1
|
Zou L, Chen J, Xie L, Zhang L, Wan L, Li W, Xu H. Landscape of T Cells in Tuberculous Pleural Effusion. THE CLINICAL RESPIRATORY JOURNAL 2025; 19:e70066. [PMID: 40170555 PMCID: PMC11962214 DOI: 10.1111/crj.70066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 12/02/2024] [Accepted: 02/11/2025] [Indexed: 04/03/2025]
Abstract
The distribution and the function of T lymphocyte subsets in pleural effusion (PE) and peripheral blood (PB) in tuberculous pleural effusion (TPE) patients remain unclear. In this study, we aimed to explore the expression patterns, regulatory mechanisms, and functions of T lymphocyte subsets in TPE patients, especially the distribution of T lymphocyte subsets at the single cell level. The CD3+ T cells were isolated from PE and PB of four TPE patients for single-cell RNA sequencing (scRNA-seq) to screen T cell subsets. T-SNE projection, Gene Set Variation Analysis (GSVA), and pseudotime analysis were performed to analyze the composition, molecular and functional properties, and developmental trajectories of T cell subsets. Finally, ELISA was carried out to identify the cytokines secreted by PE and PB. We found that CD4+CD8- T lymphocytes (Th1, Th2, and FOXP3+ Treg cells) were preferentially enriched in PE. The proportion of exhausted CD4-CD8+ cells in PE was higher than that in PB, while the proportion of initial and effector CD4-CD8+ cells was quite the reverse. We also found a large number of unexpected double positive (DP) cells in PE and PB, among which the proportion of CD4+CD8+-C10-CCL3 cells was the most different between PE and PB. Meanwhile, CD4+CD8+-C10-CCL3 was the group with the largest number of interactions with other groups. CD4-CD8- cells were mainly found in PE and may be involved in the immunomodulatory effect of PE. Furthermore, the concentrations of cytokines secreted by Th1, Th2, and Treg in PE were higher than those in PB. Our study is helpful to understand the distribution pattern and dynamic changes of T cells in PE and PB of TPE patients and further understand that the functional status and regulation of T cells will be crucial for the successful development of TPE immunotherapy.
Collapse
Affiliation(s)
- Lihui Zou
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical SciencesBeijing Hospital/National Center of Gerontology of National Health CommissionBeijingChina
| | - Jing Chen
- Beijing Economic and Technological Development ZoneAnnoroad Gene Technology Co., LtdBeijingChina
| | - Li Xie
- Department of TuberculosisBeijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research InstituteBeijingChina
| | - Lili Zhang
- Clinical Biobank, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical SciencesBeijingChina
| | - Li Wan
- Clinical Biobank, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical SciencesBeijingChina
| | - Weimin Li
- National Tuberculosis Clinical Lab of ChinaBeijing Tuberculosis and Thoracic Tumor Research Institute, Beijing Chest Hospital, Capital Medical UniversityBeijingChina
| | - Hongtao Xu
- Department of Laboratory MedicineBeijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical SciencesBeijingChina
| |
Collapse
|
2
|
Li Y, Song X, Huang Y, Zhou S, Zhong L. Genetic associations of plasma metabolites with immune cells in hyperthyroidism revealed by Mendelian randomization and GWAS-sc-eQTLs xQTLbiolinks analysis. Sci Rep 2025; 15:1377. [PMID: 39779799 PMCID: PMC11711443 DOI: 10.1038/s41598-025-85664-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 01/06/2025] [Indexed: 01/11/2025] Open
Abstract
Mendelian randomization (MR) was employed to investigate the causal relationships between immune cell phenotypes, hyperthyroidism (HD), and potential metabolic mediators. In this study, we acquired 731 immune cell phenotypes from genome-wide association studies (GWAS) (n = 18,622), HD data from the research by Handan Melike Dönertaş et al. (3,731 cases, 480,867 controls), and aggregated statistics of 1,400 blood metabolites from UK Biobank (n = 115,078). Bidirectional MR analysis was performed to explore the causal relationships between the immune cell phenotypes and HD, and two-sample and multi-variable MR were conducted to identify the potential plasma metabolites mediating in HD. In addition, sensitivity analyses were used to evaluate robustness, heterogeneity, and horizontal pleiotropy of results. Single-cell transcriptome-based exploration of potential key molecule and mechanism by which plasma metabolites regulated the immune cell differentiation in HD pathogenesis. Co-localization analysis was using single-cell eQTL (sc-eQTL) data with key molecule to probe genetically shared effects. Two-sample MRshowed that CD25 on naive-mature B cell, CD8 + NKT cell, and thymol sulfate level were found to have causal relationships with HD (P < 0.008). The causal relationship between thymol sulfate and HD were further validated in an independent cohort using the inverse-variance weighted (IVW). In addition, CD25 on naive-mature B cells and CD8 + NKT cells were both negatively correlated with thymol sulfate (P < 0.05). The results remained significant after MR-Egger and MR-PRESSO correction for horizontal pleiotropy and heterogeneity (P > 0.05). Multi-variable MR results showed that CD25 on naive-mature B cell and CD8 + NKT cell mediated 8.67% and 10.4% of the associations between thymol sulfate and HD, respectively. Moreover, thymol sulfate mediated the evolution of CD8 + NKT cells in the immune microenvironment, identifying PTPRC, PTK2B, KDM5A and TIGIT as the key participating molecules. Co-localization analysis showed that the key molecules had significant genetic sharing effects with CD8 + NKT cells (PPH4 > 0.75, R2 > 0.8, P < 0.05), with PTK2B having the broadest sharing interval. Current MR study provides evidence supporting causal relationships between several specific immune cell phenotypes and HD, as well as potential mediating metabolites. Thymol sulfate may increases the risk of HD pathogenesis by mediating the evolution of PTK2B genetic variants inducing CD8 + NKT cells in the progression of the immune microenvironment.
Collapse
Affiliation(s)
- Yutong Li
- The First School of Clinical Medicine, Guangdong Medical University, Zhanjiang, 524023, Guangdong Province, China
- The Department of General Surgery, Zhongshan City People's Hospital, Zhongshan, 528400, Guangdong Province, China
| | - Xingyu Song
- The First School of Clinical Medicine, Guangdong Medical University, Zhanjiang, 524023, Guangdong Province, China
- The Department of General Surgery, Zhongshan City People's Hospital, Zhongshan, 528400, Guangdong Province, China
| | - Yuyang Huang
- The First School of Clinical Medicine, Guangdong Medical University, Zhanjiang, 524023, Guangdong Province, China
| | - Sifan Zhou
- The First School of Clinical Medicine, Guangdong Medical University, Zhanjiang, 524023, Guangdong Province, China
- The Department of General Surgery, Zhongshan City People's Hospital, Zhongshan, 528400, Guangdong Province, China
| | - Linkun Zhong
- The Department of General Surgery, Zhongshan City People's Hospital, Zhongshan, 528400, Guangdong Province, China.
| |
Collapse
|
3
|
Jin X, Jiang C, Gan X, Zou X, Li H, Zhang L. Exploring causal relationship between the lipids, immune cells, and leiomyosarcoma: A Mendelian randomization and mediation analysis. Medicine (Baltimore) 2024; 103:e40919. [PMID: 39969344 PMCID: PMC11688058 DOI: 10.1097/md.0000000000040919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 11/06/2024] [Indexed: 02/20/2025] Open
Abstract
This study aimed to delineate the causal nexus between lipids and leiomyosarcoma (LMS), with a particular emphasis on delineating the mediating role of immune cells. Employing a 2-sample Mendelian randomization (MR) framework, we scrutinized the potential association of 179 lipid species with LMS across 179 cases and 314,193 controls. The analysis was underpinned by summary-level data derived from genome-wide association studies. The inverse variance weighting method constituted our primary analytical strategy, augmented by supplementary techniques including MR-Egger, simple mode, weighted median, and weighted mode. To ensure the integrity of our MR inferences, we conducted rigorous horizontal multiplicity, heterogeneity, and Bayesian assessments. Furthermore, a nuanced 2-step Mendelian analysis was undertaken to quantify the extent of immune cell-mediated effects of lipids on LMS. Our comprehensive MR evaluation of 179 lipids species unveiled a significant association between genetically inferred triglyceride levels and an elevated risk of LMS (odds ratio = 2.11, 95% confidence interval = 1.38-3.23, P < .001), while inversely showing no effect of LMS on triglyceride levels (odds ratio= 0.99, 95% confidence interval = 0.94-1.04, P = .83). Additionally, the examination of 731 immune cell phenotypes highlighted CD8+ natural killer T cells as contributing a 6% mediation in the causal pathway from triglycerides to LMS.
Collapse
Affiliation(s)
- Xuemei Jin
- College of Medicine, Southwest Jiaotong University, Chengdu, Sichuan, China
- Department of Oncology, The General Hospital of Western Theater Command, Chengdu, China
| | - Chaoyang Jiang
- Department of Oncology, The General Hospital of Western Theater Command, Chengdu, China
| | - Xia Gan
- Department of Oncology, The General Hospital of Western Theater Command, Chengdu, China
| | - Xinyun Zou
- College of Medicine, Southwest Jiaotong University, Chengdu, Sichuan, China
- Department of Oncology, The General Hospital of Western Theater Command, Chengdu, China
| | - Hua Li
- Department of Oncology, The General Hospital of Western Theater Command, Chengdu, China
| | - Ling Zhang
- College of Medicine, Southwest Jiaotong University, Chengdu, Sichuan, China
- Department of Oncology, The General Hospital of Western Theater Command, Chengdu, China
| |
Collapse
|
4
|
Ai N, Zhang Y, Yang J, Zhang Y, Zhao X, Feng H. Genetically predicted blood metabolites mediate the association between circulating immune cells and severe COVID-19: A Mendelian randomization study. Medicine (Baltimore) 2024; 103:e40509. [PMID: 39560514 PMCID: PMC11575977 DOI: 10.1097/md.0000000000040509] [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: 08/21/2024] [Accepted: 10/24/2024] [Indexed: 11/20/2024] Open
Abstract
Investigating the causal relationship between circulating immune cells, blood metabolites, and severe COVID-19 and revealing the role of blood metabolite-mediated circulating immune cells in disease onset and progression. Genetic variation data of 731 circulating immune cells, 1400 blood metabolites, and severe COVID-19 from genome-wide association study open-access database (https://gwas.mrcieu.ac.uk) were used as instrumental variables for bidirectional and two-step Mendelian randomization analysis. The study identified 11 circulating immune cells with unidirectional causality to severe COVID-19. Two-step Mendelian randomization analysis showed 10 blood metabolites were causally associated with severe COVID-19, and blood Myristate and Citrulline to phosphate ratio mediated the association of circulating effector memory double negative % DN and CD8dim natural killer T cell % T cells, respectively, with severe COVID-19 (Myristate mediated effect ratio was 10.20%, P = .011; Citrulline to phosphate ratio mediated effect ratio was -9.21%, P = .017). This study provides genetic evidence assessing the causal relationship between circulating immune cells, blood metabolites, and severe COVID-19, elucidates the role of blood metabolite-mediated circulating immune cells in severe COVID-19 development, and offers new insights into severe COVID-19 etiology and related preventive and targeted therapeutic strategies.
Collapse
Affiliation(s)
- Ning Ai
- Department of Gastroenterology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yan Zhang
- Department of Gastroenterology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Jing Yang
- Department of Gastroenterology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yu Zhang
- Department of Gastroenterology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xuejing Zhao
- Department of Gastroenterology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Huifen Feng
- Department of Gastroenterology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
5
|
Peng Y, Wang W, Liu X, Li S, Zhang J, Ni X, Gui J. Characterization of HPV6/11-reactive T-cell subsets in papillomas of patients with juvenile-onset recurrent respiratory papillomatosis and identification of HPV11 E7-specific candidate TCR clonotypes. J Virol 2024; 98:e0067724. [PMID: 39258910 PMCID: PMC11495051 DOI: 10.1128/jvi.00677-24] [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: 04/15/2024] [Accepted: 07/20/2024] [Indexed: 09/12/2024] Open
Abstract
Juvenile-onset recurrent respiratory papillomatosis (JORRP) is caused by persistent infection of epithelial cells by low-risk human papillomavirus (HPV) types 6 and 11. While multiple infiltrated immune cells have been reported to mediate disease progress, knowledge of HPV-reactive T-cell subsets in papillomas remains elusive. Through single-cell RNA sequencing and RNA microarray, we found that CD8+ tissue-resident memory T (CD8+ TRM) cells with strong interferon-gamma (IFN-γ) production expanded, and were negatively correlated to the disease severity in the frequency of surgery. These IFN-γ+ CD8+ memory T cells were readily activated and expanded in vitro by autologous dendritic cells loaded with HPV11 E7 peptide pool. Moreover, T cell receptor (TCR) clonal expansion was observed in JORRP papilloma tissues, indicating a biased TCR repertoire toward HPV-specific recognition. Finally, we identified and characterized HPV11 E7-specific candidate TCR clonotypes from IFN-γ+ CD8+ memory T cells, suggesting their potential application in TCR-engineered T cells (TCR-T) therapy for HPV11-related diseases. Our findings provided insights into the specific local immune response to HPV6/11 infection and highlighted the importance of IFN-γ+ CD8+ TRM cells in anti-HPV6/11 T-cell immunity.IMPORTANCEThe persistent recurrence of human papillomavirus (HPV) 6/11 infection in papillomas underscores the failure of local immune responses in patients with juvenile-onset recurrent respiratory papillomatosis (JORRP). Our previous study demonstrated that T cells constitute the predominant immune cell population in JORRP papilloma tissues. Understanding the T-cell-mediated immune responses within JORRP papilloma tissues is crucial for disease control. In the present study, we characterized CD8+ tissue-resident memory T (CD8+ TRM) cells as the primary T-cell subset responsible for local anti-HPV6/11 immunity. Moreover, we identified two HPV11 E7-specific candidate T cell receptor (TCR) clonotypes out of IFN-γ+ CD8+ memory T cells. Overall, our findings provided insights into the local immune responses to HPV6/11 infection and offered information for developing more effective immunotherapeutic strategies against JORRP.
Collapse
Affiliation(s)
- Yun Peng
- Laboratory of Tumor Immunology, Beijing Pediatric Research Institute, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China
- Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing Pediatric Research Institute, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China
| | - Wei Wang
- Laboratory of Tumor Immunology, Beijing Pediatric Research Institute, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China
- Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing Pediatric Research Institute, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China
| | - Xiangjun Liu
- Laboratory of Tumor Immunology, Beijing Pediatric Research Institute, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China
- Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing Pediatric Research Institute, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China
| | - Shilan Li
- Department of Otolaryngology, Head and Neck Surgery, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China
| | - Jie Zhang
- Department of Otolaryngology, Head and Neck Surgery, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China
| | - Xin Ni
- Laboratory of Tumor Immunology, Beijing Pediatric Research Institute, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China
- Department of Otolaryngology, Head and Neck Surgery, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China
- Beijing Key Laboratory for Pediatric Diseases of Otolaryngology, Head and Neck Surgery, Beijing Pediatric Research Institute, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China
| | - Jingang Gui
- Laboratory of Tumor Immunology, Beijing Pediatric Research Institute, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China
- Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing Pediatric Research Institute, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China
| |
Collapse
|
6
|
Meng Q, Ma J, Cui J, Gu Y, Shan Y. Subpopulation dynamics of T and B lymphocytes in Sjögren's syndrome: implications for disease activity and treatment. Front Immunol 2024; 15:1468469. [PMID: 39290700 PMCID: PMC11405198 DOI: 10.3389/fimmu.2024.1468469] [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: 07/22/2024] [Accepted: 08/27/2024] [Indexed: 09/19/2024] Open
Abstract
Sjögren's syndrome (SS) is an autoimmune disorder primarily affecting the body's exocrine glands, particularly the salivary and lacrimal glands, which lead to severe symptoms of dry eyes and mouth. The pathogenesis of SS involves the production of autoantibodies by activated immune cells, and secretion of multiple cytokines, which collectively lead to tissue damage and functional impairment. In SS, the Immune interaction among T and B cells is particularly significant. Lymphocytic infiltration in the salivary glands is predominantly composed of CD4+ T cells, whose activation cause the death of glandular epithelial cells and subsequent tissue destruction. The excessive activity of T cells contributes significantly to the disease mechanism, with helper T cells (CD4+) differentiating into various subgroups including Th1/Th2, Th17, as well as Treg, each contributing to the pathological process through distinct cytokine secretion. In patients with SS, B cells are excessively activated, leading to substantial production of autoantibodies. These antibodies can attack self-tissues, especially the lacrimal and salivary glands, causing inflammation and tissue damage. Changes in B cell subpopulations in SS patients, such as increases in plasmablasts and plasma cells, correlate positively with serum autoantibody levels and disease progression. Therapies targeting T cells and B cells are extensively researched with the aim of alleviating symptoms and improving the quality of life for patients. Understanding how these cells promote disease development through various mechanisms, and further identifying novel T and B cell subgroups with functional characterization, will facilitate the development of more effective strategies to treat SS.
Collapse
Affiliation(s)
- Qingliang Meng
- Department of Rheumatism, Henan Province Hospital of Traditional Chinese Medicine, Zhengzhou, Henan, China
| | - Junfu Ma
- Department of Rheumatism, Henan Province Hospital of Traditional Chinese Medicine, Zhengzhou, Henan, China
| | - Jiakang Cui
- Department of Rheumatism, Henan Province Hospital of Traditional Chinese Medicine, Zhengzhou, Henan, China
| | - Yangyi Gu
- Guanghua Clinical Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yu Shan
- Guanghua Clinical Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
7
|
Choi Y, Saron WA, O'Neill A, Senanayake M, Wilder-Smith A, Rathore AP, St John AL. NKT cells promote Th1 immune bias to dengue virus that governs long-term protective antibody dynamics. J Clin Invest 2024; 134:e169251. [PMID: 39088280 PMCID: PMC11405039 DOI: 10.1172/jci169251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 07/23/2024] [Indexed: 08/03/2024] Open
Abstract
NKT cells are innate-like T cells, recruited to the skin during viral infection, yet their contributions to long-term immune memory to viruses are unclear. We identified granzyme K, a product made by cytotoxic cells including NKT cells, as linked to induction of Th1-associated antibodies during primary dengue virus (DENV) infection in humans. We examined the role of NKT cells in vivo using DENV-infected mice lacking CD1d-dependent (CD1ddep) NKT cells. In CD1d-KO mice, Th1-polarized immunity and infection resolution were impaired, which was dependent on intrinsic NKT cell production of IFN-γ, since it was restored by adoptive transfer of WT but not IFN-γ-KO NKT cells. Furthermore, NKT cell deficiency triggered immune bias, resulting in higher levels of Th2-associated IgG1 than Th1-associated IgG2a, which failed to protect against a homologous DENV rechallenge and promoted antibody-dependent enhanced disease during secondary heterologous infections. Similarly, Th2 immunity, typified by a higher IgG4/IgG3 ratio, was associated with worsened human disease severity during secondary infections. Thus, CD1ddep NKT cells establish Th1 polarity during the early innate response to DENV, which promotes infection resolution, memory formation, and long-term protection from secondary homologous and heterologous infections in mice, with consistent associations observed in humans. These observations illustrate how early innate immune responses during primary infections can influence secondary infection outcomes.
Collapse
Affiliation(s)
- Youngjoo Choi
- Programme in Emerging Infectious Diseases, Duke-National University of Singapore Medical School, Singapore
| | - Wilfried Aa Saron
- Programme in Emerging Infectious Diseases, Duke-National University of Singapore Medical School, Singapore
| | - Aled O'Neill
- Programme in Emerging Infectious Diseases, Duke-National University of Singapore Medical School, Singapore
| | - Manouri Senanayake
- Department of Paediatrics, Faculty of Medicine, University of Colombo, Colombo, Sri Lanka
- Lady Ridgeway Children's Hospital, Colombo, Sri Lanka
| | - Annelies Wilder-Smith
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
- Department of Disease Control, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Abhay Ps Rathore
- Programme in Emerging Infectious Diseases, Duke-National University of Singapore Medical School, Singapore
- Department of Pathology, Duke University Medical Center, Durham, North Carolina, USA
| | - Ashley L St John
- Programme in Emerging Infectious Diseases, Duke-National University of Singapore Medical School, Singapore
- Department of Pathology, Duke University Medical Center, Durham, North Carolina, USA
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- SingHealth Duke-NUS Global Health Institute, Singapore
| |
Collapse
|
8
|
Wang Q, Chen S, Guo Z, Xia S, Zhang M. NK-like CD8 T cell: one potential evolutionary continuum between adaptive memory and innate immunity. Clin Exp Immunol 2024; 217:136-150. [PMID: 38651831 PMCID: PMC11239564 DOI: 10.1093/cei/uxae038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 03/06/2024] [Accepted: 04/22/2024] [Indexed: 04/25/2024] Open
Abstract
CD8 T cells are crucial adaptive immune cells with cytotoxicity to fight against pathogens or abnormal self-cells via major histocompatibility complex class I-dependent priming pathways. The composition of the memory CD8 T-cell pool is influenced by various factors. Physiological aging, chronic viral infection, and autoimmune diseases promote the accumulation of CD8 T cells with highly differentiated memory phenotypes. Accumulating studies have shown that some of these memory CD8 T cells also exhibit innate-like cytotoxicity and upregulate the expression of receptors associated with natural killer (NK) cells. Further analysis shows that these NK-like CD8 T cells have transcriptional profiles of both NK and CD8 T cells, suggesting the transformation of CD8 T cells into NK cells. However, the specific induction mechanism underlying NK-like transformation and the implications of this process for CD8 T cells are still unclear. This review aimed to deduce the possible differentiation model of NK-like CD8 T cells, summarize the functions of major NK-cell receptors expressed on these cells, and provide a new perspective for exploring the role of these CD8 T cells in health and disease.
Collapse
Affiliation(s)
- Qiulei Wang
- Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Shaodan Chen
- Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Zhenhong Guo
- National Key Laboratory of Medical Immunology, Institute of Immunology, Second Military Medical University, Shanghai, China
| | - Sheng Xia
- Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Minghui Zhang
- School of Medicine, Tsinghua University, Beijing, China
| |
Collapse
|
9
|
Chen Y, Jiang B, Qu C, Jiang C, Zhang C, Wang Y, Chen F, Sun X, Su L, Luo Y. Genetically predicted metabolites mediate the causal associations between autoimmune thyroiditis and immune cells. Front Endocrinol (Lausanne) 2024; 15:1424957. [PMID: 39045270 PMCID: PMC11263034 DOI: 10.3389/fendo.2024.1424957] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 06/19/2024] [Indexed: 07/25/2024] Open
Abstract
Introduction We aimed to comprehensively investigate the causal relationship between 731 immune cell traits and autoimmune thyroiditis (AIT) and to identify and quantify the role of 1400 metabolic traits as potential mediators in between. Methods Using summary-level data from genome-wide association studies (GWAS) we performed a two-sample bidirectional Mendelian randomization (MR) analysis of genetically predicted AIT and 731 immune cell traits. Furthermore, we used a two-step MR analysis to quantify the proportion of the total effects (that the immune cells exerted on the risk of AIT) mediated by potential metabolites. Results We identified 24 immune cell traits (with odds ratio (OR) ranging from 1.3166 6 to 0.6323) and 10 metabolic traits (with OR ranging from 1.7954 to 0.6158) to be causally associated with AIT, respectively. Five immune cell traits (including CD38 on IgD+ CD24-, CD28 on CD28+ CD45RA+ CD8br, HLA DR+ CD4+ AC, TD CD4+ %CD4+, and CD8 on EM CD8br) were found to be associated with the risk of AIT, which were partially mediated by metabolites (including glycolithocholate sulfate, 5alpha-androstan-3alpha,17beta-diol disulfate, arachidonoylcholine, X-15486, and kynurenine). The proportion of genetically predicted AIT mediated by the identified metabolites could range from 5.58% to 17.7%. Discussion Our study identified causal associations between AIT and immune cells which were partially mediated by metabolites, thus providing guidance for future clinical and basic research.
Collapse
Affiliation(s)
- Yongzhao Chen
- Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Bo Jiang
- Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Cheng Qu
- Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Chaoyu Jiang
- Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Chen Zhang
- Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Yanxue Wang
- Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Fei Chen
- General Surgery Center, Department of Thyroid Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Xitai Sun
- Division of Pancreas and Metabolism Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Lei Su
- Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Yuqian Luo
- Clinical Medicine Research Center, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| |
Collapse
|
10
|
Luo H, Wang K, Li B. Integrating single-cell and spatial transcriptomic analysis to unveil heterogeneity in high-grade serous ovarian cancer. Front Immunol 2024; 15:1420847. [PMID: 38975339 PMCID: PMC11224428 DOI: 10.3389/fimmu.2024.1420847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Accepted: 06/06/2024] [Indexed: 07/09/2024] Open
Abstract
High-grade serous ovarian cancer (HGSOC) presents significant challenges due to its heterogeneity and late-stage diagnoses. Using single-cell and spatial transcriptomics to elucidate the complex landscape of HGSOC to understand its underlying mechanism. Our analysis reveals significant inter- and intra-tumoral diversity, manifested through distinct cellular subpopulations and varied microenvironmental niches. Notably, our findings highlight a widespread immunosuppressive environment, marked by complex networks of cell-cell interactions, particularly evident in areas of elevated tumor cell density within metastatic samples. We identify the exclusive presence of COL14A1+ neoplastic cells in metastatic specimens, alongside a strong correlation between CD8A+ NKT cells and poor prognosis, and elevated CHODL expression in HGSOC metastasis tissues. Furthermore, knockdown experiments targeting CHODL demonstrate its role in reducing migration and invasion abilities in HGSOC cells. A pivotal discovery of our study is the delineation of specific cellular signatures correlated with adverse outcomes, notably a subset of CHODL+ neoplastic cells characterized by a distinct metabolic phenotype with a predilection for lipid metabolism. The therapeutic targeting of this metabolic pathway with existing inhibitors appears promising in curbing tumor proliferation. These findings enhance our understanding of HGSOC heterogeneity and reveal potential therapeutic targets, promising more effective management strategies for this aggressive cancer subtype.
Collapse
Affiliation(s)
| | | | - Bin Li
- Department of Gynecological Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
11
|
Luo T, Tan X, Qing G, Yu J, Liang XJ, Liang P. A natural killer T cell nanoagonist-initiated immune cascade for hepatocellular carcinoma synergistic immunotherapy. NANOSCALE 2024; 16:11126-11137. [PMID: 38787697 DOI: 10.1039/d4nr00847b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/26/2024]
Abstract
Natural killer T (NKT) cell-mediated immunotherapy shows great promise in hepatocellular carcinoma featuring an inherent immunosuppressive microenvironment. However, targeted delivery of NKT cell agonists remains challenging. Here, we developed a hyaluronic acid (HA) modified metal organic framework (zeolitic imidazolate framework-8, ZIF-8) to encapsulate α-galactosylceramide (α-Galcer), a classic NKT cell agonist, and doxorubicin (DOX) for eliminating liver cancer, denoted as α-Galcer/DOX@ZIF-8@HA. In the tumor microenvironment (TME), these pH-responsive nano-frameworks can gradually collapse to release α-Galcer for activating NKT cells and further boosting other immune cells in order to initiate an antitumor immune cascade. Along with DOX, the released α-Galcer enabled efficient NKT cell activation in TME for synergistic immunotherapy and tumor elimination, leading to evident tumor suppression and prolonged animal survival in both subcutaneous and orthotopic liver tumor models. Manipulating NKT cell agonists into functional nano-frameworks in TME may be matched with other advanced managements applied in a wider range of cancer therapies.
Collapse
Affiliation(s)
- Ting Luo
- School of Medicine, Nankai University, Tianjin, 300071, China.
- Department of Interventional Ultrasound, Fifth Medical Center of Chinese People's Liberation Army General Hospital, Beijing, 100853, China.
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, 100190, China.
| | - Xiaoqiong Tan
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, 100190, China.
| | - Guangchao Qing
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, 100190, China.
| | - Jie Yu
- Department of Interventional Ultrasound, Fifth Medical Center of Chinese People's Liberation Army General Hospital, Beijing, 100853, China.
| | - Xing-Jie Liang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, 100190, China.
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ping Liang
- School of Medicine, Nankai University, Tianjin, 300071, China.
- Department of Interventional Ultrasound, Fifth Medical Center of Chinese People's Liberation Army General Hospital, Beijing, 100853, China.
| |
Collapse
|
12
|
Miguelena Chamorro B, Hameed SA, Dechelette M, Claude JB, Piney L, Chapat L, Swaminathan G, Poulet H, Longet S, De Luca K, Mundt E, Paul S. Characterization of Canine Peyer's Patches by Multidimensional Analysis: Insights from Immunofluorescence, Flow Cytometry, and Single-Cell RNA Sequencing. Immunohorizons 2023; 7:788-805. [PMID: 38015460 PMCID: PMC10696420 DOI: 10.4049/immunohorizons.2300091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 11/06/2023] [Indexed: 11/29/2023] Open
Abstract
The oral route is effective and convenient for vaccine administration to stimulate a protective immune response. GALT plays a crucial role in mucosal immune responses, with Peyer's patches (PPs) serving as the primary site of induction. A comprehensive understanding of the structures and functions of these structures is crucial for enhancing vaccination strategies and comprehending disease mechanisms; nonetheless, our current knowledge of these structures in dogs remains incomplete. We performed immunofluorescence and flow cytometry studies on canine PPs to identify cell populations and structures. We also performed single-cell RNA sequencing (scRNA-seq) to investigate the immune cell subpopulations present in PPs at steady state in dogs. We generated and validated an Ab specifically targeting canine M cells, which will be a valuable tool for elucidating Ag trafficking into the GALT of dogs. Our findings will pave the way for future studies of canine mucosal immune responses to oral vaccination and enteropathies. Moreover, they add to the growing body of knowledge in canine immunology, further expanding our understanding of the complex immune system of dogs.
Collapse
Affiliation(s)
- Beatriz Miguelena Chamorro
- Centre International de Recherche en Infectiologie, Team GIMAP (Saint-Etienne), Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR5308, ENS Lyon, UJM, F69007 Lyon, France
- Global Innovation, Boehringer Ingelheim, Saint-Priest, France
| | | | | | | | - Lauriane Piney
- Global Innovation, Boehringer Ingelheim, Saint-Priest, France
| | - Ludivine Chapat
- Global Innovation, Boehringer Ingelheim, Saint-Priest, France
| | | | - Hervé Poulet
- Global Innovation, Boehringer Ingelheim, Saint-Priest, France
| | - Stéphanie Longet
- Centre International de Recherche en Infectiologie, Team GIMAP (Saint-Etienne), Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR5308, ENS Lyon, UJM, F69007 Lyon, France
| | - Karelle De Luca
- Global Innovation, Boehringer Ingelheim, Saint-Priest, France
| | - Egbert Mundt
- Global Innovation, Boehringer Ingelheim, Saint-Priest, France
| | - Stéphane Paul
- Centre International de Recherche en Infectiologie, Team GIMAP (Saint-Etienne), Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR5308, ENS Lyon, UJM, F69007 Lyon, France
- International Center for Infectiology Research, INSERM 1408 Vaccinology, Saint-Etienne, France
| |
Collapse
|
13
|
Almeida JS, Casanova JM, Santos-Rosa M, Tarazona R, Solana R, Rodrigues-Santos P. Natural Killer T-like Cells: Immunobiology and Role in Disease. Int J Mol Sci 2023; 24:ijms24032743. [PMID: 36769064 PMCID: PMC9917533 DOI: 10.3390/ijms24032743] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 01/27/2023] [Accepted: 01/29/2023] [Indexed: 02/04/2023] Open
Abstract
CD56+ T cells are generally recognized as a distinct population of T cells and are categorized as NKT-like cells. Although our understanding of NKT-like cells is far from satisfactory, it has been shown that aging and a number of disease situations have impacted these cells. To construct an overview of what is currently known, we reviewed the literature on human NKT-like cells. NKT-like cells are highly differentiated T cells with "CD1d-independent" antigen recognition and MHC-unrestricted cell killing. The genesis of NKT-like cells is unclear; however, it is proposed that the acquisition of innate characteristics by T cells could represent a remodeling process leading to successful aging. Additionally, it has been shown that NKT-like cells may play a significant role in several pathological conditions, making it necessary to comprehend whether these cells might function as prognostic markers. The quantification and characterization of these cells might serve as a cutting-edge indicator of individual immune health. Additionally, exploring the mechanisms that can control their killing activity in different contexts may therefore result in innovative therapeutic alternatives in a wide range of disease settings.
Collapse
Affiliation(s)
- Jani-Sofia Almeida
- Institute of Immunology, Faculty of Medicine, University of Coimbra (FMUC), 3004-504 Coimbra, Portugal
- Laboratory of Immunology and Oncology, Center for Neuroscience and Cell Biology (CNC), University of Coimbra, 3004-504 Coimbra, Portugal
- Center of Investigation in Environment, Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, 3004-504 Coimbra, Portugal
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
- Center for Innovation in Biomedicine and Biotechnology (CIBB), University of Coimbra, 3004-504 Coimbra, Portugal
- Clinical Academic Centre of Coimbra (CACC), 3000-075 Coimbra, Portugal
| | - José Manuel Casanova
- Center of Investigation in Environment, Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, 3004-504 Coimbra, Portugal
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
- Center for Innovation in Biomedicine and Biotechnology (CIBB), University of Coimbra, 3004-504 Coimbra, Portugal
- Clinical Academic Centre of Coimbra (CACC), 3000-075 Coimbra, Portugal
- University Clinic of Orthopedics, Orthopedics Service, Tumor Unit of the Locomotor Apparatus (UTAL), Coimbra Hospital and Universitary Center (CHUC), 3000-075 Coimbra, Portugal
| | - Manuel Santos-Rosa
- Institute of Immunology, Faculty of Medicine, University of Coimbra (FMUC), 3004-504 Coimbra, Portugal
- Center of Investigation in Environment, Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, 3004-504 Coimbra, Portugal
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
- Center for Innovation in Biomedicine and Biotechnology (CIBB), University of Coimbra, 3004-504 Coimbra, Portugal
- Clinical Academic Centre of Coimbra (CACC), 3000-075 Coimbra, Portugal
| | - Raquel Tarazona
- Immunology Unit, Department of Physiology, University of Extremadura, 10003 Cáceres, Spain
| | - Rafael Solana
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Reina Sofía University Hospital, 14004 Córdoba, Spain
- Immunology Unit, Department of Cell Biology, Physiology and Immunology, University of Córdoba, 14071 Córdoba, Spain
| | - Paulo Rodrigues-Santos
- Institute of Immunology, Faculty of Medicine, University of Coimbra (FMUC), 3004-504 Coimbra, Portugal
- Laboratory of Immunology and Oncology, Center for Neuroscience and Cell Biology (CNC), University of Coimbra, 3004-504 Coimbra, Portugal
- Center of Investigation in Environment, Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, 3004-504 Coimbra, Portugal
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
- Center for Innovation in Biomedicine and Biotechnology (CIBB), University of Coimbra, 3004-504 Coimbra, Portugal
- Clinical Academic Centre of Coimbra (CACC), 3000-075 Coimbra, Portugal
- Correspondence:
| |
Collapse
|
14
|
Hu X, Li F, Zeng J, Zhou Z, Wang Z, Chen J, Cao D, Hong Y, Huang L, Chen Y, Xu J, Dong F, Yu R, Zheng H. Noninvasive Low-Frequency Pulsed Focused Ultrasound Therapy for Rheumatoid Arthritis in Mice. RESEARCH (WASHINGTON, D.C.) 2022; 2022:0013. [PMID: 39290964 PMCID: PMC11407525 DOI: 10.34133/research.0013] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 10/31/2022] [Indexed: 10/16/2023]
Abstract
Rheumatoid arthritis (RA) is a common autoimmune disease characterized by chronic and progressive inflammation of the synovium. Focused ultrasound therapy is an increasingly attractive alternative for treating RA owing to its noninvasiveness; however, it remains unclear which immune subsets respond to ultrasound stimulation. In this study, we showed that spleen-targeted low-frequency pulsed focused ultrasound (LFPFU) effectively improved the severity of arthritis in an arthritis mouse model established in DBA/1J mice. Additionally, we performed in-depth immune profiling of spleen samples from RA mice, RA mice that underwent ultrasound therapy, and healthy controls using mass cytometry along with extensive antibody panels and identified the immune composition of 14 cell populations, including CD4+/CD8+ T cells, B cells, natural killer cells, and dendritic cells. Moreover, multidimensional analysis according to cell-surface markers and phenotypes helped in identifying 4 and 5 cell subpopulations among T and myeloid cells, respectively, with 6 T cell subsets and 3 myeloid cell subsets responsive to ultrasound therapy among the 3 groups. Of these cell subsets, CD8+ T cell subsets showed a unique response to ultrasound stimulation in RA mice. Specifically, CD8+ T cells show a noticeable correlation with the degree of arthritis progression and could serve as an indicator for spleen-focused ultrasound-based therapy. Furthermore, single-cell RNA sequencing of spleen cells revealed the importance of T, B, and myeloid cell populations in the anti-inflammatory pathway. These results elucidated the unique cell subsets and transcriptome of splenic cells responsive to LFPFU and demonstrated the potential of spleen-focused ultrasound stimulation in the treatment of inflammatory diseases.
Collapse
Affiliation(s)
- Xuqiao Hu
- Department of Ultrasound, First Affiliated Hospital of Southern University of Science and Technology, Second Clinical Medical College of Jinan University (Shenzhen People's Hospital), Shenzhen 518020, China
- Integrated Chinese and Western Medicine Postdoctoral Research Station, Jinan University, Guangzhou 510632, China
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, 1068 Xueyuan Avenue, Shenzhen University Town, Shenzhen 518055, China
| | - Fei Li
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, 1068 Xueyuan Avenue, Shenzhen University Town, Shenzhen 518055, China
| | - Jieying Zeng
- Department of Ultrasound, First Affiliated Hospital of Southern University of Science and Technology, Second Clinical Medical College of Jinan University (Shenzhen People's Hospital), Shenzhen 518020, China
| | - Zhenru Zhou
- Department of Ultrasound, First Affiliated Hospital of Southern University of Science and Technology, Second Clinical Medical College of Jinan University (Shenzhen People's Hospital), Shenzhen 518020, China
| | - Zhaoyang Wang
- Department of Ultrasound, First Affiliated Hospital of Southern University of Science and Technology, Second Clinical Medical College of Jinan University (Shenzhen People's Hospital), Shenzhen 518020, China
| | - Jing Chen
- Department of Ultrasound, First Affiliated Hospital of Southern University of Science and Technology, Second Clinical Medical College of Jinan University (Shenzhen People's Hospital), Shenzhen 518020, China
| | - Dongyan Cao
- Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200032, China
| | - Yifan Hong
- Institute of Molecular Physiology, Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen 518132, P.R. China
| | - Laixin Huang
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, 1068 Xueyuan Avenue, Shenzhen University Town, Shenzhen 518055, China
| | - Yongsheng Chen
- Department of Ultrasound, First Affiliated Hospital of Southern University of Science and Technology, Second Clinical Medical College of Jinan University (Shenzhen People's Hospital), Shenzhen 518020, China
| | - Jinfeng Xu
- Department of Ultrasound, First Affiliated Hospital of Southern University of Science and Technology, Second Clinical Medical College of Jinan University (Shenzhen People's Hospital), Shenzhen 518020, China
| | - Fajin Dong
- Department of Ultrasound, First Affiliated Hospital of Southern University of Science and Technology, Second Clinical Medical College of Jinan University (Shenzhen People's Hospital), Shenzhen 518020, China
| | - Rongmin Yu
- Integrated Chinese and Western Medicine Postdoctoral Research Station, Jinan University, Guangzhou 510632, China
- Department of Pharmacology, College of Pharmacy, Jinan University, 601 Huangpu Avenue West, Guangzhou 510632, China
- Biotechnological Institute of Chinese Materia Medica, Jinan University, 601 Huangpu Avenue West, Guangzhou 510632, China
| | - Hairong Zheng
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, 1068 Xueyuan Avenue, Shenzhen University Town, Shenzhen 518055, China
| |
Collapse
|
15
|
Yu W, Yuan X, Ye F, Mao C, Li J, Zhang M, Chen D, Xia S. Role of allogeneic natural killer T cells in the treatment of a patient with gefitinib-sensitive lung adenocarcinoma. Immunotherapy 2022; 14:1291-1296. [PMID: 36169194 DOI: 10.2217/imt-2022-0178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Gefitinib has shown good efficacy in patients with EGFR mutation-positive non-small-cell lung cancer, but acquired resistance is inevitable. Here we report a patient with an advanced lung adenocarcinoma with the EGFR mutation who achieved surgical opportunity and long-term survival following treatment with chemotherapy and bevacizumab, followed by sequential gefitinib combined with allogeneic haploidentical CD8+ CD56+ natural killer T cells. Our case provides a potential effective strategy for delaying acquired gefitinib resistance and extending progression-free survival among patients with non-small-cell lung cancer who harbor common EGFR mutations.
Collapse
Affiliation(s)
- Wanjun Yu
- Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu Province, 212013, China.,Department of Clinical Laboratory, QingPu Branch of Zhongshan Hospital Affiliated to Fudan University, Shanghai, 201700, China
| | - Xiao Yuan
- Department of Oncology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu Province, 212001, China
| | - Fei Ye
- Department of Oncology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu Province, 212001, China
| | - Chaoming Mao
- Department of Nuclear Medicine, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu Province, 212001, China
| | - Jian Li
- Department of Pneumology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu Province, 212001, China
| | - Minghui Zhang
- School of Medicine, Tsinghua University, Beijing, 100084, China
| | - Deyu Chen
- Department of Oncology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu Province, 212001, China
| | - Sheng Xia
- Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu Province, 212013, China
| |
Collapse
|
16
|
Multiparametric Flow Cytometry-Based Immunophenotyping of Mouse Liver Immune Cells. Methods Protoc 2022; 5:mps5050070. [PMID: 36136816 PMCID: PMC9498390 DOI: 10.3390/mps5050070] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 08/31/2022] [Accepted: 09/01/2022] [Indexed: 11/16/2022] Open
Abstract
The liver is a complex organ that governs many types of metabolisms, including energy metabolism and other cellular processes. The liver also plays a crucial role in important functions in immunity, and the activity of liver tissue-associated immunity affects the outcome of many liver pathologies. A thorough characterization of the liver immune microenvironment may contribute to a better understanding of immune signaling, the mechanisms of specific immune responses, and even to improved predictions about therapy outcomes. In this paper, we present an optimized, simple, and rapid protocol to characterize the liver-associated immune cell milieu. We believe that the most suitable technique for obtaining a complex immune cell suspension and for removing contaminating blood cells is to perform mouse liver perfusion, using only phosphate buffer saline. Combining an enzymatic digestion and a mechanical dissociation of liver tissue, followed by cell purification, improves downstream applications. This combination is an essential prerequisite for immune cell determination and characterization. We then demonstrate a flow cytometry-based multiparametric immunophenotyping along with a gating strategy to detect and quantify liver endothelial cells, T cells (helper and cytotoxic), B cells, NK cells, NKT cells, neutrophils, monocytes (subsets included), dendritic cells (subsets included), macrophages and Kupffer cells.
Collapse
|
17
|
Lee JM, Chen MH, Chou KY, Chao Y, Chen MH, Tsai CY. Novel immunoprofiling method for diagnosing SLE and evaluating therapeutic response. Lupus Sci Med 2022; 9:9/1/e000693. [PMID: 35738802 PMCID: PMC9226994 DOI: 10.1136/lupus-2022-000693] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 06/11/2022] [Indexed: 11/23/2022]
Abstract
Objective Diagnosis of SLE is based on clinical manifestations but is heterogeneous in early onset. Hence, we aimed to evaluate the feature of the immunoprofiling in patients with SLE and apply it to develop an immune signature algorithm for supporting SLE diagnosis. Methods We enrolled 13 newly diagnosed patients with SLE and 9 healthy controls (HCs) followed by analysing their immunoprofilings within their peripheral blood mononuclear cells (PBMCs) through flow cytometry. The immunoprofiling from the patients with SLE and HCs were ranked and formed an immune signature score. Besides, we enrolled four patients with SLE and monitored the changes in their immunoprofilings after immunosuppressant treatment. Results Among 93 immune cell subsets, 29 differed significantly between patients with SLE and HCs, and lower dendritic and natural killer cell percentages and a higher CD8+ T-cell percentage were identified in patients with SLE. In an investigation of immune-tolerant-related cell subsets, higher concentrations of CD8+ regulatory natural killer T cells, programmed cell death 1 (PD-1)+ T cells, and lower concentrations of programmed cell death ligand 1 (PD-L1)+ PBMCs were observed in the SLE group. The immune signature score from patients with SLE was significantly different from that from the HCs. After treatment, the disease activity of the four patients were tended to stable and percentages of PD-L1+ monocytes, PD-1+ CD4 T and CD8 T cells in patients with SLE exhibited positively and negatively correlation with the SLEDAI-2K (Systemic Lupus Erythematosus Disease Activity Index 2000) score, which might associate with the remission of SLE. Conclusions The comparison of immunprofiling between patients with SLE and HCs exhibited a distinct pattern. This difference and its application to immune signature algorithm shed light on the studies of SLE pathogenesis and immune-based diagnostic tool development in the future.
Collapse
Affiliation(s)
- Jan-Mou Lee
- Department of Advanced Research, FullHope Biomedical Co Ltd, New Taipei City, Taiwan
| | - Ming-Huang Chen
- Faculty of Medicine, National Yang Ming Chiao Tung University, Taipei City, Taiwan
| | - Kai-Yuan Chou
- Department of Advanced Research, FullHope Biomedical Co Ltd, New Taipei City, Taiwan
| | - Yee Chao
- Department of Oncology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Ming-Han Chen
- Faculty of Medicine, National Yang Ming Chiao Tung University, Taipei City, Taiwan .,Division of Allergy-Immunology-Rheumatology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Chang-Youh Tsai
- Division of Allergy-Immunology-Rheumatology, Taipei Veterans General Hospital, Taipei, Taiwan.,Faculty of Medicine, National Yang Ming Chiao Tung University, Hsinchu, Taiwan
| |
Collapse
|
18
|
Scorza BM, Mahachi KG, Cox AD, Toepp AJ, Pessoa-Pereira D, Tyrrell P, Buch J, Foltz JA, Lee D, Petersen CA. Role of NK-Like CD8 + T Cells during Asymptomatic Borrelia burgdorferi Infection. Infect Immun 2022; 90:e0055521. [PMID: 35416707 PMCID: PMC9119074 DOI: 10.1128/iai.00555-21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 03/25/2022] [Indexed: 11/20/2022] Open
Abstract
Lyme disease (LD) due to Borrelia burgdorferi is the most prevalent vector-borne disease in the United States. There is a poor understanding of how immunity contributes to bacterial control, pathology, or both during LD. Dogs in an area of endemicity were screened for B. burgdorferi and Anaplasma exposure and stratified according to seropositivity, presence of LD symptoms, and doxycycline treatment. Significantly elevated serum interleukin-21 (IL-21) and increased circulating CD3+ CD94+ lymphocytes with an NK-like CD8+ T cell phenotype were predominant in asymptomatic dogs exposed to B. burgdorferi. Both CD94+ T cells and CD3- CD94+ lymphocytes, corresponding to NK cells, from symptomatic dogs expressed gamma interferon (IFN-γ) at a 3-fold-higher frequency upon stimulation with B. burgdorferi than the same subset among endemic controls. Surface expression of activating receptor NKp46 was reduced on CD94+ T cells from LD, compared to cells after doxycycline treatment. A higher frequency of NKp46-expressing CD94+ T cells correlated with significantly increased peripheral blood mononuclear cell (PBMC) cytotoxic activity via calcein release assay. PBMCs from dogs with symptomatic LD showed significantly reduced killing ability compared with endemic control PBMCs. An elevated NK-like CD8+ T cell response was associated with protection against development of clinical LD, while excess IFN-γ was associated with clinical disease.
Collapse
Affiliation(s)
- Breanna M. Scorza
- Department of Epidemiology, University of Iowa, Iowa City, Iowa, USA
| | - Kurayi G. Mahachi
- Department of Epidemiology, University of Iowa, Iowa City, Iowa, USA
| | - Arin D. Cox
- Department of Epidemiology, University of Iowa, Iowa City, Iowa, USA
| | - Angela J. Toepp
- Department of Epidemiology, University of Iowa, Iowa City, Iowa, USA
| | | | | | - Jesse Buch
- IDEXX Laboratories, Inc., Westbrook, Maine, USA
| | - Jennifer A. Foltz
- Division of Hematology and Oncology, Nationwide Children’s Hospital, Columbus, Ohio, USA
| | - Dean Lee
- Division of Hematology and Oncology, Nationwide Children’s Hospital, Columbus, Ohio, USA
| | | |
Collapse
|
19
|
Watkinson F, Nayar SK, Rani A, Sakellariou CA, Elhage O, Papaevangelou E, Dasgupta P, Galustian C. IL-15 Upregulates Telomerase Expression and Potently Increases Proliferative Capacity of NK, NKT-Like, and CD8 T Cells. Front Immunol 2021; 11:594620. [PMID: 33537030 PMCID: PMC7848219 DOI: 10.3389/fimmu.2020.594620] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 12/02/2020] [Indexed: 11/13/2022] Open
Abstract
Interleukin-15 (IL-15) is a cytokine that has been shown to expand CD8 T cell and natural killer (NK) cell populations, and therefore has potential for potentiating adoptive immune cell therapy for cancer. Previously, IL-15 has been shown to induce proliferation of CD8 memory T cells through activation of telomerase. Here, we investigated whether telomerase is also activated during the IL-15 mediated proliferation of NK and NKT-like (CD56+CD3+) cells. We also examined the extent that each of the three signaling pathways known to be stimulated by IL-2/IL-15 (JAK-STAT, PI3K-AKT Ras-RAF/MAPK) were activated and involved in the telomerase expression in the three cell types NK, NKT, or CD8 T cells. To assess cell proliferation and doubling, peripheral blood mononuclear cells (PBMCs) or isolated NK, NKT-like or CD8 T cells were incubated with varying concentrations of IL-15 or IL-2 for 7 days. CD8 T, NK, and NKT cell expansion was determined by fluorophore-conjugated antibody staining and flow cytometry. Cell doubling was investigated using carboxyfluorescein-succinimidyl-ester (CFSE). Telomerase expression was investigated by staining cells with anti-telomerase reverse transcriptase (anti-TERT). Telomerase activity in CD56+ and CD8 T cells was also measured via Telomerase Repeat Amplification Protocol (TRAP). Analysis of cellular expansion, proliferation and TERT expression concluded that IL-15 increased cellular growth of NK, NKT, and CD8 T cells more effectively than IL-2 using low or high doses. IL-15, increased TERT expression in NK and NKT cells by up to 2.5 fold, the same increase seen in CD8 T cells. IL-2 had effects on TERT expression only at high doses (100–1000 ng/ml). Proteome profiling identified that IL-15 activated selected signaling proteins in the three pathways (JAK-STAT, PI3K-AKT, Ras-MAPK) known to mediate IL-2/IL-15 signaling, more strongly than IL-2. Evaluation by signaling pathway inhibitors revealed that JAK/STAT and PI3K/AKT pathways are important in IL-15’s ability to upregulate TERT expression in NK and NKT cells, whereas all three pathways were involved in CD8 T cell TERT expression. In conclusion, this study shows that IL-15 potently stimulates TERT upregulation in NK and NKT cells in addition to CD8 T cells and is therefore a valuable tool for adoptive cell therapies.
Collapse
Affiliation(s)
- Fiona Watkinson
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, Faculty of Life Sciences & Medicine, King's College London, Guy's Hospital, London, United Kingdom
| | - Sandeep Krishan Nayar
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, Faculty of Life Sciences & Medicine, King's College London, Guy's Hospital, London, United Kingdom
| | - Aradhana Rani
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, Faculty of Life Sciences & Medicine, King's College London, Guy's Hospital, London, United Kingdom
| | - Christina A Sakellariou
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, Faculty of Life Sciences & Medicine, King's College London, Guy's Hospital, London, United Kingdom
| | - Oussama Elhage
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, Faculty of Life Sciences & Medicine, King's College London, Guy's Hospital, London, United Kingdom.,Urology Centre, Guy's Hospital, London, United Kingdom
| | - Efthymia Papaevangelou
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, Faculty of Life Sciences & Medicine, King's College London, Guy's Hospital, London, United Kingdom
| | - Prokar Dasgupta
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, Faculty of Life Sciences & Medicine, King's College London, Guy's Hospital, London, United Kingdom.,Urology Centre, Guy's Hospital, London, United Kingdom
| | - Christine Galustian
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, Faculty of Life Sciences & Medicine, King's College London, Guy's Hospital, London, United Kingdom
| |
Collapse
|
20
|
Shen H, Gu C, Liang T, Liu H, Guo F, Liu X. Unveiling the heterogeneity of NKT cells in the liver through single cell RNA sequencing. Sci Rep 2020; 10:19453. [PMID: 33173202 PMCID: PMC7655820 DOI: 10.1038/s41598-020-76659-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Accepted: 10/29/2020] [Indexed: 11/10/2022] Open
Abstract
CD1d-dependent type I NKT cells, which are activated by lipid antigen, are known to play important roles in innate and adaptive immunity, as are a portion of type II NKT cells. However, the heterogeneity of NKT cells, especially NKT-like cells, remains largely unknown. Here, we report the profiling of NKT (NK1.1+CD3e+) cells in livers from wild type (WT), Jα18-deficient and CD1d-deficient mice by single-cell RNA sequencing. Unbiased transcriptional clustering revealed distinct cell subsets. The transcriptomic profiles identified the well-known CD1d-dependent NKT cells and defined two CD1d-independent NKT cell subsets. In addition, validation of marker genes revealed the differential organ distribution and landscape of NKT cell subsets during liver tumor progression. More importantly, we found that CD1d-independent Sca-1−CD62L+ NKT cells showed a strong ability to secrete IFN-γ after costimulation with IL-2, IL-12 and IL-18 in vitro. Collectively, our findings provide a comprehensive characterization of NKT cell heterogeneity and unveil a previously undefined functional NKT cell subset.
Collapse
Affiliation(s)
- Hao Shen
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Chan Gu
- Center for Translational Medicine, Ministry of Education Key Laboratory of Birth Defects and Related Diseases of Women and Children, Department of Obstetrics and Gynecology, West China Second Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Tao Liang
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Haifeng Liu
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Fan Guo
- Center for Translational Medicine, Ministry of Education Key Laboratory of Birth Defects and Related Diseases of Women and Children, Department of Obstetrics and Gynecology, West China Second Hospital, Sichuan University, Chengdu, 610041, Sichuan, China. .,Ministry of Education Key Laboratory of Bio-Resource and Eco-Environment, College of Life Sciences, Sichuan University, Chengdu, 610041, Sichuan, China.
| | - Xiaolong Liu
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Beijing, China. .,School of Life Sciences, Hangzhou Institute for Advanced Study, UCAS, Hangzhou, 310024, China. .,School of Life Science and Technology, ShanghaiTech University, Shanghai, 200031, China.
| |
Collapse
|
21
|
Tomaszewski M, Grywalska E, Tomaszewski A, Błaszczak P, Kurzyna M, Roliński J, Kopeć G. Overexpression of PD-1 on Peripheral Blood Lymphocytes in Patients with Idiopathic Pulmonary Arterial Hypertension and Its Association with High Viral Loads of Epstein-Barr Virus and Poor Clinical Parameters. J Clin Med 2020; 9:jcm9061966. [PMID: 32599687 PMCID: PMC7355537 DOI: 10.3390/jcm9061966] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Revised: 06/19/2020] [Accepted: 06/19/2020] [Indexed: 12/12/2022] Open
Abstract
Idiopathic pulmonary arterial hypertension (IPAH) is a rare but severe disease with the elevated blood pressure in the pulmonary arteries without a known trigger of vascular remodelling. It leads to the right heart failure with reduced survival. Changes in the immunological landscape of the lungs and the periphery are common in IPAH patients, suggesting an immune system dysfunction. A cohort of 25 IPAH patients was enrolled in our study to investigate a link between the patient’s clinical status, immune parameters of the blood, and the Epstein–Barr virus (EBV) infection. We found significant alterations of the patients’ peripheral blood parameters. Therein, T lymphocytes and NK cell counts were decreased in the IPAH patients’ blood, while the proportion of regulatory T cells was increased. Additionally, levels of proinflammatory cytokines interleukin-6 (IL-6), IL-2, and interferon-gamma (IFN-γ) were elevated. We identified a weak correlation between EBV loads and IPAH patients’ clinical state (r = 0.54) and between EBV loads and overexpression of PD-1 on helper T cells (r = 0.56). We speculate that a significant dysregulation of the immune system homeostasis observed in IPAH patients may contribute to increased susceptibility of those patients to EBV infection, yet further longitudinal studies are required to characterize this relation in detail.
Collapse
Affiliation(s)
- Michał Tomaszewski
- Department of Cardiology, Medical University of Lublin, 20-954 Lublin, Poland; (M.T.); (A.T.)
| | - Ewelina Grywalska
- Department of Clinical Immunology and Immunotherapy, Medical University of Lublin, 20-093 Lublin, Poland;
- Correspondence: ; Tel.: +48-81448-6420
| | - Andrzej Tomaszewski
- Department of Cardiology, Medical University of Lublin, 20-954 Lublin, Poland; (M.T.); (A.T.)
| | - Piotr Błaszczak
- Department of Cardiology, Cardinal Wyszynski Hospital, 20-718 Lublin, Poland;
| | - Marcin Kurzyna
- Department of Pulmonary Circulation, Thromboembolic Diseases and Cardiology, Centre of Postgraduate Medical Education, Fryderyk Chopin Hospital in European Health Centre Otwock, 05-400 Otwock, Poland;
| | - Jacek Roliński
- Department of Clinical Immunology and Immunotherapy, Medical University of Lublin, 20-093 Lublin, Poland;
| | - Grzegorz Kopeć
- Department of Cardiac and Vascular Diseases, Faculty of Medicine, Jagiellonian University Medical College, Centre for Rare Cardiovascular Diseases, John Paul II Hospital, 31-202 Krakow, Poland;
| |
Collapse
|
22
|
Magden ER, Nehete BP, Chitta S, Williams LE, Simmons JH, Abee CR, Nehete PN. Comparative Analysis of Cellular Immune Responses in Conventional and SPF Olive Baboons ( Papio anubis). Comp Med 2020; 70:160-169. [PMID: 32014083 DOI: 10.30802/aalas-cm-19-000035] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Olive baboons (P. anubis) have provided a useful model of human diseases and conditions, including cardiac, respiratory, and infectious diseases; diabetes; and involving genetics, immunology, aging, and xenotransplantation. The development of a immunologically defined SPF baboons has advanced research further, especially for studies involving the immune system and immunosuppression. In this study, we compare normal immunologic changes of PBMC subsets, and their function in age-matched conventional and SPF baboons. Our results revealed that both groups have comparable numbers of different lymphocyte subsets, but phenotypic differences in central and effector memory T-cell subsets are more pronounced in CD4+ T cells. Despite equal proportions of CD3+ T cells among the conventional and SPF baboons, PBMC from the conventional group showed greater proliferative responses to phytohemagglutinin and pokeweed mitogen and higher numbers of IFNγ-producing cells after stimulation with concanavalin A or pokeweed mitogen, whereas plasma levels of the inflammatory cytokine TNFα were significantly higher in SPF baboons. Exposure of PBMC from conventional baboons to various Toll-like (TLR) ligands, including TLR3, TLR4, and TLR8, yielded increased numbers of IFNγ producing cells, whereas PBMC from SPF baboons stimulated with TLR5 or TLR6 ligand had more IFNγ-producing cells. These findings suggest that although lymphocyte subsets share many phenotypic and functional similarities in conventional and SPF baboons, specific differences in the immune function of lymphocytes could differentially influence the quality and quantity of their innate and adaptive immune responses. These differences should be considered in interpreting experimental outcomes, specifically in studies measuring immunologic endpoints.
Collapse
Affiliation(s)
- Elizabeth R Magden
- The University of Texas MD Anderson Cancer Center Bastrop, Department of Comparative Medicine, Houston, Texas
| | - Bharti P Nehete
- The University of Texas MD Anderson Cancer Center Bastrop, Department of Comparative Medicine, Houston, Texas;,
| | - Sriram Chitta
- The University of Texas MD Anderson Cancer Center Bastrop, Department of Comparative Medicine, Houston, Texas
| | - Lawrence E Williams
- The University of Texas MD Anderson Cancer Center Bastrop, Department of Comparative Medicine, Houston, Texas
| | - Joe H Simmons
- The University of Texas MD Anderson Cancer Center Bastrop, Department of Comparative Medicine, Houston, Texas
| | - Christian R Abee
- The University of Texas MD Anderson Cancer Center Bastrop, Department of Comparative Medicine, Houston, Texas
| | - Pramod N Nehete
- The University of Texas MD Anderson Cancer Center Bastrop, Department of Comparative Medicine, Houston, Texas; The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, Texas
| |
Collapse
|
23
|
Novickij V, Čėsna R, Perminaitė E, Zinkevičienė A, Characiejus D, Novickij J, Šatkauskas S, Ruzgys P, Girkontaitė I. Antitumor Response and Immunomodulatory Effects of Sub-Microsecond Irreversible Electroporation and Its Combination with Calcium Electroporation. Cancers (Basel) 2019; 11:cancers11111763. [PMID: 31717542 PMCID: PMC6896087 DOI: 10.3390/cancers11111763] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 11/04/2019] [Accepted: 11/07/2019] [Indexed: 01/04/2023] Open
Abstract
In this work, we have investigated the feasibility of sub-microsecond range irreversible electroporation (IRE) with and without calcium electroporation in vivo. As a model, BALB/C mice were used and bioluminescent SP2/0 myeloma tumor models were developed. Tumors were treated with two separate pulsed electric field (PEF) pulsing protocols PEF1: 12 kV/cm × 200 ns × 500 (0.006 J/pulse) and PEF2: 12 kV/cm × 500 ns × 500 (0.015 J/pulse), which were delivered with and without Ca2+ (168 mM) using parallel plate electrodes at a repetition frequency of 100 Hz. Both PEF1 and PEF2 treatments reduced tumor growth and prolonged the life span of the mice, however, the PEF2 protocol was more efficient. The delay in tumor renewal was the biggest when a combination of IRE with calcium electroporation was used, however, we did not obtain significant differences in the final mouse survival compared to PEF2 alone. Anti-tumor immune responses were also investigated after treatment with PEF2 and PEF2+Ca. In both cases the treated mice had enlarged spleens and increased spleen T cell numbers, lower percentages of suppressor cell subsets (conventional CD4+CD25+ Treg, CD4+CD25−DX5+ Tr1, CD8+DX5+, CD4+CD28−, CD8+CD28−), changed proportions of Tcm and Tef/Tem T cells in the spleen and increased amount of tumor cell specific antibodies in the sera. The treatment based on IRE was effective against primary tumors, destroyed the tumor microenvironment and induced an anti-tumor immune response, however, it was not sufficient for complete control of tumor metastasis.
Collapse
Affiliation(s)
- Vitalij Novickij
- Faculty of Electronics, Vilnius Gediminas Technical University, 03227 Vilnius, Lithuania
- Correspondence: (V.N.); (I.G.)
| | - Robertas Čėsna
- Department of Immunology, State Research Institute Centre for Innovative Medicine, 08410 Vilnius, Lithuania
| | - Emilija Perminaitė
- Department of Immunology, State Research Institute Centre for Innovative Medicine, 08410 Vilnius, Lithuania
| | - Auksė Zinkevičienė
- Department of Immunology, State Research Institute Centre for Innovative Medicine, 08410 Vilnius, Lithuania
| | - Dainius Characiejus
- Department of Immunology, State Research Institute Centre for Innovative Medicine, 08410 Vilnius, Lithuania
| | - Jurij Novickij
- Faculty of Electronics, Vilnius Gediminas Technical University, 03227 Vilnius, Lithuania
| | - Saulius Šatkauskas
- Biophysical Research Group, Vytautas Magnus University, 44404 Kaunas, Lithuania
| | - Paulius Ruzgys
- Biophysical Research Group, Vytautas Magnus University, 44404 Kaunas, Lithuania
| | - Irutė Girkontaitė
- Department of Immunology, State Research Institute Centre for Innovative Medicine, 08410 Vilnius, Lithuania
- Correspondence: (V.N.); (I.G.)
| |
Collapse
|
24
|
Li Z, Wu Y, Wang C, Zhang M. Mouse CD8 +NKT-like cells exert dual cytotoxicity against mouse tumor cells and myeloid-derived suppressor cells. Cancer Immunol Immunother 2019; 68:1303-1315. [PMID: 31278476 PMCID: PMC6682577 DOI: 10.1007/s00262-019-02363-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Accepted: 06/26/2019] [Indexed: 02/06/2023]
Abstract
Our previous work has demonstrated the high efficiency of CD8+ natural killer T (NKT)-like cells in killing antigen-bearing dendritic cells. To evaluate their role in the tumor microenvironment, we performed in vitro and in vivo antitumor experiments to investigate whether CD8+NKT-like cells could kill Yac-1 and B16 cells like NK cells and kill EL4-OVA8 cells in an antigen-specific manner like cytotoxic T lymphocytes (CTLs). Unlike NK1.1−CTLs, CD8+NKT-like cells also exhibit the capability to kill myeloid-derived suppressor cells (MDSCs) in an antigen-specific manner, indicative of their potential role in clearing tumor antigen-bearing MDSCs to improve the antitumor microenvironment. In vitro blocking experiments showed that granzyme B inhibitor efficiently suppressed the cytotoxicity of CD8+NKT-like cells against tumor cells and MDSCs, while Fas ligand (FasL) or tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) inhibition failed to produce similar effects. Transcriptomic and phenotypic analyses of CD8+NKT-like cells, NK cells, and NK1.1−CTLs indicated that CD8+NKT-like cells expressed both T-cell activation markers and NK cell markers, thus bearing features of both the activated T cells and NK cells. Taken together, CD8+NKT-like cells could exert NK- and CTL-like antitumor effects through the elimination of both tumor cells and MDSCs in a granzyme B-dependent manner.
Collapse
Affiliation(s)
- Zhengyuan Li
- School of Medicine, Tsinghua University, Beijing, 100084, China
| | - Yiqing Wu
- School of Medicine, Tsinghua University, Beijing, 100084, China
| | - Chao Wang
- School of Medicine, Tsinghua University, Room B343, Haidian District, Beijing, 100084, China.
| | - Minghui Zhang
- School of Medicine, Tsinghua University, Room B343, Haidian District, Beijing, 100084, China.
| |
Collapse
|
25
|
Zhang W, Yi Z, Wei C, Keung KL, Sun Z, Xi C, Woytovich C, Farouk S, Gallon L, Menon MC, Magee C, Najafian N, Samaniego MD, Djamali A, Alexander SI, Rosales IA, Smith RN, O'Connell PJ, Colvin R, Cravedi P, Murphy B. Pretransplant transcriptomic signature in peripheral blood predicts early acute rejection. JCI Insight 2019; 4:127543. [PMID: 31167967 DOI: 10.1172/jci.insight.127543] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2019] [Accepted: 04/23/2019] [Indexed: 12/19/2022] Open
Abstract
Commonly available clinical parameters fail to predict early acute cellular rejection (EAR, occurring within 6 months after transplant), a major risk factor for graft loss after kidney transplantation. We performed whole-blood RNA sequencing at the time of transplant in 235 kidney transplant recipients enrolled in a prospective cohort study (Genomics of Chronic Allograft Rejection [GoCAR]) and evaluated the relationship of pretransplant transcriptomic profiles with EAR. EAR was associated with downregulation of NK and CD8+ T cell gene signatures in pretransplant blood. We identified a 23-gene set that predicted EAR in the discovery (n = 81, and AUC = 0.80) and validation (n = 74, and AUC = 0.74) sets. Exclusion of recipients with 5 or 6 HLA donor mismatches increased the AUC to 0.89. The risk score derived from the gene set was also significantly associated with acute cellular rejection after 6 months, antibody-mediated rejection and/or de novo donor-specific antibodies, and graft loss in a cohort of 154 patients, combining the validation set and additional GoCAR patients with surveillance biopsies between 6 and 24 months (n = 80) posttransplant. This 23-gene set is a potentially important new tool for determination of the recipient's immunological risk before kidney transplantation, and facilitation of an individualized approach to immunosuppressive therapy.
Collapse
Affiliation(s)
- Weijia Zhang
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Zhengzi Yi
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Chengguo Wei
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Karen L Keung
- Department of Medicine, Westmead Clinical School, The University of Sydney, Sydney, Australia
| | - Zeguo Sun
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Caixia Xi
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Christopher Woytovich
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Samira Farouk
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Lorenzo Gallon
- Department of Medicine-Nephrology and Surgery-Organ Transplantation, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Madhav C Menon
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Ciara Magee
- Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Nader Najafian
- Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | | | - Arjang Djamali
- Division of Nephrology, Department of Medicine, University of Wisconsin, Madison, Wisconsin, USA
| | - Stephen I Alexander
- Department of Medicine, Westmead Clinical School, The University of Sydney, Sydney, Australia
| | - Ivy A Rosales
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Rex Neal Smith
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Philip J O'Connell
- Department of Medicine, Westmead Clinical School, The University of Sydney, Sydney, Australia
| | - Robert Colvin
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Paolo Cravedi
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Barbara Murphy
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
26
|
Wikenheiser DJ, Brown SL, Lee J, Stumhofer JS. NK1.1 Expression Defines a Population of CD4 + Effector T Cells Displaying Th1 and Tfh Cell Properties That Support Early Antibody Production During Plasmodium yoelii Infection. Front Immunol 2018; 9:2277. [PMID: 30374346 PMCID: PMC6196288 DOI: 10.3389/fimmu.2018.02277] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 09/13/2018] [Indexed: 11/23/2022] Open
Abstract
Early plasmablast induction is a hallmark of Plasmodium infection and is thought to contribute to the control of acute parasite burden. Although long understood to be a T-cell dependent phenomenon, regulation of early plasmablast differentiation, however, is poorly understood. Here, we identify a population of CD4+ T cells that express the innate NK cell marker NK1.1 as an important source of T cell help for early plasmablast and parasite-specific Ab production. Interestingly, NK1.1+ CD4+ T cells arise from conventional, naive NK1.1− CD4+ T cells, and their generation is independent of CD1d but critically reliant on MHC-II. CD4+ T cells that express NK1.1 early after activation produce IFN-γ and IL-21, and express the follicular helper T (Tfh) cell markers ICOS, PD-1 and CXCR5 more frequently than NK1.1− CD4+ T cells. Further analysis of this population revealed that NK1.1+ Tfh-like cells were more regularly complexed with plasmablasts than NK1.1− Tfh-like cells. Ultimately, depletion of NK1.1+ cells impaired class-switched parasite-specific antibody production during early Plasmodium yoelii infection. Together, these data suggest that expression of NK1.1 defines a population of rapidly expanding effector CD4+ T cells that specifically promote plasmablast induction during Plasmodium infection and represent a subset of T cells whose modulation could promote effective vaccine design.
Collapse
Affiliation(s)
- Daniel J Wikenheiser
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Susie L Brown
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Juhyung Lee
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Jason S Stumhofer
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| |
Collapse
|
27
|
Sharapova TN, Ivanova OK, Prasolov VS, Romanova EA, Sashchenko LP, Yashin DV. Innate immunity protein Tag7 (PGRP-S) activates lymphocytes capable of Fasl-Fas-dependent contact killing of virus-infected cells. IUBMB Life 2017; 69:971-977. [PMID: 29083508 DOI: 10.1002/iub.1688] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Accepted: 10/05/2017] [Indexed: 12/12/2022]
Abstract
The innate immunity protein Tag7 (PGRP-S, PGLYRP1) is involved in antimicrobial and antitumor defense. As shown in our previous studies, Tag7 specifically interacts with the major heat shock protein Hsp70 to form a stable Tag7-Hsp70 complex with cytotoxic activity against tumor cells. A stable complex of Tag7 with the calcium-binding protein Mts1 (S100A4) stimulates migration of lymphocytes. Moreover, Tag7 can activate cytotoxic lymphocytes that recognize and kill HLA-negative tumor cells. Here, we have shown that Tag 7 treatment of human peripheral blood mononuclear cells (PBMCs) results in activation of different cytotoxic lymphocyte populations-natural killer (NK) cells and CD8+ NKG2D+ T lymphocytes-that kill Moloney murine leukemia virus (MMLV) infected SC-1 cells using different mechanisms of cell death induction. This mechanism in NK cells is based on the release of granzymes, which activate apoptosis in target cells, while CD8+ NKG2D+ T lymphocytes recognize the noncanonical MicA antigen on the surface of virus-containing cells and kill them via the FasL-Fas interaction, triggering the apoptotic or necroptotic cell death pathway. Preliminary incubation of PBMCs with virus-infected cells and following incubation with Tag7 results in activation of lymphocytes with a different phenotype. These lymphocytes change the spectrum of target cells and the mechanism of cell death induction, and their interaction with target cells is not species-specific. © 2017 IUBMB Life, 69(12):971-977, 2017.
Collapse
Affiliation(s)
| | | | - Vladimir S Prasolov
- Laboratory of Cell Biology, Engelhardt Institute of Molecular Biology RAS, Moscow, Russia
| | | | | | | |
Collapse
|
28
|
Pluangnooch P, Timalsena S, Wongkajornsilp A, Soontrapa K. Cytokine-induced killer cells: A novel treatment for allergic airway inflammation. PLoS One 2017; 12:e0186971. [PMID: 29073213 PMCID: PMC5658108 DOI: 10.1371/journal.pone.0186971] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Accepted: 10/11/2017] [Indexed: 12/25/2022] Open
Abstract
The effectiveness of cytokine-induced killer (CIK) cells for treatment of cancers has long been appreciated. Here, we report for the first time that CIK cells can be applied to treat allergic airway inflammation. Adopting from an established protocol with some modifications, we generated CIK cells ex vivo from mouse T cells, and examined their effectiveness in treatment of allergic airway inflammation using the ovalbumin-induced model of allergic airway inflammation. Based upon evaluation of bronchoalveolar lavage cellularity, T helper type2 cytokine levels and lung histology, all of which are important parameters for determining the severity of allergic airway inflammation, diseased mice treated with CIK cells showed significant reductions in all the parameters without any obvious adverse effects. Interestingly, the observed effects were comparable to those treated with dexamethasone. Thus, our study provides a novel application of CIK cells in treatment of allergic airway inflammation.
Collapse
Affiliation(s)
- Panwadee Pluangnooch
- Department of Pharmacology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Sunita Timalsena
- Department of Pharmacology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Adisak Wongkajornsilp
- Department of Pharmacology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Kitipong Soontrapa
- Department of Pharmacology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- * E-mail:
| |
Collapse
|
29
|
T cell subtypes and reciprocal inflammatory mediator expression differentiate P. falciparum memory recall responses in asymptomatic and symptomatic malaria patients in southeastern Haiti. PLoS One 2017; 12:e0174718. [PMID: 28369062 PMCID: PMC5378365 DOI: 10.1371/journal.pone.0174718] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Accepted: 03/14/2017] [Indexed: 12/20/2022] Open
Abstract
Asymptomatic Plasmodium falciparum infection is responsible for maintaining malarial disease within human populations in low transmission countries such as Haiti. Investigating differential host immune responses to the parasite as a potential underlying mechanism could help provide insight into this highly complex phenomenon and possibly identify asymptomatic individuals. We performed a cross-sectional analysis of individuals who were diagnosed with malaria in Sud-Est, Haiti by comparing the cellular and humoral responses of both symptomatic and asymptomatic subjects. Plasma samples were analyzed with a P. falciparum protein microarray, which demonstrated serologic reactivity to 3,877 P. falciparum proteins of known serologic reactivity; however, no antigen-antibody reactions delineating asymptomatics from symptomatics were identified. In contrast, differences in cellular responses were observed. Flow cytometric analysis of patient peripheral blood mononuclear cells co-cultured with P. falciparum infected erythrocytes demonstrated a statistically significant increase in the proportion of T regulatory cells (CD4+ CD25+ CD127-), and increases in unique populations of both NKT-like cells (CD3+ CD8+ CD56+) and CD8mid T cells in asymptomatics compared to symptomatics. Also, CD38+/HLA-DR+ expression on γδ T cells, CD8mid (CD56-) T cells, and CD8mid CD56+ NKT-like cells decreased upon exposure to infected erythrocytes in both groups. Cytometric bead analysis of the co-culture supernatants demonstrated an upregulation of monocyte-activating chemokines/cytokines in asymptomatics, while immunomodulatory soluble factors were elevated in symptomatics. Principal component analysis of these expression values revealed a distinct clustering of individual responses within their respective phenotypic groups. This is the first comprehensive investigation of immune responses to P. falciparum in Haiti, and describes unique cell-mediated immune repertoires that delineate individuals into asymptomatic and symptomatic phenotypes. Future investigations using large scale biological data sets analyzing multiple components of adaptive immunity, could collectively define which cellular responses and molecular correlates of disease outcome are malaria region specific, and which are truly generalizable features of asymptomatic Plasmodium immunity, a research goal of critical priority.
Collapse
|
30
|
Wang G, Yu G, Wang D, Guo S, Shan F. Comparison of the purity and vitality of natural killer cells with different isolation kits. Exp Ther Med 2017; 13:1875-1883. [PMID: 28565780 PMCID: PMC5443303 DOI: 10.3892/etm.2017.4189] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Accepted: 12/23/2016] [Indexed: 01/21/2023] Open
Abstract
Natural killer (NK) cells are innate lymphocytes that aid in the protection of the host from infectious diseases and cancer. In vitro studies of NK cells have provided a foundation for developing clinical adoptive NK-cell transferred immunotherapy against human tumors. To elucidate the functions and mechanisms of NK cell populations, it is important to develop an optimal, highly reproducible and reliable isolation method. The present comparative study was performed with four different NK cell isolation kits of magnetic bead labeling made by Miltenyi and Stemcell companies, including positive selection kits [cluster of differentiation (CD)-49b, using the monoclonal antibody DX5) MicroBeads] and negative selection kits. In addition, the viability of NK cells isinterleukin-2 (IL-2)-dependent in vitro and thus the concentration of IL-2 is critical for maintaining longer cell viability of NK cells. NK cell purity and viability after culturing, for 24, 48 or 72 h, with or without IL-2 (0, 100, 300 or 500 U/ml) was investigated in the present study. Purity of NK cells varied depending on the purification kit used, despite the same method being applied. Furthermore, more granulocytes were present in purified NK cells using Miltenyi sorting kits, particularly when using the negative selection kit. The main disadvantage of DX5-positive selection using the Stemcell and Miltenyi kits was that a high percentage of CD3ε+ cells were mixed into the isolated NK cells. Additionally, a significant difference of NK cell purity (P=0.003) was observed while purification was performed using different surface markers. As a consequence, the use of the positive selection kit was modified and subsequently a significantly higher purity (P=0.002) and yield (P=0.004) of NK cells was obtained. Moreover, the purity of NK cells and viability with or without a range of concentrations of IL-2 was compared. Results indicated that with a higher IL-2 concentration, the NK cell purity and viability were significantly higher (P<0.05). To our knowledge, this is the first report that has compared the disadvantages of four commercial NK cell isolation kits from two well-known companies, and identified the effect of NK cell purity and viability, using different concentrations of IL-2. To conclude, the results of the present study are fundamental in aiding the further development of NK cell therapy protocols for murine in vivo models.
Collapse
Affiliation(s)
- Guangchuan Wang
- Department of Immunology, School of Basic Medical Science, China Medical University, Shenyang, Liaoning 110122, P.R. China.,Department of Immunology, School of Basic Medical Science, Jinzhou Medical University, Jinzhou, Liaoning 121000, P.R. China
| | - Guang Yu
- Department of Immunology, School of Basic Medical Science, Jinzhou Medical University, Jinzhou, Liaoning 121000, P.R. China
| | - Dongmei Wang
- Department of Immunology, School of Basic Medical Science, China Medical University, Shenyang, Liaoning 110122, P.R. China
| | - Shengnan Guo
- Department of Immunology, School of Basic Medical Science, China Medical University, Shenyang, Liaoning 110122, P.R. China
| | - Fengping Shan
- Department of Immunology, School of Basic Medical Science, China Medical University, Shenyang, Liaoning 110122, P.R. China
| |
Collapse
|
31
|
de Andrés C, Fernández-Paredes L, Tejera-Alhambra M, Alonso B, Ramos-Medina R, Sánchez-Ramón S. Activation of Blood CD3 +CD56 +CD8 + T Cells during Pregnancy and Multiple Sclerosis. Front Immunol 2017; 8:196. [PMID: 28280497 PMCID: PMC5322280 DOI: 10.3389/fimmu.2017.00196] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Accepted: 02/09/2017] [Indexed: 01/24/2023] Open
Abstract
A striking common feature of most autoimmune diseases is their female predominance, with at least twice as common among women than men in relapsing–remitting multiple sclerosis (MS), the prevailing MS clinical form with onset at childbearing age. This fact, together with the protective effect on disease activity during pregnancy, when there are many biological changes including high levels of estrogens and progesterone, puts sex hormones under the spotlight. The role of natural killer (NK) and NKT cells in MS disease beginning and course is still to be elucidated. The uterine NK (uNK) cells are the most predominant immune population in early pregnancy, and the number and function of uNK cells infiltrating the endometrium are sex-hormones’ dependent. However, there is controversy on the role of estrogen or progesterone on circulating NK (CD56dim and CD56bright) and NKT cells’ subsets. Here, we show a significantly increased activation of CD3+CD56+CD8+ cells in pregnant MS women (MSP) compared with non-pregnant MS women (NPMS) (p < 0.001) and even with respect to healthy pregnant women (HP, p < 0.001), remaining increased even after delivery. The dynamics of expression of early activation marker CD69 on CD3+CD56+CD8+ cells showed a progressive statistically significant increase along the gestation trimesters (T) and at postpartum (PP) with respect to NPMS (1T: p = 0.018; 2T: p = 0.004; 3T: p < 0.001; PP: p = 0.001). In addition, early activation expression of CD69 on CD3+CD56+CD8+ cells was higher in MSP than HP in the first two trimesters of gestation (p = 0.004 and p = 0.015, respectively). NPMS showed significantly increased cytotoxic/regulatory NK ratio compared with healthy controls (p < 0.001). On the other hand, gender studies showed no differences between MS women and men in NK and CD3+CD56+CD8+ cells’ subsets. Our findings may add on the understanding of the regulatory axis in MS during pregnancy. Further studies on specific CD8+ NKT cells function and their role in pregnancy beneficial effects on MS are warranted to move forward more effective MS treatments.
Collapse
Affiliation(s)
- Clara de Andrés
- Department of Neurology, Hospital General Universitario Gregorio Marañón , Madrid , Spain
| | | | - Marta Tejera-Alhambra
- Department of Immunology, Hospital General Universitario Gregorio Marañón , Madrid , Spain
| | - Bárbara Alonso
- Department of Immunology, Hospital General Universitario Gregorio Marañón , Madrid , Spain
| | - Rocío Ramos-Medina
- Department of Immunology, Hospital General Universitario Gregorio Marañón , Madrid , Spain
| | - Silvia Sánchez-Ramón
- Department of Clinical Immunology, IdISSC, Hospital Clínico San Carlos, Madrid, Spain; Department of Microbiology I, Complutense University School of Medicine, Madrid, Spain
| |
Collapse
|
32
|
Pita-López ML, Pera A, Solana R. Adaptive Memory of Human NK-like CD8 + T-Cells to Aging, and Viral and Tumor Antigens. Front Immunol 2016; 7:616. [PMID: 28066426 PMCID: PMC5165258 DOI: 10.3389/fimmu.2016.00616] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Accepted: 12/06/2016] [Indexed: 12/13/2022] Open
Abstract
Human natural killer (NK)-like CD8+ T-cells are singular T-cells that express both T and NK cell markers such as CD56; their frequencies depend on their differentiation and activation during their lifetime. There is evidence of the presence of these innate CD8+ T-cells in the human umbilical cord, highlighting the necessity of investigating whether the NK-like CD8+ T-cells arise in the early stages of life (gestation). Based on the presence of cell surface markers, these cells have also been referred to as CD8+KIR+ T-cells, innate CD8+ T-cells, CD8+CD28−KIR+ T-cells or NKT-like CD8+CD56+ cells. However, the functional and co-signaling significance of these NK cell receptors on NK-like CD8+ T-cells is less clear. Also, the diverse array of costimulatory and co-inhibitory receptors are spatially and temporally regulated and may have distinct overlapping functions on NK-like CD8+ T-cell priming, activation, differentiation, and memory responses associated with different cell phenotypes. Currently, there is no consensus regarding the functional properties and phenotypic characterization of human NK-like CD8+ T-cells. Environmental factors, such as aging, autoimmunity, inflammation, viral antigen re-exposure, or the presence of persistent tumor antigens have been shown to allow differentiation (“adaptation”) of the NK-like CD8+ T-cells; the elucidation of this differentiation process and a greater understanding of the characteristics of these cells could be important for their eventual in potential therapeutic applications aimed at improving protective immunity. This review will attempt to elucidate an understanding of the characteristics of these cells with the goal toward their eventual use in potential therapeutic applications aimed at improving protective immunity.
Collapse
Affiliation(s)
- María Luisa Pita-López
- Research Center in Molecular Biology of Chronic Diseases (CIBIMEC), CUSUR University of Guadalajara , Guzmán , Mexico
| | - Alejandra Pera
- Clinical Division, Brighton and Sussex Medical School, University of Sussex, Brighton, UK; Maimonides Biomedicine Institute of Cordoba (IMIBIC), Reina Sofia Hospital, University of Córdoba, Córdoba, Spain
| | - Rafael Solana
- Maimonides Biomedicine Institute of Cordoba (IMIBIC), Reina Sofia Hospital, University of Córdoba , Córdoba , Spain
| |
Collapse
|