studies linking immune inflammation with IBS have been inconsistent. To further elucidate this relationship, we conducted a Mendelian randomization (MR) analysis of 731 immune cell markers to dissect the influence of various immune phenotypes on IBS. Our goal was to deepen our understanding of the disrupted brain-gut axis in IBS and to identify novel therapeutic targets. AIM To leverage publicly available data to perform MR analysis on 731 immune cell markers and explore their impact on IBS. We aimed to uncover immunophenotypic associations with IBS that could inform future drug development and therapeutic strategies. METHODS We performed a comprehensive two-sample MR analysis to evaluate the causal relationship between immune cell markers and IBS. By utilizing genetic data from public databases, we examined the causal associations between 731 immune cell markers, encompassing median fluorescence intensity, relative cell abundance, absolute cell count, and morphological parameters, with IBS susceptibility. Sensitivity analyses were conducted to validate our findings and address potential heterogeneity and pleiotropy. RESULTS Bidirectional false discovery rate correction indicated no significant influence of IBS on immunophenotypes. However, our analysis revealed a causal impact of IBS on 30 out of 731 immune phenotypes (P < 0.05). Nine immune phenotypes demonstrated a protective effect against IBS [inverse variance weighting (IVW) < 0.05, odd ratio (OR) < 1], while 21 others were associated with an increased risk of IBS onset (IVW ≥ 0.05, OR ≥ 1). CONCLUSION Our findings underscore a substantial genetic correlation between immune cell phenotypes and IBS, providing valuable insights into the pathophysiology of the condition. These results pave the way for the development of more precise biomarkers and targeted therapies for IBS. Furthermore, this research enriches our comprehension of immune cell roles in IBS pathogenesis, offering a foundation for more effective, personalized treatment approaches. These advancements hold promise for improving IBS patient quality of life and reducing the disease burden on individuals and their families.
Collapse
Affiliation(s)
- Wei-Hao Chai
- Department of Graduate School, Xinjiang Medical University, Urumqi 830000, Xinjiang Uygur Autonomous Region, China
| | - Yan Ma
- Department of Anesthesiology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi 830054, Xinjiang Uygur Autonomous Region, China
| | - Jia-Jia Li
- Key Laboratory of Special Environmental Medicine of Xinjiang, General Hospital of Xinjiang Military Command of the PLA, Urumqi 830000, Xinjiang Uygur Autonomous Region, China
| | - Fei Guo
- Department of Emergency Trauma Surgery, The First Affiliated Hospital of Xinjiang Medical University, Urumqi 830054, Xinjiang Uygur Autonomous Region, China
| | - Yi-Zhan Wu
- Department of Graduate School, Xinjiang Medical University, Urumqi 830000, Xinjiang Uygur Autonomous Region, China
| | - Jiang-Wei Liu
- Key Laboratory of Special Environmental Medicine of Xinjiang, General Hospital of Xinjiang Military Command of the PLA, Urumqi 830000, Xinjiang Uygur Autonomous Region, China
| |
Collapse
|
9
|
Yao M, Chen H, Chen Z, Wang Y, Shi D, Wu D, Li W, Huang J, Chen G, Zheng Q, Ye Z, Zheng C, Yang Y. Genomic and transcriptomic significance of multiple primary lung cancers detected by next-generation sequencing in clinical settings. Carcinogenesis 2024; 45:387-398. [PMID: 38693810 DOI: 10.1093/carcin/bgae026] [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: 09/27/2023] [Revised: 03/18/2024] [Accepted: 04/26/2024] [Indexed: 05/03/2024] Open
Abstract
Effective diagnosis and understanding of the mechanism of intrapulmonary metastasis (IM) from multiple primary lung cancers (MPLC) aid clinical management. However, the actual detection panels used in the clinic are variable. Current research on tumor microenvironment (TME) of MPLC and IM is insufficient. Therefore, additional investigation into the differential diagnosis and discrepancies in TME between two conditions is crucial. Two hundred and fourteen non-small cell lung cancer patients with multiple tumors were enrolled and 507 samples were subjected to DNA sequencing (NGS 10). Then, DNA and RNA sequencing (master panel) were performed on the specimens from 32 patients, the TME profiles between tumors within each patient and across patients and the differentially expressed genes were compared. Four patients were regrouped with NGS 10 results. Master panel resolved the classifications of six undetermined patients. The TME in MPLC exhibited a high degree of infiltration by natural killer (NK) cells, CD56dim NK cells, endothelial cells, etc., P < 0.05. Conversely, B cells, activated B cells, regulatory cells, immature dendritic cells, etc., P < 0.001, were heavily infiltrated in the IM. NECTIN4 and LILRB4 mRNA were downregulated in the MPLC (P < 0.0001). Additionally, NECTIN4 (P < 0.05) and LILRB4 were linked to improved disease-free survival in the MPLC. In conclusion, IM is screened from MPLC by pathology joint NGS 10 detections, followed by a large NGS panel for indistinguishable patients. A superior prognosis of MPLC may be associated with an immune-activating TME and the downregulation of NECTIN4 and LILRB4 considered as potential drug therapeutic targets.
Collapse
Affiliation(s)
- Meihong Yao
- Department of Pathology, Fujian Medical University Union Hospital, No.29 Xinquan Road, Gulou District, Fuzhou 350001, Fujian Province, China
| | - Hu Chen
- Department of Pathology, Fujian Medical University Union Hospital, No.29 Xinquan Road, Gulou District, Fuzhou 350001, Fujian Province, China
| | - Zui Chen
- Department of Pathology, Fujian Medical University Union Hospital, No.29 Xinquan Road, Gulou District, Fuzhou 350001, Fujian Province, China
| | - Yingying Wang
- Department of Pathology, Fujian Medical University Union Hospital, No.29 Xinquan Road, Gulou District, Fuzhou 350001, Fujian Province, China
| | - Dongliang Shi
- Department of Pathology, Fujian Medical University Union Hospital, No.29 Xinquan Road, Gulou District, Fuzhou 350001, Fujian Province, China
| | - Dan Wu
- Department of Pathology, Fujian Medical University Union Hospital, No.29 Xinquan Road, Gulou District, Fuzhou 350001, Fujian Province, China
| | - Wen Li
- Department of Pathology, Fujian Medical University Union Hospital, No.29 Xinquan Road, Gulou District, Fuzhou 350001, Fujian Province, China
| | - Jianping Huang
- Department of Pathology, Fujian Medical University Union Hospital, No.29 Xinquan Road, Gulou District, Fuzhou 350001, Fujian Province, China
| | - Guizhen Chen
- Department of Pathology, Fujian Medical University Union Hospital, No.29 Xinquan Road, Gulou District, Fuzhou 350001, Fujian Province, China
| | - Qiaoling Zheng
- Department of Pathology, Fujian Medical University Union Hospital, No.29 Xinquan Road, Gulou District, Fuzhou 350001, Fujian Province, China
| | - Zhengtao Ye
- Department of Pathology, Fujian Medical University Union Hospital, No.29 Xinquan Road, Gulou District, Fuzhou 350001, Fujian Province, China
| | - Chenxin Zheng
- School of Economics, Xiamen University, No.422 Siming South Road, Siming District, Xiamen 361005, Fujian Province, China
| | - Yinghong Yang
- Department of Pathology, Fujian Medical University Union Hospital, No.29 Xinquan Road, Gulou District, Fuzhou 350001, Fujian Province, China
| |
Collapse
|
10
|
Zhang L, Xu Z, Li Y, Wu KJ, Yu C, Zhu W, Sun DL, Zhu L, Zhou J. Glutamine supplementation improves the activity and immunosuppressive action of induced regulatory T cells in vitro and in vivo. Transpl Immunol 2024; 84:102044. [PMID: 38663757 DOI: 10.1016/j.trim.2024.102044] [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: 10/12/2023] [Revised: 04/04/2024] [Accepted: 04/14/2024] [Indexed: 05/12/2024]
Abstract
BACKGROUND Glutamine is crucial for the activation and efficacy of T cells, and may play a role in regulating the immune environment. This study aimed to investigate the potential role of glutamine in the activation and proliferation of induced regulatory T cells (iTregs). METHODS CD4+CD45RA+T cells were sorted from peripheral blood mononuclear cells and cultured to analyze iTreg differentiation. Glutamine was then added to the culture system to evaluate the effects of glutamine on iTregs by determining oxidative phosphorylation (OXPHOS), apoptosis, and cytokine secretion. Additionally, a humanized murine graft-versus-host disease (GVHD) model was constructed to confirm the efficacy of glutamine-treated iTregs in vivo. RESULTS After being cultured in vitro, glutamine significantly enhanced the levels of Foxp3, CTLA-4, CD39, CD69, IL-10, TGF-β, and Ki67 (CTLA-4, IL-10, TGF-β are immunosuppressive markers of iTregs) compared with that of the control iTregs (P < 0.05). Furthermore, the growth curve showed that the proliferative ability of glutamine-treated iTregs was better than that of the control iTregs (P < 0.01). Compared with the control iTregs, glutamine supplementation significantly increased oxygen consumption rates and ATP production (P < 0.05), significantly downregulated Annexin V and Caspase 3, and upregulated BCL2 (P < 0.05). However, GPNA significantly reversed the effects of glutamine (P < 0.05). Finally, a xeno-GVHD mouse model was successfully established to confirm that glutamine-treated iTregs increased the mice survival rate, delayed weight loss, and alleviated colon injury. CONCLUSION Glutamine supplementation can improve the activity and immunosuppressive action of iTregs, and the possible mechanisms by which this occurs are related to cell proliferation, apoptosis, and OXPHOS.
Collapse
Affiliation(s)
- Li Zhang
- The Third Affiliated Hospital of Soochow University, Changzhou 213000, China; Liyang People's Hospital, Liyang 213300, China
| | - Zhongya Xu
- Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Yuanjiu Li
- The Third Affiliated Hospital of Soochow University, Changzhou 213000, China
| | - Ke-Jia Wu
- The Third Affiliated Hospital of Soochow University, Changzhou 213000, China
| | - Chongyuan Yu
- The Third Affiliated Hospital of Soochow University, Changzhou 213000, China
| | - Wenjie Zhu
- Kangda College of Nanjing Medical University, Nanjing, China
| | - Dong-Lin Sun
- The Third Affiliated Hospital of Soochow University, Changzhou 213000, China.
| | - Li Zhu
- The Third Affiliated Hospital of Soochow University, Changzhou 213000, China.
| | - Jun Zhou
- Children's Hospital of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
11
|
Blinova VG, Zhdanov DD. Many Faces of Regulatory T Cells: Heterogeneity or Plasticity? Cells 2024; 13:959. [PMID: 38891091 PMCID: PMC11171907 DOI: 10.3390/cells13110959] [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: 04/23/2024] [Revised: 05/21/2024] [Accepted: 05/22/2024] [Indexed: 06/21/2024] Open
Abstract
Regulatory T cells (Tregs) are essential for maintaining the immune balance in normal and pathological conditions. In autoimmune diseases and transplantation, they restrain the loss of self-tolerance and promote engraftment, whereas in cancer, an increase in Treg numbers is mostly associated with tumor growth and poor prognosis. Numerous markers and their combinations have been used to identify Treg subsets, demonstrating the phenotypic diversity of Tregs. The complexity of Treg identification can be hampered by the unstable expression of some markers, the decrease in the expression of a specific marker over time or the emergence of a new marker. It remains unclear whether such phenotypic shifts are due to new conditions or whether the observed changes are due to initially different populations. In the first case, cellular plasticity is observed, whereas in the second, cellular heterogeneity is observed. The difference between these terms in relation to Tregs is rather blurred. Considering the promising perspectives of Tregs in regenerative cell-based therapy, the existing confusing data on Treg phenotypes require further investigation and analysis. In our review, we introduce criteria that allow us to distinguish between the heterogeneity and plasticity of Tregs normally and pathologically, taking a closer look at their diversity and drawing the line between two terms.
Collapse
Affiliation(s)
- Varvara G. Blinova
- Laboratory of Medical Biotechnology, Institute of Biomedical Chemistry, Pogodinskaya st. 10/8, 119121 Moscow, Russia;
| | - Dmitry D. Zhdanov
- Laboratory of Medical Biotechnology, Institute of Biomedical Chemistry, Pogodinskaya st. 10/8, 119121 Moscow, Russia;
- Department of Biochemistry, People’s Friendship University of Russia Named after Patrice Lumumba (RUDN University), Miklukho-Maklaya st. 6, 117198 Moscow, Russia
| |
Collapse
|
12
|
Mamun AA, Shao C, Geng P, Wang S, Xiao J. Recent advances in molecular mechanisms of skin wound healing and its treatments. Front Immunol 2024; 15:1395479. [PMID: 38835782 PMCID: PMC11148235 DOI: 10.3389/fimmu.2024.1395479] [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: 03/04/2024] [Accepted: 05/03/2024] [Indexed: 06/06/2024] Open
Abstract
The skin, being a multifaceted organ, performs a pivotal function in the complicated wound-healing procedure, which encompasses the triggering of several cellular entities and signaling cascades. Aberrations in the typical healing process of wounds may result in atypical scar development and the establishment of a persistent condition, rendering patients more vulnerable to infections. Chronic burns and wounds have a detrimental effect on the overall quality of life of patients, resulting in higher levels of physical discomfort and socio-economic complexities. The occurrence and frequency of prolonged wounds are on the rise as a result of aging people, hence contributing to escalated expenditures within the healthcare system. The clinical evaluation and treatment of chronic wounds continue to pose challenges despite the advancement of different therapeutic approaches. This is mainly owing to the prolonged treatment duration and intricate processes involved in wound healing. Many conventional methods, such as the administration of growth factors, the use of wound dressings, and the application of skin grafts, are used to ease the process of wound healing across diverse wound types. Nevertheless, these therapeutic approaches may only be practical for some wounds, highlighting the need to advance alternative treatment modalities. Novel wound care technologies, such as nanotherapeutics, stem cell treatment, and 3D bioprinting, aim to improve therapeutic efficacy, prioritize skin regeneration, and minimize adverse effects. This review provides an updated overview of recent advancements in chronic wound healing and therapeutic management using innovative approaches.
Collapse
Affiliation(s)
- Abdullah Al Mamun
- Central Laboratory of The Lishui Hospital of Wenzhou Medical University, Lishui People’s Hospital, Lishui, Zhejiang, China
| | - Chuxiao Shao
- Central Laboratory of The Lishui Hospital of Wenzhou Medical University, Lishui People’s Hospital, Lishui, Zhejiang, China
| | - Peiwu Geng
- Central Laboratory of The Lishui Hospital of Wenzhou Medical University, Lishui People’s Hospital, Lishui, Zhejiang, China
| | - Shuanghu Wang
- Central Laboratory of The Lishui Hospital of Wenzhou Medical University, Lishui People’s Hospital, Lishui, Zhejiang, China
| | - Jian Xiao
- Central Laboratory of The Lishui Hospital of Wenzhou Medical University, Lishui People’s Hospital, Lishui, Zhejiang, China
- Molecular Pharmacology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
- Department of Wound Healing, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
13
|
Headen K, Jakaite V, Mesaric VA, Scotta C, Lombardi G, Nicolaides KH, Shangaris P. The Role of Regulatory T Cells and Their Therapeutic Potential in Hypertensive Disease of Pregnancy: A Literature Review. Int J Mol Sci 2024; 25:4884. [PMID: 38732104 PMCID: PMC11084408 DOI: 10.3390/ijms25094884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 04/22/2024] [Accepted: 04/23/2024] [Indexed: 05/13/2024] Open
Abstract
Hypertensive disorders of pregnancy (HDP), including preeclampsia (PE) and gestational hypertension (GH), are major causes of maternal and foetal morbidity and mortality. This review elucidates the role of regulatory T cells (Tregs) in the immunological aspects of HDP and explores their therapeutic potential. Tregs, which play a critical role in maintaining immune homeostasis, are crucial in pregnancy to prevent immune-mediated rejection of the foetus. The review highlights that Tregs contribute to immunological adaptation in normal pregnancy, ensuring foetal acceptance. In contrast, HDP is associated with Treg dysfunction, which is marked by decreased numbers and impaired regulatory capacity, leading to inadequate immune tolerance and abnormal placental development. This dysfunction is particularly evident in PE, in which Tregs fail to adequately modulate the maternal immune response against foetal antigens, contributing to the pathophysiology of the disorder. Therapeutic interventions aiming to modulate Treg activity represent a promising avenue for HDP management. Studies in animal models and limited clinical trials suggest that enhancing Treg functionality could mitigate HDP symptoms and improve pregnancy outcomes. However, given the multifactorial nature of HDP and the intricate regulatory mechanisms of Tregs, the review explores the complexities of translating in vitro and animal model findings into effective clinical therapies. In conclusion, while the precise role of Tregs in HDP is still being unravelled, their central role in immune regulation during pregnancy is indisputable. Further research is needed to fully understand the mechanisms by which Tregs contribute to HDP and to develop targeted therapies that can safely and effectively harness their regulatory potential for treating hypertensive diseases of pregnancy.
Collapse
Affiliation(s)
- Kyle Headen
- Department of Women and Children’s Health, School of Life Course Sciences, Faculty of Life Sciences & Medicine, King’s College London, London SE5 8AF, UK; (K.H.); (K.H.N.)
- Harris Birthright Research Centre for Fetal Medicine, King’s College Hospital, London SE5 8BB, UK; (V.J.); (V.A.M.)
| | - Vaidile Jakaite
- Harris Birthright Research Centre for Fetal Medicine, King’s College Hospital, London SE5 8BB, UK; (V.J.); (V.A.M.)
| | - Vita Andreja Mesaric
- Harris Birthright Research Centre for Fetal Medicine, King’s College Hospital, London SE5 8BB, UK; (V.J.); (V.A.M.)
| | - Cristiano Scotta
- Peter Gorer Department of Immunobiology, School of Immunology & Microbial Sciences, Faculty of Life Sciences & Medicine, King’s College London, London SE1 1UL, UK; (C.S.); (G.L.)
| | - Giovanna Lombardi
- Peter Gorer Department of Immunobiology, School of Immunology & Microbial Sciences, Faculty of Life Sciences & Medicine, King’s College London, London SE1 1UL, UK; (C.S.); (G.L.)
| | - Kypros H. Nicolaides
- Department of Women and Children’s Health, School of Life Course Sciences, Faculty of Life Sciences & Medicine, King’s College London, London SE5 8AF, UK; (K.H.); (K.H.N.)
- Harris Birthright Research Centre for Fetal Medicine, King’s College Hospital, London SE5 8BB, UK; (V.J.); (V.A.M.)
| | - Panicos Shangaris
- Department of Women and Children’s Health, School of Life Course Sciences, Faculty of Life Sciences & Medicine, King’s College London, London SE5 8AF, UK; (K.H.); (K.H.N.)
- Harris Birthright Research Centre for Fetal Medicine, King’s College Hospital, London SE5 8BB, UK; (V.J.); (V.A.M.)
- Peter Gorer Department of Immunobiology, School of Immunology & Microbial Sciences, Faculty of Life Sciences & Medicine, King’s College London, London SE1 1UL, UK; (C.S.); (G.L.)
- Immunoregulation Laboratory, Faculty of Life Sciences & Medicine, 5th Floor, Bermondsey Wing, Guy’s Hospital, London SE1 9RT, UK
| |
Collapse
|
14
|
Bi Y, Kong R, Peng Y, Cai D, Zhang Y, Yang F, Li X, Deng W, Liu F, He B, Cao C, Deng C, Tang X, Fan L, Yu H, Zhou Z. Multiply restimulated human cord blood-derived Tregs maintain stabilized phenotype and suppressive function and predict their therapeutic effects on autoimmune diabetes. Diabetol Metab Syndr 2024; 16:71. [PMID: 38515175 PMCID: PMC10956208 DOI: 10.1186/s13098-024-01277-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Accepted: 01/24/2024] [Indexed: 03/23/2024] Open
Abstract
BACKGROUND Regulatory T cells (Tregs) are involved in the maintenance of immune homeostasis and immune regulation. Clinical trials on the adoptive transfer of Tregs have been ongoing for > 10 years. However, many unresolved issues remain in the production of readymade Treg products and selection of patients. Hence, this study aimed to develop a method to expand off-the-shelf Tregs derived from umbilical cord blood (UCB-Tregs) in vitro without changing their phenotype and inhibitory function. In addition, the study intended to design an approach to precisely select patients who are more likely to benefit from the adoptive Treg transfer therapy. METHODS UCB-Tregs were isolated and cultured in a medium containing human recombinant IL-2 and rapamycin and then multiply restimulated with human T-activator CD3/CD28 dynabeads. The phenotype and suppressive capacity of Tregs were assessed on days 18 and 42. The relationship between the suppressive function of UCB-Tregs in vitro and clinical indicators was analyzed, and the ability of the in vitro suppressive capacity to predict the in vivo therapeutic effects was evaluated. RESULTS UCB-Tregs expanded 123-fold and 5,981-fold at 18 and 42 days, respectively. The suppressive function of UCB-Tregs on the proliferation of immune cells at 42 days was not significantly different compared with that of UCB-Tregs obtained at 18 days. The suppression rate of UCB-Tregs to PBMCs was negatively correlated with the course of diabetes. Moreover, the high-suppression group exhibited a better treatment response than the low-suppression group during the 12-month follow-up period. CONCLUSIONS Multiply restimulated UCB-Tregs expanded at a large scale without any alterations in their classical phenotypic features and inhibitory functions. The suppressive function of Tregs in vitro was negatively correlated with the disease duration. The present study revealed the possibility of predicting the in vivo therapeutic effects via the in vitro inhibition assay. Thus, these findings provided a method to obtain off-the-shelf Treg products and facilitated the selection of patients who are likely to respond to the treatment, thereby moving toward the goal of precision treatment.
Collapse
Affiliation(s)
- Yuanjie Bi
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, Hunan Engineering Research Center of Cell Therapy for Diabetes, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Ran Kong
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, Hunan Engineering Research Center of Cell Therapy for Diabetes, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Yani Peng
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, Hunan Engineering Research Center of Cell Therapy for Diabetes, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Donghua Cai
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, Hunan Engineering Research Center of Cell Therapy for Diabetes, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Yu Zhang
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, Hunan Engineering Research Center of Cell Therapy for Diabetes, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Fan Yang
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, Hunan Engineering Research Center of Cell Therapy for Diabetes, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Xia Li
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, Hunan Engineering Research Center of Cell Therapy for Diabetes, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Wen Deng
- Department of Obstetrics and Gynecology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Fang Liu
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, Hunan Engineering Research Center of Cell Therapy for Diabetes, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Binbin He
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, Hunan Engineering Research Center of Cell Therapy for Diabetes, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Chuqing Cao
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, Hunan Engineering Research Center of Cell Therapy for Diabetes, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Chao Deng
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, Hunan Engineering Research Center of Cell Therapy for Diabetes, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Xiaohan Tang
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, Hunan Engineering Research Center of Cell Therapy for Diabetes, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Li Fan
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, Hunan Engineering Research Center of Cell Therapy for Diabetes, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Haibo Yu
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, Hunan Engineering Research Center of Cell Therapy for Diabetes, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Zhiguang Zhou
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, Hunan Engineering Research Center of Cell Therapy for Diabetes, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China.
| |
Collapse
|
15
|
Zhang C, Li Y, Yu Y, Li Z, Xu X, Talifu Z, Liu W, Yang D, Gao F, Wei S, Zhang L, Gong H, Peng R, Du L, Li J. Impact of inflammation and Treg cell regulation on neuropathic pain in spinal cord injury: mechanisms and therapeutic prospects. Front Immunol 2024; 15:1334828. [PMID: 38348031 PMCID: PMC10859493 DOI: 10.3389/fimmu.2024.1334828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 01/03/2024] [Indexed: 02/15/2024] Open
Abstract
Spinal cord injury is a severe neurological trauma that can frequently lead to neuropathic pain. During the initial stages following spinal cord injury, inflammation plays a critical role; however, excessive inflammation can exacerbate pain. Regulatory T cells (Treg cells) have a crucial function in regulating inflammation and alleviating neuropathic pain. Treg cells release suppressor cytokines and modulate the function of other immune cells to suppress the inflammatory response. Simultaneously, inflammation impedes Treg cell activity, further intensifying neuropathic pain. Therefore, suppressing the inflammatory response while enhancing Treg cell regulatory function may provide novel therapeutic avenues for treating neuropathic pain resulting from spinal cord injury. This review comprehensively describes the mechanisms underlying the inflammatory response and Treg cell regulation subsequent to spinal cord injury, with a specific focus on exploring the potential mechanisms through which Treg cells regulate neuropathic pain following spinal cord injury. The insights gained from this review aim to provide new concepts and a rationale for the therapeutic prospects and direction of cell therapy in spinal cord injury-related conditions.
Collapse
Affiliation(s)
- Chunjia Zhang
- School of Rehabilitation, Capital Medical University, Beijing, China
- Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research Center, Beijing, China
| | - Yan Li
- Institute of Rehabilitation medicine, China Rehabilitation Research Center, Beijing, China
| | - Yan Yu
- Institute of Rehabilitation medicine, China Rehabilitation Research Center, Beijing, China
| | - Zehui Li
- School of Rehabilitation, Capital Medical University, Beijing, China
- Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research Center, Beijing, China
| | - Xin Xu
- School of Rehabilitation, Capital Medical University, Beijing, China
- Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research Center, Beijing, China
| | - Zuliyaer Talifu
- School of Population Medicine and Public Health, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing, China
| | - Wubo Liu
- School of Rehabilitation, Capital Medical University, Beijing, China
- Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research Center, Beijing, China
- Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Degang Yang
- School of Rehabilitation, Capital Medical University, Beijing, China
- Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research Center, Beijing, China
| | - Feng Gao
- School of Rehabilitation, Capital Medical University, Beijing, China
- Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research Center, Beijing, China
| | - Song Wei
- School of Rehabilitation, Capital Medical University, Beijing, China
- Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research Center, Beijing, China
| | - Liang Zhang
- School of Rehabilitation, Capital Medical University, Beijing, China
- Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research Center, Beijing, China
| | - Han Gong
- School of Rehabilitation, Capital Medical University, Beijing, China
- Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research Center, Beijing, China
| | - Run Peng
- School of Rehabilitation, Capital Medical University, Beijing, China
- Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research Center, Beijing, China
| | - Liangjie Du
- School of Rehabilitation, Capital Medical University, Beijing, China
- Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research Center, Beijing, China
| | - Jianjun Li
- School of Rehabilitation, Capital Medical University, Beijing, China
- Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research Center, Beijing, China
- Institute of Rehabilitation medicine, China Rehabilitation Research Center, Beijing, China
- Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| |
Collapse
|
16
|
Carriero F, Rubino V, Leone S, Montanaro R, Brancaleone V, Ruggiero G, Terrazzano G. Regulatory T R3-56 Cells in the Complex Panorama of Immune Activation and Regulation. Cells 2023; 12:2841. [PMID: 38132162 PMCID: PMC10742044 DOI: 10.3390/cells12242841] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 12/11/2023] [Accepted: 12/13/2023] [Indexed: 12/23/2023] Open
Abstract
The interplay between immune activation and immune regulation is a fundamental aspect of the functional harmony of the immune system. This delicate balance is essential to triggering correct and effective immune responses against pathogens while preventing excessive inflammation and the immunopathogenic mechanisms of autoimmunity. The knowledge of all the mechanisms involved in immune regulation is not yet definitive, and, probably, the overall picture is much broader than what has been described in the scientific literature so far. Given the plasticity of the immune system and the diversity of organisms, it is highly probable that numerous other cells and molecules are still to be ascribed to the immune regulation process. Here, we report a general overview of how immune activation and regulation interact, based on the involvement of molecules and cells specifically dedicated to these processes. In addition, we discuss the role of TR3-56 lymphocytes as a new cellular candidate in the immune regulation landscape.
Collapse
Affiliation(s)
- Flavia Carriero
- Department of Sciences, University of Basilicata, 85100 Potenza, Italy; (F.C.); (R.M.); (V.B.)
| | - Valentina Rubino
- Department of Translational Medicine, University of Naples Federico II, 80131 Naples, Italy; (V.R.); (G.R.)
| | - Stefania Leone
- Hematopoietic Stem Cell Transplantation Unit, Azienda Ospedaliera A. Cardarelli, 80131 Naples, Italy;
| | - Rosangela Montanaro
- Department of Sciences, University of Basilicata, 85100 Potenza, Italy; (F.C.); (R.M.); (V.B.)
| | - Vincenzo Brancaleone
- Department of Sciences, University of Basilicata, 85100 Potenza, Italy; (F.C.); (R.M.); (V.B.)
| | - Giuseppina Ruggiero
- Department of Translational Medicine, University of Naples Federico II, 80131 Naples, Italy; (V.R.); (G.R.)
| | - Giuseppe Terrazzano
- Department of Sciences, University of Basilicata, 85100 Potenza, Italy; (F.C.); (R.M.); (V.B.)
| |
Collapse
|
17
|
Dou H, Brandon NR, Koper KE, Xu Y. Fingerprint of Circulating Immunocytes as Biomarkers for the Prognosis of Brain Inflammation and Neuronal Injury after Cardiac Arrest. ACS Chem Neurosci 2023; 14:4115-4127. [PMID: 37967214 DOI: 10.1021/acschemneuro.3c00397] [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] [Indexed: 11/17/2023] Open
Abstract
Cardiac arrest is one of the most dangerous health problems in the world. Outcome prognosis is largely based on cerebral performance categories determined by neurological evaluations. Few systemic tests are currently available to predict survival to hospital discharge. Here, we present the results from the preclinical studies of cardiac arrest and resuscitation (CAR) in mice to identify signatures of circulating immune cells as blood-derived biomarkers to predict outcomes after CAR. Two flow cytometry panels for circulating blood lymphocytes and myeloid-derived cells, respectively, were designed to correlate with neuroinflammation and neuronal and dendritic losses in the selectively vulnerable regions of bilateral hippocampi. We found that CD4+CD25+ regulatory T cells, CD11b+CD11c- and CD11b+Ly6C+Ly6G+ myeloid-derived cells, and cells positive for the costimulatory molecules CD80 and CD86 in the blood were correlated with activation of microglia and astrocytosis, and CD4+CD25+ T cells are additionally correlated with neuronal and dendritic losses. A fingerprint pattern of blood T cells and monocytes is devised as a diagnostic tool to predict CAR outcomes. Blood tests aimed at identifying these immunocyte patterns in cardiac arrest patients will guide future clinical trials to establish better prognostication tools to avoid unnecessary early withdrawal from life-sustaining treatment.
Collapse
Affiliation(s)
- Huanyu Dou
- Department of Molecular and Translational Medicine, Paul L. Foster School of Medicine, and Graduate School of Biomedical Sciences, Texas Tech University Health Science Center, El Paso, Texas 79905, United States
| | - Nicole R Brandon
- Departments of Anesthesiology and Perioperative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15213, United States
| | - Kerryann E Koper
- Departments of Anesthesiology and Perioperative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15213, United States
| | - Yan Xu
- Departments of Anesthesiology and Perioperative Medicine, Pharmacology and Chemical Biology, and Structural Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15213, United States
- Department of Physics and Astronomy, The Dietrich School of Arts and Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, United States
| |
Collapse
|
18
|
Hong Y, Chen P, Gao J, Lin Y, Chen L, Shang X. Sepsis-associated encephalopathy: From pathophysiology to clinical management. Int Immunopharmacol 2023; 124:110800. [PMID: 37619410 DOI: 10.1016/j.intimp.2023.110800] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 07/20/2023] [Accepted: 08/11/2023] [Indexed: 08/26/2023]
Abstract
Sepsis-associated encephalopathy, which presents as delirium and coma, is a significant complication of sepsis characterized by acute brain dysfunction. The presence of inflammatory pathological changes in the brain of sepsis patients and animal models has been recognized since the 1920 s, initially attributed to the entry of microbial toxins into the brain. In the early 2000 s, attention shifted towards the impact of oxidative stress, the cholinergic system, and cytokines on brain function following sepsis onset. More recently, sepsis-associated encephalopathy has been defined as a diffuse brain dysfunction not directly caused by pathogenic infection of the brain. Currently, there is no evidence-based standard for diagnosing sepsis-associated encephalopathy, and clinical management is primarily focused on symptomatic and supportive measures. This review aims to explore the pathophysiology of sepsis-associated encephalopathy and establish the connection between pathophysiological mechanisms and clinical characteristics. We hope that this work will spark the interest of researchers from various fields and contribute to the advancement of sepsis-associated encephalopathy research.
Collapse
Affiliation(s)
- Yixiao Hong
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China; The Third Department of Critical Care Medicine, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fujian Provincial Center for Critical Care Medicine, Fujian Provincial Key Laboratory of Critical Care Medicine, Fuzhou, China
| | - Peiling Chen
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China; The Third Department of Critical Care Medicine, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fujian Provincial Center for Critical Care Medicine, Fujian Provincial Key Laboratory of Critical Care Medicine, Fuzhou, China
| | - Jingqi Gao
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China; The Third Department of Critical Care Medicine, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fujian Provincial Center for Critical Care Medicine, Fujian Provincial Key Laboratory of Critical Care Medicine, Fuzhou, China
| | - Yingying Lin
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China; The Third Department of Critical Care Medicine, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fujian Provincial Center for Critical Care Medicine, Fujian Provincial Key Laboratory of Critical Care Medicine, Fuzhou, China
| | - Linfang Chen
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China; The Third Department of Critical Care Medicine, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fujian Provincial Center for Critical Care Medicine, Fujian Provincial Key Laboratory of Critical Care Medicine, Fuzhou, China
| | - Xiuling Shang
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China; The Third Department of Critical Care Medicine, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fujian Provincial Center for Critical Care Medicine, Fujian Provincial Key Laboratory of Critical Care Medicine, Fuzhou, China.
| |
Collapse
|
19
|
Ou Q, Power R, Griffin MD. Revisiting regulatory T cells as modulators of innate immune response and inflammatory diseases. Front Immunol 2023; 14:1287465. [PMID: 37928540 PMCID: PMC10623442 DOI: 10.3389/fimmu.2023.1287465] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 10/05/2023] [Indexed: 11/07/2023] Open
Abstract
Regulatory T cells (Treg) are known to be critical for the maintenance of immune homeostasis by suppressing the activation of auto- or allo-reactive effector T cells through a diverse repertoire of molecular mechanisms. Accordingly, therapeutic strategies aimed at enhancing Treg numbers or potency in the setting of autoimmunity and allogeneic transplants have been energetically pursued and are beginning to yield some encouraging outcomes in early phase clinical trials. Less well recognized from a translational perspective, however, has been the mounting body of evidence that Treg directly modulate most aspects of innate immune response under a range of different acute and chronic disease conditions. Recognizing this aspect of Treg immune modulatory function provides a bridge for the application of Treg-based therapies to common medical conditions in which organ and tissue damage is mediated primarily by inflammation involving myeloid cells (mononuclear phagocytes, granulocytes) and innate lymphocytes (NK cells, NKT cells, γδ T cells and ILCs). In this review, we comprehensively summarize pre-clinical and human research that has revealed diverse modulatory effects of Treg and specific Treg subpopulations on the range of innate immune cell types. In each case, we emphasize the key mechanistic insights and the evidence that Treg interactions with innate immune effectors can have significant impacts on disease severity or treatment. Finally, we discuss the opportunities and challenges that exist for the application of Treg-based therapeutic interventions to three globally impactful, inflammatory conditions: type 2 diabetes and its end-organ complications, ischemia reperfusion injury and atherosclerosis.
Collapse
Affiliation(s)
- Qifeng Ou
- Regenerative Medicine Institute (REMEDI) at CÚRAM SFI Research Centre for Medical Devices, School of Medicine, College of Medicine, Nursing and Health Sciences, University of Galway, Galway, Ireland
| | - Rachael Power
- Regenerative Medicine Institute (REMEDI) at CÚRAM SFI Research Centre for Medical Devices, School of Medicine, College of Medicine, Nursing and Health Sciences, University of Galway, Galway, Ireland
| | - Matthew D. Griffin
- Regenerative Medicine Institute (REMEDI) at CÚRAM SFI Research Centre for Medical Devices, School of Medicine, College of Medicine, Nursing and Health Sciences, University of Galway, Galway, Ireland
- Nephrology Department, Galway University Hospitals, Saolta University Healthcare Group, Galway, Ireland
| |
Collapse
|
20
|
Bi Y, Kong R, Peng Y, Yu H, Zhou Z. Umbilical cord blood and peripheral blood-derived regulatory T cells therapy: Progress in type 1 diabetes. Clin Immunol 2023; 255:109716. [PMID: 37544491 DOI: 10.1016/j.clim.2023.109716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 07/25/2023] [Accepted: 08/03/2023] [Indexed: 08/08/2023]
Abstract
Regulatory T cells (Tregs) are key regulators for the inflammatory response and play a role in maintaining the immune tolerance. Type 1 diabetes (T1D) is a relatively common autoimmune disease that results from the loss of immune tolerance to β-cell-associated antigens. Preclinical models have demonstrated the safety and efficacy of Tregs given in transplant rejection and autoimmune diseases such as T1D. Adoptive transfer of Tregs has been utilized in clinical trials for over a decade. However, the achievement of the adoptive transfer of Tregs therapy in clinical application remains challenging. In this review, we highlight the characterization of Tregs and compare the differences between umbilical cord blood and adult peripheral blood-derived Tregs. Additionally, we summarize conditional modifications in the expansion of Tregs in clinical trials, especially for the treatment of T1D. Finally, we discuss the existing technical challenges for Tregs in clinical trials for the treatment of T1D.
Collapse
Affiliation(s)
- Yuanjie Bi
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, Hunan Engineering Research Center of Cell Therapy for Diabetes, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Ran Kong
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, Hunan Engineering Research Center of Cell Therapy for Diabetes, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Yani Peng
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, Hunan Engineering Research Center of Cell Therapy for Diabetes, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Haibo Yu
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, Hunan Engineering Research Center of Cell Therapy for Diabetes, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China.
| | - Zhiguang Zhou
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, Hunan Engineering Research Center of Cell Therapy for Diabetes, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China.
| |
Collapse
|
21
|
Zhang QQ, Zhang WJ, Chang S. HDAC6 inhibition: a significant potential regulator and therapeutic option to translate into clinical practice in renal transplantation. Front Immunol 2023; 14:1168848. [PMID: 37545520 PMCID: PMC10401441 DOI: 10.3389/fimmu.2023.1168848] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Accepted: 05/30/2023] [Indexed: 08/08/2023] Open
Abstract
Histone deacetylase 6 (HDAC6), an almost exclusively cytoplasmic enzyme, plays an essential role in many biological processes and exerts its deacetylation-dependent/independent effects on a variety of target molecules, which has contributed to the flourishing growth of relatively isoform-specific enzyme inhibitors. Renal transplantation (RT) is one of the alternatively preferred treatments and the most cost-effective treatment approaches for the great majority of patients with end-stage renal disease (ESRD). HDAC6 expression and activity have recently been shown to be increased in kidney disease in a number of studies. To date, a substantial amount of validated studies has identified HDAC6 as a pivotal modulator of innate and adaptive immunity, and HDAC6 inhibitors (HDAC6i) are being developed and investigated for use in arrays of immune-related diseases, making HDAC6i a promising therapeutic candidate for the management of a variety of renal diseases. Based on accumulating evidence, HDAC6i markedly open up new avenues for therapeutic intervention to protect against oxidative stress-induced damage, tip the balance in favor of the generation of tolerance-related immune cells, and attenuate fibrosis by inhibiting multiple activations of cell profibrotic signaling pathways. Taken together, we have a point of view that targeting HDAC6 may be a novel approach for the therapeutic strategy of RT-related complications, including consequences of ischemia-reperfusion injury, induction of immune tolerance in transplantation, equilibrium of rejection, and improvement of chronic renal graft interstitial fibrosis after transplantation in patients. Herein, we will elaborate on the unique function of HDAC6, which focuses on therapeutical mechanism of action related to immunological events with a general account of the tantalizing potential to the clinic.
Collapse
Affiliation(s)
- Qian-qian Zhang
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China
| | - Wei-jie Zhang
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China
| | - Sheng Chang
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China
| |
Collapse
|
22
|
Hoffmann AD, Weinberg SE, Swaminathan S, Chaudhuri S, Almubarak HF, Schipma MJ, Mao C, Wang X, El-Shennawy L, Dashzeveg NK, Wei J, Mehl PJ, Shihadah LJ, Wai CM, Ostiguin C, Jia Y, D'Amico P, Wang NR, Luo Y, Demonbreun AR, Ison MG, Liu H, Fang D. Unique molecular signatures sustained in circulating monocytes and regulatory T cells in convalescent COVID-19 patients. Clin Immunol 2023; 252:109634. [PMID: 37150240 PMCID: PMC10162478 DOI: 10.1016/j.clim.2023.109634] [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: 02/20/2023] [Revised: 04/19/2023] [Accepted: 04/26/2023] [Indexed: 05/09/2023]
Abstract
Over two years into the COVID-19 pandemic, the human immune response to SARS-CoV-2 during the active disease phase has been extensively studied. However, the long-term impact after recovery, which is critical to advance our understanding SARS-CoV-2 and COVID-19-associated long-term complications, remains largely unknown. Herein, we characterized single-cell profiles of circulating immune cells in the peripheral blood of 100 patients, including convalescent COVID-19 and sero-negative controls. Flow cytometry analyses revealed reduced frequencies of both short-lived monocytes and long-lived regulatory T (Treg) cells within the patients who have recovered from severe COVID-19. sc-RNA seq analysis identifies seven heterogeneous clusters of monocytes and nine Treg clusters featuring distinct molecular signatures in association with COVID-19 severity. Asymptomatic patients contain the most abundant clusters of monocytes and Tregs expressing high CD74 or IFN-responsive genes. In contrast, the patients recovered from a severe disease have shown two dominant inflammatory monocyte clusters featuring S100 family genes: one monocyte cluster of S100A8 & A9 coupled with high HLA-I and another cluster of S100A4 & A6 with high HLA-II genes, a specific non-classical monocyte cluster with distinct IFITM family genes, as well as a unique TGF-β high Treg Cluster. The outpatients and seronegative controls share most of the monocyte and Treg clusters patterns with high expression of HLA genes. Surprisingly, while presumably short-lived monocytes appear to have sustained alterations over 4 months, the decreased frequencies of long-lived Tregs (high HLA-DRA and S100A6) in the outpatients restore over the tested convalescent time (≥ 4 months). Collectively, our study identifies sustained and dynamically altered monocytes and Treg clusters with distinct molecular signatures after recovery, associated with COVID-19 severity.
Collapse
Affiliation(s)
- Andrew D Hoffmann
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Sam E Weinberg
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Suchitra Swaminathan
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; Division of Rheumatology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Shuvam Chaudhuri
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Hannah Faisal Almubarak
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Matthew J Schipma
- NUseq Core Facility, Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Chengsheng Mao
- Division of Health and Biomedical Informatics, Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Xinkun Wang
- NUseq Core Facility, Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Lamiaa El-Shennawy
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Nurmaa K Dashzeveg
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Juncheng Wei
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Paul J Mehl
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Laura J Shihadah
- NUseq Core Facility, Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Ching Man Wai
- NUseq Core Facility, Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Carolina Ostiguin
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Yuzhi Jia
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Paolo D'Amico
- Division of Hematology and Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Neale R Wang
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Yuan Luo
- Division of Health and Biomedical Informatics, Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Alexis R Demonbreun
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Michael G Ison
- Division of Infectious Disease, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; Division of Organ Transplantation, Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; Respiratory Diseases Branch, Division of Microbiology and Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20892, USA.
| | - Huiping Liu
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; Division of Hematology and Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA.
| | - Deyu Fang
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA.
| |
Collapse
|
23
|
Wu W, Chen L, Jia G, Tang Q, Han B, Xia S, Jiang Q, Liu H. Inhibition of FGFR3 upregulates MHC-I and PD-L1 via TLR3/NF-kB pathway in muscle-invasive bladder cancer. Cancer Med 2023; 12:15676-15690. [PMID: 37283287 PMCID: PMC10417096 DOI: 10.1002/cam4.6172] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 05/16/2023] [Accepted: 05/19/2023] [Indexed: 06/08/2023] Open
Abstract
BACKGROUND Improving the potency of immune response is paramount among issues concerning immunotherapy of muscle-invasive bladder cancer (MIBC). METHODS On the basis of immune subtypes, we investigated possible molecular mechanisms involved in tumor immune escape in MIBC. According to the 312 immune-related genes, three MIBC immune subtypes were clustered. RESULTS Cluster 2 subtype is characterized by FGFR3 mutations and has a better clinical prognosis. However, the expression levels of MHC-I and immune checkpoints genes were the lowest, indicating that this subtype is subject to immune escape and has a low response rate to immunotherapy. Bioinformatics analysis and immunofluorescence staining of clinical samples revealed that the FGFR3 is involved in the immune escape in MIBC. Besides, after FGFR3 knockout with siRNA in RT112 and UMUC14 cells, the TLR3/NF-kB pathway was significantly activated and was accompanied by upregulation of MHC-I and PD-L1 gene expression. Furthermore, the use of TLR3 agonists poly(I:C) can further improve the effect. CONCLUSION Together, our results suggest that FGFR3 might involve in immunosuppression by inhibition of NF-kB pathway in BC. Considering that TLR3 agonists are currently approved for clinical treatment as immunoadjuvants, our study might provide more insights for improving the efficacy of immunotherapy in MIBC.
Collapse
Affiliation(s)
- WenBo Wu
- Department of UrologyShanghai General HospitalShanghaiChina
- Shanghai JiaoTong University School of MedicineShanghaiChina
| | - Lei Chen
- Department of UrologyShanghai General HospitalShanghaiChina
| | - GaoZhen Jia
- Department of UrologyShanghai General HospitalShanghaiChina
| | - QiLin Tang
- Department of UrologyShanghai General HospitalShanghaiChina
- Shanghai JiaoTong University School of MedicineShanghaiChina
| | - BangMin Han
- Department of UrologyShanghai General HospitalShanghaiChina
| | - ShuJie Xia
- Department of UrologyShanghai General HospitalShanghaiChina
| | - Qi Jiang
- Department of UrologyShanghai General HospitalShanghaiChina
| | - HaiTao Liu
- Department of UrologyShanghai General HospitalShanghaiChina
- Shanghai JiaoTong University School of MedicineShanghaiChina
| |
Collapse
|
24
|
Chen H, Li Y, Wang J, Zheng T, Wu C, Cui M, Feng Y, Ye H, Dong Z, Dang Y. Plant Polyphenols Attenuate DSS-induced Ulcerative Colitis in Mice via Antioxidation, Anti-inflammation and Microbiota Regulation. Int J Mol Sci 2023; 24:10828. [PMID: 37446006 DOI: 10.3390/ijms241310828] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 06/11/2023] [Accepted: 06/16/2023] [Indexed: 07/15/2023] Open
Abstract
The pathogenesis of ulcerative colitis (UC) is associated with inflammation, oxidative stress, and gut microbiota imbalance. Although most researchers have demonstrated the antioxidant bioactivity of the phenolic compounds in plants, their UC-curing ability and underlying mechanisms still need to be further and adequately explored. Herein, we studied the antioxidation-structure relationship of several common polyphenols in plants including gallic acid, proanthocyanidin, ellagic acid, and tannic acid. Furthermore, the in vivo effects of the plant polyphenols on C57BL/6 mice with dextran-sulfate-sodium-induced UC were evaluated and the action mechanisms were explored. Moreover, the interplay of several mechanisms was determined. The higher the number of phenolic hydroxyl groups, the stronger the antioxidant activity. All polyphenols markedly ameliorated the symptoms and pathological progression of UC in mice. Furthermore, inflammatory cytokine levels were decreased and the intestinal barrier was repaired. The process was regulated by the antioxidant-signaling pathway of nuclear-erythroid 2-related factor 2. Moreover, the diversity of the intestinal microbiota, Firmicutes-to-Bacteroides ratio, and relative abundance of beneficial bacteria were increased. An interplay was observed between microbiota regulation and oxidative stress, immunity, and inflammatory response. Furthermore, intestinal barrier repair was found to be correlated with inflammatory responses. Our study results can form a basis for comprehensively developing plant-polyphenol-related medicinal products.
Collapse
Affiliation(s)
- Huan Chen
- Drug Delivery Research Center, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100193, China
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Hebei Medical University, Shijiazhuang 050017, China
| | - Ying Li
- Drug Delivery Research Center, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100193, China
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100094, China
- Key Laboratory of New Drug Discovery Based on Classic Chinese Medicine Prescription, Beijing 100700, China
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing 100700, China
| | - Jinrui Wang
- Drug Delivery Research Center, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100193, China
| | - Tingting Zheng
- Drug Delivery Research Center, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100193, China
| | - Chenyang Wu
- Drug Delivery Research Center, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100193, China
| | - Mengyao Cui
- Drug Delivery Research Center, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100193, China
| | - Yifan Feng
- Drug Delivery Research Center, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100193, China
| | - Hanyi Ye
- Drug Delivery Research Center, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100193, China
| | - Zhengqi Dong
- Drug Delivery Research Center, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100193, China
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100094, China
- Key Laboratory of New Drug Discovery Based on Classic Chinese Medicine Prescription, Beijing 100700, China
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing 100700, China
| | - Yunjie Dang
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Hebei Medical University, Shijiazhuang 050017, China
| |
Collapse
|
25
|
Stroukov W, Mastronicola D, Albany CJ, Catak Z, Lombardi G, Scottà C. OMIP-090: A 20-parameter flow cytometry panel for rapid analysis of cell diversity and homing capacity in human conventional and regulatory T cells. Cytometry A 2023; 103:362-367. [PMID: 36740883 PMCID: PMC10952450 DOI: 10.1002/cyto.a.24720] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 01/11/2023] [Accepted: 01/21/2023] [Indexed: 02/07/2023]
Abstract
The panel was developed and optimized for monitoring changes in homing capacity and functional diversity of human CD4+ conventional and regulatory T cell subsets. The analysis was based on expression of only surface markers in freshly isolated peripheral blood mononuclear cells (PBMCs) to reduce at minimum any alteration due to permeabilization or freezing/thawing procedures. We included markers to assess the distribution of naïve and memory populations based on the expression of CD45RA, CCR7, CD25, CD28 and CD95 in both conventional and regulatory T cells. The identification of major functional subsets was performed using CCR4, CCR6, CCR10, CXCR3 and CXCR5. Homing capacity of these subsets to skin, airway tract, gut and inflammatory lesions could finally be assessed with the markers CLA, CCR3, CCR5 and integrin β7. The panel was tested on freshly isolated PBMCs from healthy donors and patients with allergic rhinitis or autoimmune disorders.
Collapse
Affiliation(s)
- Wladislaw Stroukov
- “Peter Gorer” Department of Immunobiology, School of Immunology & Microbiological SciencesKing's College LondonLondonUK
| | - Daniela Mastronicola
- “Peter Gorer” Department of Immunobiology, School of Immunology & Microbiological SciencesKing's College LondonLondonUK
| | - Caraugh Jane Albany
- “Peter Gorer” Department of Immunobiology, School of Immunology & Microbiological SciencesKing's College LondonLondonUK
- British Heart Foundation Centre, School of Cardiovascular Medicine and SciencesKing's College LondonLondonUK
| | - Zeynep Catak
- “Peter Gorer” Department of Immunobiology, School of Immunology & Microbiological SciencesKing's College LondonLondonUK
| | - Giovanna Lombardi
- “Peter Gorer” Department of Immunobiology, School of Immunology & Microbiological SciencesKing's College LondonLondonUK
| | - Cristiano Scottà
- “Peter Gorer” Department of Immunobiology, School of Immunology & Microbiological SciencesKing's College LondonLondonUK
| |
Collapse
|
26
|
Cheng Y, Li L, Wei X, Xu F, Huang X, Qi F, Zhang Y, Li X. HNRNPC suppresses tumor immune microenvironment by activating Treg cells promoting the progression of prostate cancer. Cancer Sci 2023; 114:1830-1845. [PMID: 36718950 PMCID: PMC10154801 DOI: 10.1111/cas.15745] [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: 11/01/2022] [Revised: 01/20/2023] [Accepted: 01/27/2023] [Indexed: 02/01/2023] Open
Abstract
Immune microenvironment could affect the biological progress in prostate cancer (PCa) through N6 methyl adenosine (m6A) methylation. The purpose of this study was to investigate the crosstalk between m6A methylation and immune microenvironment and explore potential biomarkers to improve the immunotherapeutic response. Firstly, according to 11 differentially expressed m6A genes between normal and tumor samples, PCa patients were divided into immune microenvironment subtype 1 (IMS1) and IMS2 based on m6A gene profiles extracted from The Cancer Genome Atlas (TCGA) database. IMS2 showed an immune "cold" phenotype with worse prognoses, and HNRNPC was identified as the biomarker of IMS2 by the protein-protein interaction network. Furthermore, through bioinformatics analyses and in vitro experiments, we found that HNRNPC-high patients showed a suppressive immune-infiltrating tumor microenvironment with a higher infiltration of regulatory T (Treg) cells. Finally, we cocultured transfected PCa cells with peripheral blood mononuclear cells (PBMC) and verified that HNRNPC inhibits tumor immunity by elevating the activation of Treg cells and suppression of effector CD8 T cell. In conclusion, we identified a "cold" immune phenotype in PCa, and HNRNPC regulating the activation of Treg cells. Activation of the immune microenvironment through targeting HNRNPC may be a potential therapeutic option for advanced PCa.
Collapse
Affiliation(s)
- Yifei Cheng
- Department of Urologic SurgeryJiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & Affiliated Cancer Hospital of Nanjing Medical UniversityNanjingChina
- Department of UrologyThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| | - Lu Li
- State Key Laboratory of Translational Medicine and Innovative Drug DevelopmentJiangsu Simcere Diagnostics Co., Ltd.NanjingChina
| | - Xiyi Wei
- Department of UrologyThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
- The State Key Lab of ReproductiveDepartment of UrologyThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| | - Fan Xu
- Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & Affiliated Cancer Hospital of Nanjing Medical UniversityNanjingChina
| | - Xiaochen Huang
- Department of PathologyJiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & Affiliated Cancer Hospital of Nanjing Medical UniversityNanjingChina
| | - Feng Qi
- Department of Urologic SurgeryJiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & Affiliated Cancer Hospital of Nanjing Medical UniversityNanjingChina
| | - Yanyan Zhang
- Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & Affiliated Cancer Hospital of Nanjing Medical UniversityNanjingChina
| | - Xiao Li
- Department of Urologic SurgeryJiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & Affiliated Cancer Hospital of Nanjing Medical UniversityNanjingChina
- Department of Scientific ResearchJiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & Affiliated Cancer Hospital of Nanjing Medical UniversityNanjingChina
| |
Collapse
|
27
|
Liu Y, Hoang TK, Park ES, Freeborn J, Okeugo B, Tran DQ, Rhoads JM. Probiotic-educated Tregs are more potent than naïve Tregs for immune tolerance in stressed new-born mice. Benef Microbes 2023; 14:73-84. [PMID: 36815493 PMCID: PMC10124588 DOI: 10.3920/bm2022.0095] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/24/2023]
Abstract
When new-born mice are subjected to acute maternal separation stress, cow-milk based formula feeding, and brief recurrent hypoxia with cold stress, they develop gut inflammation similar to the phenotype of neonatal necrotizing enterocolitis, characterised by an increase in gut mucosal effector T (Teffs) and reduced Foxp3+ regulatory T (Tregs) cells. The imbalance can be prevented by probiotic Limosilactobacillus reuteri DSM 17938 (LR 17938). We hypothesised that LR 17938 could potentiate a tolerogenic function of Tregs. To analyse whether LR 17938 can educate Tregs to improve their tolerogenic potency during neonatal stress, we isolated T cells (Tregs and Teffs) from 'donor' mice fed with either LR 17938 (107 cfu) or control media. The cells were adoptively transferred (AT) by intraperitoneal injection (5 × 105 cells/mouse) to new-born (d5) recipient mice. Mice were then separated from their dams, fed formula by gavage, and exposed to hypoxia and cold stress (NeoStress) for 4 days. We analysed the percentage of Tregs in CD4+T helper cells in the intestine (INT) and mesenteric lymph nodes (MLN) of recipient mice. We found that: (1) the percentage of Tregs in the INT and MLN following NeoStress were significantly reduced compared to dam-fed unstressed mice; (2) AT of either naïve Tregs or LR-educated Tregs to mice with Neostress increased the percentage of Tregs in the INT and MLN compared to the percentage in NeoStress mice without Treg treatment; however, LR-educated Tregs increased the Tregs significantly more than naïve Tregs; and (3) AT of LR-educated Tregs reduced pro-inflammatory CD44+Foxp3-NonTregs and inflammatory CX3CR1+ dendritic cells in the intestinal mucosa of NeoStress mice. In conclusion, adoptive transfer of Tregs promotes the generation of and/or migration of endogenous Tregs in the intestinal mucosa of recipient mice. Importantly, probiotic-educated Tregs are more potent than naïve Tregs to enhance immune tolerance following neonatal stress.
Collapse
Affiliation(s)
- Y Liu
- Department of Pediatrics, Division of Gastroenterology, McGovern Medical School, University of Texas Health Science Center at Houston, 6431 Fannin Street, MSB 3.137, Houston, TX 77030, USA
| | - T K Hoang
- Department of Pediatrics, Division of Gastroenterology, McGovern Medical School, University of Texas Health Science Center at Houston, 6431 Fannin Street, MSB 3.137, Houston, TX 77030, USA
| | - E S Park
- Department of Pediatrics, Division of Gastroenterology, McGovern Medical School, University of Texas Health Science Center at Houston, 6431 Fannin Street, MSB 3.137, Houston, TX 77030, USA
| | - J Freeborn
- Department of Pediatrics, Division of Gastroenterology, McGovern Medical School, University of Texas Health Science Center at Houston, 6431 Fannin Street, MSB 3.137, Houston, TX 77030, USA
| | - B Okeugo
- Department of Pediatrics, Division of Gastroenterology, McGovern Medical School, University of Texas Health Science Center at Houston, 6431 Fannin Street, MSB 3.137, Houston, TX 77030, USA
| | - D Q Tran
- Department of Pediatrics, Division of Gastroenterology, McGovern Medical School, University of Texas Health Science Center at Houston, 6431 Fannin Street, MSB 3.137, Houston, TX 77030, USA
| | - J M Rhoads
- Department of Pediatrics, Division of Gastroenterology, McGovern Medical School, University of Texas Health Science Center at Houston, 6431 Fannin Street, MSB 3.137, Houston, TX 77030, USA
| |
Collapse
|
28
|
Zhao Y, Zheng X, Li M, Zhao J, Wang X, Zhu H. ADAR1 improved Treg cell function through the miR-21b/Foxp3 axis and inhibits the progression of acute graft-versus-host disease after allogeneic hematopoietic stem cell transplantation. Int Immunopharmacol 2023; 115:109620. [PMID: 36577155 DOI: 10.1016/j.intimp.2022.109620] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Revised: 12/04/2022] [Accepted: 12/17/2022] [Indexed: 12/27/2022]
Abstract
Th17/Treg equilibrium towards the pro-inflammatory Th17 side contributes greatly to the rejection during allogeneic hematopoietic stem cell transplantation (allo-HSCT). Forkhead box P3 (Foxp3) is important in the pathogenic conversion between Th17 and Treg cells. However, how Foxp3 expression was regulated is largely unknown. Here, we investigated the role of RNA-editing enzyme ADAR1 in Foxp3-mediated Th17/Treg imbalance and progression of acute graft-versus-host disease (aGVHD), a most serious complication in patients received allo-HSCT. Th1, Th17 and Treg cells were respectively isolated from peripheral blood CD4 + T cells of allo-HSCT patients, and we found that proportions of Th1 and Th17 were markedly increased, while Treg proportion was significantly decreased in aGVHD patients post transplantation compared with non-aGVHD patients, accompanied by decreased ADAR1 and increased miR-21b levels. RNA-immunoprecipitation (RIP) combined with gain- and loss-of-function experiments demonstrated that ADAR1 improved Treg cell functions and negatively regulated the production of miR-21b, a Foxp3-targeting miRNA. Inhibition of miR-21b improved Treg functions, and Foxp3 knockdown could eliminate the effect of miR-21b inhibition or ADAR1 overexpression on Treg function. Finally, an aGVHD mouse model was established and Ad-O/E-ADAR1 was injected into aGVHD mice to verify the effect of ADAR1 on aGVHD progression in vivo. The results showed that ADAR1 overexpression decreased Th17 proportion and increased Treg proportion in aGVHD mice and obviously improved tissue necrosis and reticular structure of aGVHD liver and lung in vivo. Collectively, ADAR1 suppresses miR-21b production and improves Foxp3-mediated Treg cell function to inhibit the progression of aGVHD after allo-HSCT.
Collapse
Affiliation(s)
- Yanru Zhao
- Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, PR China
| | - Xiaoyan Zheng
- Department of Hematology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, PR China
| | - Miaojing Li
- Department of Hematology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, PR China
| | - Jing Zhao
- Department of Hematology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, PR China
| | - Xiaoning Wang
- Department of Hematology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, PR China
| | - Huachao Zhu
- Department of Hematology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, PR China.
| |
Collapse
|
29
|
Povoleri GAM, Fleskens V, Taams LS. Isolation and Functional Characterization of Regulatory CD4+ T Cells from the Inflamed Joints of Patients with Rheumatoid Arthritis. Methods Mol Biol 2023; 2559:189-203. [PMID: 36180634 DOI: 10.1007/978-1-0716-2647-4_13] [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] [Indexed: 06/16/2023]
Abstract
Regulatory T cells play a critical role in maintaining immune homeostasis and in preventing and controlling unwanted immune activation. These cells are often studied in the context of human peripheral blood, but can also be isolated from other biofluids. Here we describe methods for the isolation and functional characterization of human CD4+ CD25hi CD127low regulatory T cells from the synovial fluid of patients with inflammatory arthritis.
Collapse
Affiliation(s)
- Giovanni A M Povoleri
- Centre for Inflammation Biology and Cancer Immunology (CIBCI), Department of Inflammation Biology, School of Immunology & Microbial Sciences, King's College London, London, UK
| | - Veerle Fleskens
- Centre for Inflammation Biology and Cancer Immunology (CIBCI), Department of Inflammation Biology, School of Immunology & Microbial Sciences, King's College London, London, UK
| | - Leonie S Taams
- Centre for Inflammation Biology and Cancer Immunology (CIBCI), Department of Inflammation Biology, School of Immunology & Microbial Sciences, King's College London, London, UK.
| |
Collapse
|
30
|
Tauber PA, Kratzer B, Schatzlmaier P, Smole U, Köhler C, Rausch L, Kranich J, Trapin D, Neunkirchner A, Zabel M, Jutz S, Steinberger P, Gadermaier G, Brocker T, Stockinger H, Derdak S, Pickl WF. The small molecule inhibitor BX-795 uncouples IL-2 production from inhibition of Th2 inflammation and induces CD4 + T cells resembling iTreg. Front Immunol 2023; 14:1094694. [PMID: 37090735 PMCID: PMC10117943 DOI: 10.3389/fimmu.2023.1094694] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 03/06/2023] [Indexed: 04/25/2023] Open
Abstract
Background Treg cells have been shown to be an important part of immune-homeostasis and IL-2 which is produced upon T cell receptor (TCR)-dependent activation of T lymphocytes has been demonstrated to critically participate in Treg development. Objective To evaluate small molecule inhibitors (SMI) for the identification of novel IL-2/Treg enhancing compounds. Materials and methods We used TCR-dependent and allergen-specific cytokine secretion of human and mouse T cells, next generation messenger ribonucleic acid sequencing (RNA-Seq) and two different models of allergic airway inflammation to examine lead SMI-compounds. Results We show here that the reported 3-phosphoinositide dependent kinase-1 (PDK1) SMI BX-795 increased IL-2 in culture supernatants of Jurkat E6-1 T cells, human peripheral blood mononuclear cells (hPBMC) and allergen-specific mouse T cells upon TCR-dependent and allergen-specific stimulation while concomitantly inhibiting Th2 cytokine secretion. RNA-Seq revealed that the presence of BX-795 during allergen-specific activation of T cells induces a bona fide Treg cell type highly similar to iTreg but lacking Foxp3 expression. When applied in mugwort pollen and house dust mite extract-based models of airway inflammation, BX-795 significantly inhibited Th2 inflammation including expression of Th2 signature transcription factors and cytokines and influx into the lungs of type 2-associated inflammatory cells such as eosinophils. Conclusions BX-795 potently uncouples IL-2 production from Th2 inflammation and induces Th-IL-2 cells, which highly resemble induced (i)Tregs. Thus, BX-795 may be a useful new compound for the treatment of allergic diseases.
Collapse
Affiliation(s)
- Peter A. Tauber
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Bernhard Kratzer
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Philipp Schatzlmaier
- Institute of Hygiene and Applied Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Ursula Smole
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Cordula Köhler
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Lisa Rausch
- Institute for Immunology, Biomedical Center (BMC), Faculty of Medicine, Ludwig Maximilian University (LMU) Munich, Munich, Germany
| | - Jan Kranich
- Institute for Immunology, Biomedical Center (BMC), Faculty of Medicine, Ludwig Maximilian University (LMU) Munich, Munich, Germany
| | - Doris Trapin
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Alina Neunkirchner
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Maja Zabel
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Sabrina Jutz
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Peter Steinberger
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | | | - Thomas Brocker
- Institute for Immunology, Biomedical Center (BMC), Faculty of Medicine, Ludwig Maximilian University (LMU) Munich, Munich, Germany
| | - Hannes Stockinger
- Institute of Hygiene and Applied Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Sophia Derdak
- Core Facilities, Medical University of Vienna, Vienna, Austria
| | - Winfried F. Pickl
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
- Karl Landsteiner University of Healthcare, Krems, Austria
- *Correspondence: Winfried F. Pickl,
| |
Collapse
|
31
|
Yu Y, Bai H, Wu F, Chen J, Li B, Li Y. Tissue adaptation of regulatory T cells in adipose tissue. Eur J Immunol 2022; 52:1898-1908. [PMID: 36369886 DOI: 10.1002/eji.202149527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 08/05/2022] [Accepted: 10/27/2022] [Indexed: 11/15/2022]
Abstract
Foxp3+ regulatory T (Treg) cells critically suppress over-activated immune responses and therefore maintain immune homeostasis. Adipose tissue-resident Treg (AT Treg) cells are known for modulating immunity and metabolism in adipose tissue microenvironment through various physiological signals, as well as their heterogeneous subsets, which potentially play disparate roles in aging and obesity. Recent single-cell studies of Treg cells have revealed specialized trajectories of their tissue adaptation and development in lymphoid tissues and at barrier sites. Here, we reviewed a T Cell Receptor (TCR)-primed environmental cue-boosted model of adipose Treg cells' tissue adaptation, especially in response to IL-33, IFN-α, insulin, and androgen signals, which trigger sophisticated transcriptional cascades and ultimately establish unique transcriptional modules in adipose Treg cell subsets. In addition, we further discuss potential therapeutic strategies against aging and obesity by blocking detrimental environmental cues, strengthening the functions of specific AT Treg subsets and modifying the communications between AT Treg subsets and adipocytes.
Collapse
Affiliation(s)
- Yimeng Yu
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Department of Respiratory and Critical Care Medicine of Ruijin Hospital, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hongyu Bai
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Department of Respiratory and Critical Care Medicine of Ruijin Hospital, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fenglin Wu
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Department of Respiratory and Critical Care Medicine of Ruijin Hospital, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jieqiong Chen
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Department of Respiratory and Critical Care Medicine of Ruijin Hospital, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bin Li
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Department of Respiratory and Critical Care Medicine of Ruijin Hospital, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yangyang Li
- Unit of Immune and Metabolic Regulation, School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| |
Collapse
|
32
|
Agosto‐Burgos C, Wu EY, Iannone MA, Hu Y, Hogan SL, Henderson CD, Kennedy KB, Blazek L, Herrera CA, Munson D, Falk RJ, Ciavatta DJ, Free ME. The frequency of Treg subsets distinguishes disease activity in ANCA vasculitis. Clin Transl Immunology 2022; 11:e1428. [PMID: 36381498 PMCID: PMC9652144 DOI: 10.1002/cti2.1428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 10/20/2022] [Accepted: 10/21/2022] [Indexed: 11/13/2022] Open
Abstract
Objectives T regulatory cells (Tregs) are a heterogeneous group of immunoregulatory cells that dampen self-harming immune responses and prevent the development of autoimmune diseases. In anti-neutrophil cytoplasmic autoantibody (ANCA) vasculitis, Tregs possess diminished suppressive capacity, which has been attributed to the expression of a FOXP3 splice-variant lacking exon 2 in T cells (FOXP3Δ2 CD4+ T cells). However, the suppressive capacity of Tregs varies between subsets. We evaluated the frequency of Treg subsets in ANCA vasculitis as a potential explanation for diminished suppressive capacity. Methods We developed a custom mass cytometry panel and performed deep immune profiling of Tregs in healthy controls, patients with active disease and in remission. Using these data, we performed multidimensional reduction and discriminant analysis to identify associations between Treg subsets and disease activity. Results Total Tregs were expanded in ANCA vasculitis, which was associated with remission and the administration of rituximab and/or prednisone. The frequency of FOXP3Δ2 CD4+ T cells did not distinguish disease activity and this population had high expression levels of CD127 and lacked both CD25 and Helios, suggesting that they are not conventional Tregs. The frequency of CXCR3+, CD103+ and CCR7+ Tregs distinguished disease activity, and the combination of the frequency of these three Treg subsets segregated active patients from patients in remission and healthy controls. From these three subsets, the frequency of CXCR3+ Tregs distinguished patients with active disease with renal involvement. Conclusion Treg heterogeneity can discriminate disease activity and should be explored as a biomarker of disease activity in ANCA vasculitis.
Collapse
Affiliation(s)
- Christian Agosto‐Burgos
- Division of Nephrology and Hypertension, Department of Medicine, UNC Kidney CenterUniversity of North Carolina at Chapel HillChapel HillNCUSA
- Department of Pathology and Laboratory MedicineUniversity of North Carolina at Chapel HillChapel HillNCUSA
| | - Eveline Y Wu
- Division of Pediatric Allergy, Immunology and Rheumatology, Department of PediatricsUniversity of North CarolinaChapel HillNCUSA
| | - Marie A Iannone
- Lineberger Comprehensive Cancer CenterUniversity of North Carolina at Chapel HillChapel HillNCUSA
| | - Yichun Hu
- Division of Nephrology and Hypertension, Department of Medicine, UNC Kidney CenterUniversity of North Carolina at Chapel HillChapel HillNCUSA
| | - Susan L Hogan
- Division of Nephrology and Hypertension, Department of Medicine, UNC Kidney CenterUniversity of North Carolina at Chapel HillChapel HillNCUSA
| | - Candace D Henderson
- Division of Nephrology and Hypertension, Department of Medicine, UNC Kidney CenterUniversity of North Carolina at Chapel HillChapel HillNCUSA
| | - Kristin B Kennedy
- Division of Nephrology and Hypertension, Department of Medicine, UNC Kidney CenterUniversity of North Carolina at Chapel HillChapel HillNCUSA
| | - Lauren Blazek
- Division of Nephrology and Hypertension, Department of Medicine, UNC Kidney CenterUniversity of North Carolina at Chapel HillChapel HillNCUSA
| | - Carolina A Herrera
- Division of Nephrology and Hypertension, Department of Medicine, UNC Kidney CenterUniversity of North Carolina at Chapel HillChapel HillNCUSA
- Department of Pathology and Laboratory MedicineUniversity of North Carolina at Chapel HillChapel HillNCUSA
| | - Dominique Munson
- Division of Nephrology and Hypertension, Department of Medicine, UNC Kidney CenterUniversity of North Carolina at Chapel HillChapel HillNCUSA
- Department of Pathology and Laboratory MedicineUniversity of North Carolina at Chapel HillChapel HillNCUSA
| | - Ronald J Falk
- Division of Nephrology and Hypertension, Department of Medicine, UNC Kidney CenterUniversity of North Carolina at Chapel HillChapel HillNCUSA
- Department of MedicineUniversity of North Carolina at Chapel HillChapel HillNCUSA
| | - Dominic J Ciavatta
- Division of Nephrology and Hypertension, Department of Medicine, UNC Kidney CenterUniversity of North Carolina at Chapel HillChapel HillNCUSA
- Department of GeneticsUniversity of North Carolina at Chapel HillChapel HillNCUSA
| | - Meghan E Free
- Division of Nephrology and Hypertension, Department of Medicine, UNC Kidney CenterUniversity of North Carolina at Chapel HillChapel HillNCUSA
- Department of Pathology and Laboratory MedicineUniversity of North Carolina at Chapel HillChapel HillNCUSA
- Department of MedicineUniversity of North Carolina at Chapel HillChapel HillNCUSA
| |
Collapse
|
33
|
Osborne DG, Domenico J, Fujita M. Expression of IL-37 Induces a Regulatory T-Cell-like Phenotype and Function in Jurkat Cells. Cells 2022; 11:2565. [PMID: 36010641 PMCID: PMC9406943 DOI: 10.3390/cells11162565] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 08/10/2022] [Accepted: 08/16/2022] [Indexed: 11/17/2022] Open
Abstract
The anti-inflammatory cytokine interleukin-37 (IL-37) plays a key role in inhibiting innate and adaptive immunity. Past results have shown that IL-37 is elevated in human Treg cells compared to other T cell subsets and contributes to enhancing the Treg transcription factor, forkhead box protein P3 (FOXP3). However, it is unknown if ectopic expression of IL-37 in non-Treg CD4+ T cells can lead to the development of Treg phenotype and function. In the present study, we used a PrimeFlow® RNA assay and confirmed elevated IL37 expression in human Treg cells. We then stably transfected the non-Treg CD4+ T cell leukemia cell line, E6 Jurkat cells, with IL37 and found significant induction of the Treg phenotype. These IL-37-expressing Jurkat cells had elevated CTLA-4 and FOXP3 and produced IL-10. In conjunction with the Treg phenotype, IL-37-expressing Jurkat cells suppressed T cell activation/proliferation, comparable to human primary Treg cells. The creation of this stable human Treg-like cell line has the potential to provide further assistance for in vitro studies of human Treg cells, as it is more convenient than the use of primary human Treg cells. Furthermore, it provides insights into Treg cell biology and function.
Collapse
Affiliation(s)
- Douglas Grant Osborne
- Department of Dermatology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Joanne Domenico
- Department of Dermatology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Mayumi Fujita
- Department of Dermatology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Department of Veterans Affairs Medical Center, VA Eastern Colorado Health Care System, Aurora, CO 80045, USA
| |
Collapse
|
34
|
Schlöder J, Shahneh F, Schneider FJ, Wieschendorf B. Boosting regulatory T cell function for the treatment of autoimmune diseases – That’s only half the battle! Front Immunol 2022; 13:973813. [PMID: 36032121 PMCID: PMC9400058 DOI: 10.3389/fimmu.2022.973813] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 07/18/2022] [Indexed: 01/04/2023] Open
Abstract
Regulatory T cells (Treg) represent a subset of specialized T cells that are essential for the regulation of immune responses and maintenance of peripheral tolerance. Once activated, Treg exert powerful immunosuppressive properties, for example by inhibiting T cell-mediated immune responses against self-antigens, thereby protecting our body from autoimmunity. Autoimmune diseases such as multiple sclerosis, rheumatoid arthritis or systemic lupus erythematosus, exhibit an immunological imbalance mainly characterized by a reduced frequency and impaired function of Treg. In addition, there has been increasing evidence that – besides Treg dysfunction – immunoregulatory mechanisms fail to control autoreactive T cells due to a reduced responsiveness of T effector cells (Teff) for the suppressive properties of Treg, a process termed Treg resistance. In order to efficiently treat autoimmune diseases and thus fully induce immunological tolerance, a combined therapy aimed at both enhancing Treg function and restoring Teff responsiveness could most likely be beneficial. This review provides an overview of immunomodulating drugs that are currently used to treat various autoimmune diseases in the clinic and have been shown to increase Treg frequency as well as Teff sensitivity to Treg-mediated suppression. Furthermore, we discuss strategies on how to boost Treg activity and function, and their potential use in the treatment of autoimmunity. Finally, we present a humanized mouse model for the preclinical testing of Treg-activating substances in vivo.
Collapse
Affiliation(s)
- Janine Schlöder
- Department of Dermatology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
- ActiTrexx GmbH, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
- *Correspondence: Janine Schlöder,
| | - Fatemeh Shahneh
- Department of Dermatology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Franz-Joseph Schneider
- Department of Dermatology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
- ActiTrexx GmbH, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Björn Wieschendorf
- Department of Dermatology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
- ActiTrexx GmbH, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| |
Collapse
|
35
|
Matsuda M, Terada T, Kitatani K, Kawata R, Nabe T. Roles of type 1 regulatory T (Tr1) cells in allergen-specific immunotherapy. FRONTIERS IN ALLERGY 2022; 3:981126. [PMID: 35991310 PMCID: PMC9381954 DOI: 10.3389/falgy.2022.981126] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 07/20/2022] [Indexed: 12/03/2022] Open
Abstract
Allergen-specific immunotherapy (AIT) is the only causative treatment for allergic diseases by modification of the immune response to allergens. A key feature of AIT is to induce immunotolerance to allergens by generating antigen-specific regulatory T (Treg) cells in allergic patients. Type 1 regulatory T (Tr1) cells and forkhead box protein 3 (Foxp3)-expressing Treg cells are well known among Treg cell subsets. Foxp3 was identified as a master transcription factor of Treg cells, and its expression is necessary for their suppressive activity. In contrast to Foxp3+ Treg cells, the master transcription factor of Tr1 cells has not been elucidated. Nevertheless, Tr1 cells are generally considered as a distinct subset of Treg cells induced in the periphery during antigen exposure in tolerogenic conditions and can produce large amounts of anti-inflammatory cytokines such as interleukin-10 and transforming growth factor-β, followed by down-regulation of the function of effector immune cells independently of Foxp3 expression. Since the discovery of Tr1 cells more than 20 years ago, research on Tr1 cells has expanded our understanding of the mechanism of AIT. Although the direct precursors and true identity of these cells continues to be disputed, we and others have demonstrated that Tr1 cells are induced in the periphery by AIT, and the induced cells are re-activated by antigens, followed by suppression of allergic symptoms. In this review, we discuss the immune mechanisms for the induction of Tr1 cells by AIT and the immune-suppressive roles of Tr1 cells in AIT.
Collapse
Affiliation(s)
- Masaya Matsuda
- Laboratory of Immunopharmacology, Faculty of Pharmaceutical Sciences, Setsunan University, Hirakata, Japan
| | - Tetsuya Terada
- Department of Otolaryngology, Head & Neck Surgery, Osaka Medical and Pharmaceutical University, Takatsuki, Japan
| | - Kazuyuki Kitatani
- Laboratory of Immunopharmacology, Faculty of Pharmaceutical Sciences, Setsunan University, Hirakata, Japan
| | - Ryo Kawata
- Department of Otolaryngology, Head & Neck Surgery, Osaka Medical and Pharmaceutical University, Takatsuki, Japan
| | - Takeshi Nabe
- Laboratory of Immunopharmacology, Faculty of Pharmaceutical Sciences, Setsunan University, Hirakata, Japan
- Correspondence: Takeshi Nabe
| |
Collapse
|
36
|
Brown ME, Peters LD, Hanbali SR, Arnoletti JM, Sachs LK, Nguyen KQ, Carpenter EB, Seay HR, Fuhrman CA, Posgai AL, Shapiro MR, Brusko TM. Human CD4 +CD25 +CD226 - Tregs Demonstrate Increased Purity, Lineage Stability, and Suppressive Capacity Versus CD4 +CD25 +CD127 lo/- Tregs for Adoptive Cell Therapy. Front Immunol 2022; 13:873560. [PMID: 35693814 PMCID: PMC9178079 DOI: 10.3389/fimmu.2022.873560] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 04/28/2022] [Indexed: 01/21/2023] Open
Abstract
Regulatory T cell (Treg) adoptive cell therapy (ACT) represents an emerging strategy for restoring immune tolerance in autoimmune diseases. Tregs are commonly purified using a CD4+CD25+CD127lo/- gating strategy, which yields a mixed population: 1) cells expressing the transcription factors, FOXP3 and Helios, that canonically define lineage stable thymic Tregs and 2) unstable FOXP3+Helios- Tregs. Our prior work identified the autoimmune disease risk-associated locus and costimulatory molecule, CD226, as being highly expressed not only on effector T cells but also, interferon-γ (IFN-γ) producing peripheral Tregs (pTreg). Thus, we sought to determine whether isolating Tregs with a CD4+CD25+CD226- strategy yields a population with increased purity and suppressive capacity relative to CD4+CD25+CD127lo/- cells. After 14d of culture, expanded CD4+CD25+CD226- cells displayed a decreased proportion of pTregs relative to CD4+CD25+CD127lo/- cells, as measured by FOXP3+Helios- expression and the epigenetic signature at the FOXP3 Treg-specific demethylated region (TSDR). Furthermore, CD226- Tregs exhibited decreased production of the effector cytokines, IFN-γ, TNF, and IL-17A, along with increased expression of the immunoregulatory cytokine, TGF-β1. Lastly, CD226- Tregs demonstrated increased in vitro suppressive capacity as compared to their CD127lo/- counterparts. These data suggest that the exclusion of CD226-expressing cells during Treg sorting yields a population with increased purity, lineage stability, and suppressive capabilities, which may benefit Treg ACT for the treatment of autoimmune diseases.
Collapse
Affiliation(s)
- Matthew E. Brown
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, Diabetes Institute, University of Florida, Gainesville, FL, United States
| | - Leeana D. Peters
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, Diabetes Institute, University of Florida, Gainesville, FL, United States
| | - Seif R. Hanbali
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, Diabetes Institute, University of Florida, Gainesville, FL, United States
| | - Juan M. Arnoletti
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, Diabetes Institute, University of Florida, Gainesville, FL, United States
| | - Lindsey K. Sachs
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, Diabetes Institute, University of Florida, Gainesville, FL, United States
| | - Kayla Q. Nguyen
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, Diabetes Institute, University of Florida, Gainesville, FL, United States
| | - Emma B. Carpenter
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, Diabetes Institute, University of Florida, Gainesville, FL, United States
| | - Howard R. Seay
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, Diabetes Institute, University of Florida, Gainesville, FL, United States
- ROSALIND, Inc., San Diego, CA, United States
| | - Christopher A. Fuhrman
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, Diabetes Institute, University of Florida, Gainesville, FL, United States
- NanoString Technologies, Inc., Seattle, WA, United States
| | - Amanda L. Posgai
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, Diabetes Institute, University of Florida, Gainesville, FL, United States
| | - Melanie R. Shapiro
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, Diabetes Institute, University of Florida, Gainesville, FL, United States
| | - Todd M. Brusko
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, Diabetes Institute, University of Florida, Gainesville, FL, United States
- Department of Pediatrics, College of Medicine, Diabetes Institute, University of Florida, Gainesville, FL, United States
| |
Collapse
|
37
|
Yan H, Li B, Su R, Gao C, Li X, Wang C. Preliminary Study on the Imbalance Between Th17 and Regulatory T Cells in Antiphospholipid Syndrome. Front Immunol 2022; 13:873644. [PMID: 35603166 PMCID: PMC9121099 DOI: 10.3389/fimmu.2022.873644] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 04/13/2022] [Indexed: 11/13/2022] Open
Abstract
ObjectivePatients with antiphospholipid syndrome (APS) have immune cell abnormalities that remain poorly understood. This study compared primary APS (PAPS) and secondary APS (SAPS) patients with healthy controls with respect to peripheral blood lymphocytes, CD4+T cell subsets, and cytokine levels. The correlation between antiphospholipid antibody titres and T helper 17 (Th17) and T regulatory (Treg) cell subsets was also analyzed, together with the correlations between cytokine profiles and the clinical characteristics of APS patients.MethodsThe retrospective study population consisted of 67 APS patients (12 with PAPS, 55 with SAPS) and 40 healthy controls. Absolute numbers of peripheral blood lymphocyte subsets and CD4+ T cell subsets were detected by flow cytometry, and serum cytokine levels by flow cytometry bead array.ResultsPatients with SAPS had lower absolute values of T, B and CD4+T cells than the healthy control group, while only natural killer (NK) cell levels were decreased in patients with PAPS. Absolute numbers of T, B, NK, and CD4+T cells were significantly higher in the PAPS than SAPS group. The trends in CD4+T cell subsets were the same in PAPS and SAPS patients as in healthy controls, with increased Th1, decreased Th2, and decreased Treg levels, and thus an increased Th17/Treg ratio. Th2, Th17, and Treg cell counts were higher in the PAPS than SAPS group. Cytokine analysis showed that only IL-10 levels differed between the two APS groups. However, the levels of all of the studied cytokines were higher in APS patients than healthy controls, and correlated with the clinical characteristics of the patients. In the PAPS group, the titres of two autoantibodies correlated positively with the Th17/Treg ratio and negatively with the levels of D-dimer and Treg subsets.ConclusionsOur study clearly showed that APS patients have immune disturbances, the most prominent of which is an increase in the Th17/Treg ratio, due to a decrease in the number of Treg cells. These abnormalities may be involved in the occurrence and progression of APS. An additional finding was a higher level of peripheral blood lymphocytes in PAPS than SAPS patients, which may be related to the immunosuppressive treatment of SAPS patients.
Collapse
Affiliation(s)
- Huanhuan Yan
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Baochen Li
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Rui Su
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Chong Gao
- Pathology, Joint Program in Transfusion Medicine, Brigham and Women’s Hospital/Children’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Xiaofeng Li
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Caihong Wang
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, China
- *Correspondence: Caihong Wang,
| |
Collapse
|
38
|
Krause N, Mengwasser J, Phithak E, Beato F, Appis M, Milford EL, Pratschke J, Sauer I, Kuehl A, Vogel A, Goodyear M, Hammerich L, Tacke F, Haas JF, Müller T, Utku N. Immune Regulatory 1 Cells: A Novel and Potent Subset of Human T Regulatory Cells. Front Immunol 2022; 12:790775. [PMID: 35222353 PMCID: PMC8867398 DOI: 10.3389/fimmu.2021.790775] [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: 10/07/2021] [Accepted: 12/22/2021] [Indexed: 11/23/2022] Open
Abstract
A subset of T regulatory cells (Tregs), identified by TIRC7 (T cell immune response cDNA 7) expression is designated as Immune Regulatory 1 Cells (IR1 cells). TIRC7 is an immune checkpoint inhibitor, co-localized with the T- cell receptor, HLA-DR and CTLA-4 during T-cell activation, which delivers regulatory signals via binding to its ligand, HLA-DR α2 domain. IR1 cells express FOXP3, and multiple other markers associated with immune suppression. They constitute as much as 10% of Tregs. IR1 cells strongly inhibit proliferation in mixed lymphocyte reactions, where they express high levels of IL-10. Ex vivo expansion of Tregs over 2 weeks in the presence of an agonist TIRC7 antibody disproportionately expands the IR1 Treg subset, while maintaining high expression of suppressive markers including CD39, IL-10, LAP and GARP. Ex vivo expanded IR1 cells are a potent, homogeneous, stable set of suppressor Tregs with the potential to modulate immune dysregulation. The characteristics of IR1 cells suggest a therapeutic advantage over polyclonal Tregs for therapeutic interventions. Early restoration of immune homeostasis using IR1 cells has the potential to fundamentally alter the natural history of conditions characterized by abnormalities in the T regulatory cell compartment.
Collapse
Affiliation(s)
- Nicolas Krause
- Institute for Medical Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Department of Hepatology and Gastroenterology, Universitätsmedizin Berlin, Berlin, Germany
| | - Jörg Mengwasser
- Department of Surgery, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Elpida Phithak
- Institute for Medical Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Department of Hepatology and Gastroenterology, Universitätsmedizin Berlin, Berlin, Germany
| | - Francisca Beato
- Department of Gastroenterology, Moffit Cancer Center, Tampa, FL, United States
| | - Marc Appis
- Department of Biochemistry, Freie Universität, Berlin, Germany
| | - Edgar Louis Milford
- Department of Medicine, Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Johan Pratschke
- Department of Surgery, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Igor Sauer
- Department of Surgery, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Anja Kuehl
- Department of Hepatology and Gastroenterology, Universitätsmedizin Berlin, Berlin, Germany
| | - Arndt Vogel
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Michael Goodyear
- Department of Medicine, Dalhousie University, Halifax, NS, Canada
| | - Linda Hammerich
- Department of Hepatology and Gastroenterology, Charite, Berlin, Germany
| | - Frank Tacke
- Department of Hepatology and Gastroenterology, Charite, Berlin, Germany
| | - Johanna Faith Haas
- Institute for Medical Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Tobias Müller
- Department of Hepatology and Gastroenterology, Universitätsmedizin Berlin, Berlin, Germany
| | - Nalan Utku
- Institute for Medical Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Sachs Incubator for Translational Medicine, Sächsische Inkubator für Klinische Translation (SIKT), University of Leipzig, Leipzig, Germany
| |
Collapse
|
39
|
Braga A, Neves E, Guimarães J, Braga J, Vasconcelos C. The dynamics of Th17 / Treg ratio in SLE patients during pregnancy. J Reprod Immunol 2022; 151:103622. [DOI: 10.1016/j.jri.2022.103622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 03/22/2022] [Accepted: 04/06/2022] [Indexed: 11/17/2022]
|
40
|
Hoffmann AD, Weinberg SE, Swaminathan S, Chaudhuri S, Mubarak HF, Schipma MJ, Mao C, Wang X, El-Shennawy L, Dashzeveg NK, Wei J, Mehl PJ, Shihadah LJ, Wai CM, Ostiguin C, Jia Y, D'Amico P, Wang NR, Luo Y, Demonbreun AR, Ison MG, Liu H, Fang D. Unique molecular signatures sustained in circulating monocytes and regulatory T cells in Convalescent COVID-19 patients. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2022:2022.03.26.485922. [PMID: 35378753 PMCID: PMC8978941 DOI: 10.1101/2022.03.26.485922] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/01/2023]
Abstract
Over two years into the COVID-19 pandemic, the human immune response to SARS-CoV-2 during the active disease phase has been extensively studied. However, the long-term impact after recovery, which is critical to advance our understanding SARS-CoV-2 and COVID-19-associated long-term complications, remains largely unknown. Herein, we characterized multi-omic single-cell profiles of circulating immune cells in the peripheral blood of 100 patients, including covenlesent COVID-19 and sero-negative controls. The reduced frequencies of both short-lived monocytes and long-lived regulatory T (Treg) cells are significantly associated with the patients recovered from severe COVID-19. Consistently, sc-RNA seq analysis reveals seven heterogeneous clusters of monocytes (M0-M6) and ten Treg clusters (T0-T9) featuring distinct molecular signatures and associated with COVID-19 severity. Asymptomatic patients contain the most abundant clusters of monocyte and Treg expressing high CD74 or IFN-responsive genes. In contrast, the patients recovered from a severe disease have shown two dominant inflammatory monocyte clusters with S100 family genes: S100A8 & A9 with high HLA-I whereas S100A4 & A6 with high HLA-II genes, a specific non-classical monocyte cluster with distinct IFITM family genes, and a unique TGF-β high Treg Cluster. The outpatients and seronegative controls share most of the monocyte and Treg clusters patterns with high expression of HLA genes. Surprisingly, while presumably short-ived monocytes appear to have sustained alterations over 4 months, the decreased frequencies of long-lived Tregs (high HLA-DRA and S100A6) in the outpatients restore over the tested convalescent time (>= 4 months). Collectively, our study identifies sustained and dynamically altered monocytes and Treg clusters with distinct molecular signatures after recovery, associated with COVID-19 severity.
Collapse
|
41
|
Gao YL, Liu YC, Zhang X, Shou ST, Chai YF. Insight Into Regulatory T Cells in Sepsis-Associated Encephalopathy. Front Neurol 2022; 13:830784. [PMID: 35370925 PMCID: PMC8965708 DOI: 10.3389/fneur.2022.830784] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 02/18/2022] [Indexed: 01/09/2023] Open
Abstract
Sepsis-associated encephalopathy (SAE) is a diffuse central nervous system (CNS) dysfunction during sepsis, and is associated with increased mortality and poor outcomes in septic patients. Despite the high incidence and clinical relevance, the exact mechanisms driving SAE pathogenesis are not yet fully understood, and no specific therapeutic strategies are available. Regulatory T cells (Tregs) have a role in SAE pathogenesis, thought to be related with alleviation of sepsis-induced hyper-inflammation and immune responses, promotion of T helper (Th) 2 cells functional shift, neuroinflammation resolution, improvement of the blood-brain barrier (BBB) function, among others. Moreover, in a clinical point of view, these cells have the potential value of improving neurological and psychiatric/mental symptoms in SAE patients. This review aims to provide a general overview of SAE from its initial clinical presentation to long-term cognitive impairment and summarizes the main features of its pathogenesis. Additionally, a detailed overview on the main mechanisms by which Tregs may impact SAE pathogenesis is given. Finally, and considering that Tregs may be a novel target for immunomodulatory intervention in SAE, different therapeutic options, aiming to boost peripheral and brain infiltration of Tregs, are discussed.
Collapse
Affiliation(s)
- Yu-lei Gao
- Department of Emergency Medicine, Tianjin Medical University General Hospital, Tianjin, China
- Yu-lei Gao
| | - Yan-cun Liu
- Department of Emergency Medicine, Tianjin Medical University General Hospital, Tianjin, China
| | - Xiang Zhang
- Department of Emergency Medicine, Rizhao People's Hospital of Shandong Province, Rizhao, China
| | - Song-tao Shou
- Department of Emergency Medicine, Tianjin Medical University General Hospital, Tianjin, China
| | - Yan-fen Chai
- Department of Emergency Medicine, Tianjin Medical University General Hospital, Tianjin, China
- *Correspondence: Yan-fen Chai
| |
Collapse
|
42
|
Bandaru SS, Boyilla R, Merchant N, Nagaraju GP, El-Rayes B. Targeting T regulatory cells: their role in colorectal carcinoma progression and current clinical trials. Pharmacol Res 2022; 178:106197. [DOI: 10.1016/j.phrs.2022.106197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 03/24/2022] [Accepted: 03/25/2022] [Indexed: 10/18/2022]
|
43
|
Liu Y, Tu M, Wang L. Pan-Cancer Analysis Predicts FOXS1 as a Key Target in Prognosis and Tumor Immunotherapy. Int J Gen Med 2022; 15:2171-2185. [PMID: 35241932 PMCID: PMC8887970 DOI: 10.2147/ijgm.s354195] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Accepted: 02/18/2022] [Indexed: 12/14/2022] Open
Abstract
Purpose Only a few studies have reported the role of FOXS1, a transcriptional factor, in the tumor development process. In this article, we investigate the function of FOXS1 in distinct neoplastic development and the tumor immune microenvironment (TIME). Patients and Methods The latent roles of FOXS1 in various tumors were prospected based on TCGA, GTEx, CCLE, GEPIA2, cBioPortal, TIMER, ImmuCellAI databases, GSVA datasets, GSEA datasets, and R packages. The expression difference, gene alteration, clinical characteristics, prognostic values, biological mechanism, potential pathways, tumor microenvironment, and immune cell infiltration related to FOXS1 were appraised. Results FOXS1 was strongly expressed in pan-cancer, and this gene was associated with low survival rates. FOXS1 was linked to many pathways that are cancer-promoting and immune-related. The expression of this transcriptional factor in cancers was positively related to immune cell infiltration, especially M2-like macrophages and Treg cells. In addition to that, FOXS1 demonstrated a positive relationship with many immune-suppression genes, such as TGFB1 and ARORA2A. Conclusion Our study identified an oncogenic effect of FOXS1, which may play a vital role as a prognosticative biological marker in pan-cancer. Exorbitant expression of FOXS1 is associated with high TAMs and Treg cells infiltration. These cells have an immunosuppressive function and promote the development of the immunosuppressive tumor microenvironment. The research of FOXS1 provided a potential drug target for tumor immunotherapy.
Collapse
Affiliation(s)
- Yunqiang Liu
- School of Medical College, Guangdong Medical University, Zhanjiang, Guang Dong Province, 524023, People’s Republic of China
| | - Mengjun Tu
- Eye Hospital and School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, Zhejiang Province, 325027, People’s Republic of China
- National Clinical Research Center for Ocular Diseases, Wenzhou, Zhejiang Province, 325027, People’s Republic of China
| | - Lingling Wang
- Department of Clinical Laboratory Center of the Second Affiliated Hospital & Yu Ying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, 325027, People’s Republic of China
- School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang Province, 325035, People’s Republic of China
- Correspondence: Lingling Wang, Department of Clinical Laboratory Center of The Second Affiliated Hospital & Yu Ying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, 325027, People’s Republic of China, Tel +8618867786629, Email
| |
Collapse
|
44
|
Hall BM, Hall RM, Tran GT, Robinson CM, Wilcox PL, Rakesh PK, Wang C, Sharland AF, Verma ND, Hodgkinson SJ. Interleukin-5 (IL-5) Therapy Prevents Allograft Rejection by Promoting CD4 +CD25 + Ts2 Regulatory Cells That Are Antigen-Specific and Express IL-5 Receptor. Front Immunol 2021; 12:714838. [PMID: 34912327 PMCID: PMC8667344 DOI: 10.3389/fimmu.2021.714838] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 11/01/2021] [Indexed: 12/26/2022] Open
Abstract
CD4+CD25+Foxp3+T cell population is heterogenous and contains three major sub-groups. First, thymus derived T regulatory cells (tTreg) that are naïve/resting. Second, activated/memory Treg that are produced by activation of tTreg by antigen and cytokines. Third, effector lineage CD4+CD25+T cells generated from CD4+CD25- T cells' activation by antigen to transiently express CD25 and Foxp3. We have shown that freshly isolated CD4+CD25+T cells are activated by specific alloantigen and IL-4, not IL-2, to Ts2 cells that express the IL-5 receptor alpha. Ts2 cells are more potent than naïve/resting tTreg in suppressing specific alloimmunity. Here, we showed rIL-5 promoted further activation of Ts2 cells to Th2-like Treg, that expressed foxp3, irf4, gata3 and il5. In vivo, we studied the effects of rIL-5 treatment on Lewis heart allograft survival in F344 rats. Host CD4+CD25+T cells were assessed by FACS, in mixed lymphocyte culture and by RT-PCR to examine mRNA of Ts2 or Th2-like Treg markers. rIL-5 treatment given 7 days after transplantation reduced the severity of rejection and all grafts survived ≥60d whereas sham treated rats fully rejected by day 31 (p<0.01). Treatment with anti-CD25 or anti-IL-4 monoclonal antibody abolished the benefits of treatment with rIL-5 and accelerated rejection. After 10d treatment with rIL-5, hosts' CD4+CD25+ cells expressed more Il5ra and responded to specific donor Lewis but not self. Enriched CD4+CD25+ cells from rIL-5 treated rats with allografts surviving >60 days proliferated to specific donor only when rIL-5 was present and did not proliferate to self or third party. These cells had more mRNA for molecules expressed by Th2-like Treg including Irf4, gata3 and Il5. These findings were consistent with IL-5 treatment preventing rejection by activation of Ts2 cells and Th2-like Treg.
Collapse
Affiliation(s)
- Bruce M Hall
- Immune Tolerance Laboratory, South West Clinical School, University of New South Wales (UNSW) Sydney, Liverpool, NSW, Australia.,Ingham Institute of Applied Medical Research, Liverpool Hospital, Liverpool, NSW, Australia
| | - Rachael M Hall
- Immune Tolerance Laboratory, South West Clinical School, University of New South Wales (UNSW) Sydney, Liverpool, NSW, Australia.,Ingham Institute of Applied Medical Research, Liverpool Hospital, Liverpool, NSW, Australia
| | - Giang T Tran
- Immune Tolerance Laboratory, South West Clinical School, University of New South Wales (UNSW) Sydney, Liverpool, NSW, Australia.,Ingham Institute of Applied Medical Research, Liverpool Hospital, Liverpool, NSW, Australia
| | - Catherine M Robinson
- Immune Tolerance Laboratory, South West Clinical School, University of New South Wales (UNSW) Sydney, Liverpool, NSW, Australia.,Ingham Institute of Applied Medical Research, Liverpool Hospital, Liverpool, NSW, Australia
| | - Paul L Wilcox
- Immune Tolerance Laboratory, South West Clinical School, University of New South Wales (UNSW) Sydney, Liverpool, NSW, Australia.,Ingham Institute of Applied Medical Research, Liverpool Hospital, Liverpool, NSW, Australia
| | - Prateek K Rakesh
- Immune Tolerance Laboratory, South West Clinical School, University of New South Wales (UNSW) Sydney, Liverpool, NSW, Australia.,Ingham Institute of Applied Medical Research, Liverpool Hospital, Liverpool, NSW, Australia
| | - Chuanmin Wang
- Transplantation Immunobiology Group, Central Clinical School, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Alexandra F Sharland
- Transplantation Immunobiology Group, Central Clinical School, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Nirupama D Verma
- Immune Tolerance Laboratory, South West Clinical School, University of New South Wales (UNSW) Sydney, Liverpool, NSW, Australia.,Ingham Institute of Applied Medical Research, Liverpool Hospital, Liverpool, NSW, Australia
| | - Suzanne J Hodgkinson
- Immune Tolerance Laboratory, South West Clinical School, University of New South Wales (UNSW) Sydney, Liverpool, NSW, Australia.,Ingham Institute of Applied Medical Research, Liverpool Hospital, Liverpool, NSW, Australia
| |
Collapse
|
45
|
Tang XD, Ji TT, Dong JR, Feng H, Chen FQ, Chen X, Zhao HY, Chen DK, Ma WT. Pathogenesis and Treatment of Cytokine Storm Induced by Infectious Diseases. Int J Mol Sci 2021; 22:13009. [PMID: 34884813 PMCID: PMC8658039 DOI: 10.3390/ijms222313009] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Revised: 11/24/2021] [Accepted: 11/29/2021] [Indexed: 12/15/2022] Open
Abstract
Cytokine storm is a phenomenon characterized by strong elevated circulating cytokines that most often occur after an overreactive immune system is activated by an acute systemic infection. A variety of cells participate in cytokine storm induction and progression, with profiles of cytokines released during cytokine storm varying from disease to disease. This review focuses on pathophysiological mechanisms underlying cytokine storm induction and progression induced by pathogenic invasive infectious diseases. Strategies for targeted treatment of various types of infection-induced cytokine storms are described from both host and pathogen perspectives. In summary, current studies indicate that cytokine storm-targeted therapies can effectively alleviate tissue damage while promoting the clearance of invading pathogens. Based on this premise, "multi-omics" immune system profiling should facilitate the development of more effective therapeutic strategies to alleviate cytokine storms caused by various diseases.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - De-Kun Chen
- College of Veterinary Medicine, Northwest A&F University, Yangling District, Xianyang 712100, China; (X.-D.T.); (T.-T.J.); (J.-R.D.); (H.F.); (F.-Q.C.); (X.C.); (H.-Y.Z.)
| | - Wen-Tao Ma
- College of Veterinary Medicine, Northwest A&F University, Yangling District, Xianyang 712100, China; (X.-D.T.); (T.-T.J.); (J.-R.D.); (H.F.); (F.-Q.C.); (X.C.); (H.-Y.Z.)
| |
Collapse
|
46
|
Pham MN, Khoryati L, Jamison BL, Hayes E, Sullivan JM, Campbell DJ, Gavin MA. In Vivo Expansion of Antigen-Specific Regulatory T Cells through Staggered Fc.IL-2 Mutein Dosing and Antigen-Specific Immunotherapy. Immunohorizons 2021; 5:782-791. [PMID: 34583939 PMCID: PMC11034776 DOI: 10.4049/immunohorizons.2100051] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 08/30/2021] [Indexed: 11/19/2022] Open
Abstract
In mice, Ag administration in the absence of adjuvant typically elicits tolerogenic immune responses through the deletion or inactivation of conventional CD4 T cells and the formation or expansion of regulatory CD4 T cells (Treg). Although these "Ag-specific immunotherapy" (ASI) approaches are currently under clinical development to treat autoinflammatory conditions, efficacy and safety may be variable and unpredictable because of the diverse activation states of immune cells in subjects with autoimmune and allergic diseases. To reliably induce Ag-specific tolerance in patients, novel methods to control T cell responses during ASI are needed, and strategies that permanently increase Treg frequencies among Ag-specific CD4 T cells may provide long-lasting immunosuppression between treatments. In this study, we present an approach to durably increase the frequency of Ag-specific Treg in mice by administering ASI when Treg numbers are transiently increased with individual doses of a half-life-extended Treg-selective IL-2 mutein. Repeated weekly cycles of IL-2 mutein doses (day 0) followed by ASI (day 3) resulted in a 3- to 5-fold enrichment in Treg among Ag-responsive CD4 T cells. Expanded Ag-specific Treg persisted for more than 3 wk following treatment cessation, as well as through an inflammatory T cell response to an Ag-expressing virus. Combining Treg enrichment with ASI has the potential to durably treat autoimmune disease or allergy by increasing the Treg/conventional CD4 T cell ratio among autoantigen- or allergen-specific T cells.
Collapse
Affiliation(s)
- Minh N Pham
- Benaroya Research Institute, Seattle, WA; and
| | | | | | - Erika Hayes
- Benaroya Research Institute, Seattle, WA; and
| | | | | | - Marc A Gavin
- Benaroya Research Institute, Seattle, WA; and
- Omeros Corp., Seattle, WA
| |
Collapse
|
47
|
Li X, Xu H, Huang J, Luo D, Lv S, Lu X, Xiao C. Dysfunctions, Molecular Mechanisms, and Therapeutic Strategies of Regulatory T Cells in Rheumatoid Arthritis. Front Pharmacol 2021; 12:716081. [PMID: 34512345 PMCID: PMC8428974 DOI: 10.3389/fphar.2021.716081] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 07/19/2021] [Indexed: 12/20/2022] Open
Abstract
Regulatory T cells (Tregs) represent a distinct subpopulation of CD4+ T lymphocytes that promote immune tolerance and maintain immune system homeostasis. The dysfunction of Tregs is tightly associated with rheumatoid arthritis (RA). Although the complex pathogenic processes of RA remain unclear, studies on Tregs in RA have achieved substantial progress not only in fundamental research but also in clinical application. This review discusses the current knowledge of the characterizations, functions, and molecular mechanisms of Tregs in the pathogenesis of RA, and potential therapies for these disorders are also involved.
Collapse
Affiliation(s)
- Xiaoya Li
- The Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing, China.,Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, China
| | - Huihui Xu
- Beijing Key Laboratory of Research of Chinese Medicine on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jing Huang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Dan Luo
- Department of Ophthalmology, Traditional Chinese Medicine Hospital of Changping District, Beijing, China
| | - Shuang Lv
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, China
| | - Xiangchen Lu
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, China.,School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Cheng Xiao
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, China.,Department of Emergency, China-Japan Friendship Hospital, Beijing, China
| |
Collapse
|
48
|
Aghabi YO, Yasin A, Kennedy JI, Davies SP, Butler AE, Stamataki Z. Targeting Enclysis in Liver Autoimmunity, Transplantation, Viral Infection and Cancer. Front Immunol 2021; 12:662134. [PMID: 33953725 PMCID: PMC8089374 DOI: 10.3389/fimmu.2021.662134] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 03/22/2021] [Indexed: 12/14/2022] Open
Abstract
Persistent liver inflammation can lead to cirrhosis, which associates with significant morbidity and mortality worldwide. There are no curative treatments beyond transplantation, followed by long-term immunosuppression. The global burden of end stage liver disease has been increasing and there is a shortage of donor organs, therefore new therapies are desperately needed. Harnessing the power of the immune system has shown promise in certain autoimmunity and cancer settings. In the context of the liver, regulatory T cell (Treg) therapies are in development. The hypothesis is that these specialized lymphocytes that dampen inflammation may reduce liver injury in patients with chronic, progressive diseases, and promote transplant tolerance. Various strategies including intrinsic and extracorporeal expansion of Treg cells, aim to increase their abundance to suppress immune responses. We recently discovered that hepatocytes engulf and delete Treg cells by enclysis. Herein, we propose that inhibition of enclysis may potentiate existing regulatory T cell therapeutic approaches in patients with autoimmune liver diseases and in patients receiving a transplant. Moreover, in settings where the abundance of Treg cells could hinder beneficial immunity, such us in chronic viral infection or liver cancer, enhancement of enclysis could result in transient, localized reduction of Treg cell numbers and tip the balance towards antiviral and anti-tumor immunity. We describe enclysis as is a natural process of liver immune regulation that lends itself to therapeutic targeting, particularly in combination with current Treg cell approaches.
Collapse
Affiliation(s)
| | | | | | | | | | - Zania Stamataki
- College of Medical and Dental Sciences, Institute for Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
49
|
Wardell CM, MacDonald KN, Levings MK, Cook L. Cross talk between human regulatory T cells and antigen-presenting cells: Lessons for clinical applications. Eur J Immunol 2020; 51:27-38. [PMID: 33301176 DOI: 10.1002/eji.202048746] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Revised: 10/04/2020] [Accepted: 12/01/2020] [Indexed: 12/14/2022]
Abstract
Regulatory T cells (Tregs) have a critical role in maintaining self-tolerance and immune homeostasis. There is much interest in using Tregs as a cell therapy to re-establish tolerance in conditions such as inflammatory bowel disease and type 1 diabetes, with many ongoing clinical studies testing the safety and efficacy of this approach. Manufacturing of Tregs for therapy typically involves ex vivo expansion to obtain sufficient cell numbers for infusion and comes with the risk of altering the activity of key biological processes. However, this process also offers an opportunity to tailor Treg function to maximize in vivo activity. In this review, we focus on the roles of antigen-presenting cells (APCs) in the generation and function of Tregs in humans. In addition to stimulating the development of Tregs, APCs activate Tregs and provide signals that induce specialized functional and homing marker expression. Cross talk between Tregs and APCs is a critical, often under-appreciated, aspect of Treg biology, with APCs mediating the key properties of infectious tolerance and bystander suppression. Understanding the biology of human Treg-APC interactions will reveal new ways to optimize Treg-based therapeutic approaches.
Collapse
Affiliation(s)
- Christine M Wardell
- Department of Surgery, University of British Columbia, Vancouver, British Columbia, Canada.,BC Children's Hospital Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Katherine N MacDonald
- BC Children's Hospital Research Institute, University of British Columbia, Vancouver, British Columbia, Canada.,School of Biomedical Engineering, University of British Columbia, Vancouver, British Columbia, Canada.,Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada
| | - Megan K Levings
- Department of Surgery, University of British Columbia, Vancouver, British Columbia, Canada.,BC Children's Hospital Research Institute, University of British Columbia, Vancouver, British Columbia, Canada.,School of Biomedical Engineering, University of British Columbia, Vancouver, British Columbia, Canada.,Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Laura Cook
- BC Children's Hospital Research Institute, University of British Columbia, Vancouver, British Columbia, Canada.,Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|