1
|
Qing J, Zhao Y, Wu J. The impact of rising peripheral blood naïve CD8 + T cell levels on chronic kidney disease onset: a Mendelian randomization study. Ren Fail 2025; 47:2486564. [PMID: 40230080 PMCID: PMC12001844 DOI: 10.1080/0886022x.2025.2486564] [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: 01/20/2025] [Revised: 03/11/2025] [Accepted: 03/13/2025] [Indexed: 04/16/2025] Open
Abstract
BACKGROUND The global incidence of chronic kidney disease (CKD) is rising rapidly. Immune cells play a crucial role in the onset and progression of CKD, however, the causal relationships and underlying immunological mechanisms remain incompletely elucidated. This deficiency hinders the development and application of early interventions and immunotherapies for CKD. METHODS In this study, we hypothesize that alterations in immune cell phenotypes (ICPs) in the blood may influence the onset of CKD. We collated Genome Wide Association Studies (GWAS) data for 731 ICPs, alongside summary data for CKD and estimated glomerular filtration rate (eGFR). Utilizing bidirectional mendelian randomization analysis (MR), we identified the impact of ICPs on the onset of CKD. RESULTS Preliminary MR analyses revealed three ICPs positively associated with CKD onset: the absolute number of CD45RA+ CD28- CD8+ T cells (p = 1.209 × 10-15, 95% CI: 1.0002-1.0003), the percentage of CD28+ CD45RA+ CD8+ T cells of total T cells (p = 5.831 × 10-6, 95% CI: 1.0028-1.0070), and the percentage of CD45RA- CD28- CD8+ T cells of total T cells (p = 4.292 × 10-5, 95% CI: 1.0005-1.0015). After conducting sensitivity and reverse MR analyses, only the percentage of CD28+ CD45RA+ CD8+ T cells (naïve CD8+ T Cells) was found to have a sufficiently robust causal impact on CKD. CONCLUSION We are the first to demonstrate a significant positive association between the percentage of naïve CD8+ T cells and CKD onset. This finding offers new insights for early prevention and treatment of CKD.
Collapse
Affiliation(s)
- Jianbo Qing
- Department of Nephrology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yiting Zhao
- Department of Nephrology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Junnan Wu
- Department of Nephrology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
2
|
Ren Q, Wang F, Du F, He C, Wang X, Wang J, Zhang Z, Sun Y. Asiaticoside enhances the anti-tumor effect of anti-PDL1 by regulating T cell activity through increasing LCK activity. Pathol Res Pract 2025; 271:155995. [PMID: 40373489 DOI: 10.1016/j.prp.2025.155995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 03/12/2025] [Accepted: 05/05/2025] [Indexed: 05/17/2025]
Abstract
Anti-PD-L1 antibody confers anti-tumor effects, but its long-term use can provoke resistance and adverse effects. Asiaticoside, a bioactive triterpene glycoside from Centella asiatica L., regulates immune function and induces apoptosis of hepatocellular carcinoma (HCC) cells. T cells play a vital role in killing tumor cells and require lymphocyte-specific protein tyrosine kinase (LCK) for activation. Here, we examined whether a combined asiaticoside and anti-PD-L1 treatment regulates T cells via LCK activation to enhance the anti-tumor effect in vivo. We established a subcutaneous mouse HCC model using Hepa1-6 cells and measured spleen and tumor weight. Morphological changes of tumor tissues were assessed by hematoxylin-eosin staining. Tumor cell apoptosis and proliferation were determined by TUNEL staining and KI67 immunohistochemistry. The proportion of activated T cells in the spleen was detected by flow cytometry, and the levels of phosphorylated p-LCK and p-AKT in the spleen were determined by Western blotting. Changes in the levels of serum inflammatory factors were detected with ELISA. Our results revealed that the combined asiaticoside and anti-PD-L1 treatment inhibited tumor growth by enhancing apoptosis and reducing tumor cell proliferation. The treatment activated T cells to increase the proportion of effector T cells in the spleen, evidenced by upregulated p-LCK and p-AKT levels. It also increased the level of TNF-α in the serum and decreased IL-6, implying an enhanced immune response. In conclusion, the combined asiaticoside and anti-PD-L1 treatment enhances the anti-HCC effect in vivo by promoting LCK activation to regulate T cells.
Collapse
Affiliation(s)
- Qingyi Ren
- Green Pharmaceutical Technology Key Laboratory of Luzhou City, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Fang Wang
- Green Pharmaceutical Technology Key Laboratory of Luzhou City, School of Pharmacy, Southwest Medical University, Luzhou, China; Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China; Central Nervous System Drug Key Laboratory of Sichuan Province
| | - Fei Du
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Chenxi He
- Green Pharmaceutical Technology Key Laboratory of Luzhou City, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Xiaodong Wang
- Department of Hepatobiliary Disease, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Jun Wang
- Green Pharmaceutical Technology Key Laboratory of Luzhou City, School of Pharmacy, Southwest Medical University, Luzhou, China; Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Zhuo Zhang
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China.
| | - Yuhong Sun
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China.
| |
Collapse
|
3
|
Wang Q, Wang S, Cui L, Zhang Y, Waterhouse GIN, Sun-Waterhouse D, Ma C, Kang W. Flammulina velutipes polysaccharide exerts immunomodulatory function involving RSAD2 to regulate the NF-κB/MAPK signaling pathway in RAW264.7 macrophage cells and in mouse spleen cells. Int J Biol Macromol 2025; 309:142985. [PMID: 40210026 DOI: 10.1016/j.ijbiomac.2025.142985] [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: 01/14/2025] [Revised: 04/06/2025] [Accepted: 04/07/2025] [Indexed: 04/12/2025]
Abstract
There are ongoing efforts to explore the potential of natural bioactive substances including polysaccharides in immunological regulation and understand the mechanisms under their immune-regulating function. In this study, a polysaccharide from Flammulina velutipes (FVP-1) exhibited immunomodulatory in RAW264.7 macrophage cells and mouse spleen cells. FVP-1 increased the secretion of cytokines (like TNF-α, IL-6 and IL-1β) and their mRNA expression, upregulated the transcription and translation expression of COX-2 and iNOS, and enhanced the release of reactive oxygen species the phagocytic activity in macrophages, thereby promoting the maturation and transformation of certain lymphocytes. All these functions of FVP-1 depended to some extent on its concentration. The RSAD2 effector was involved in the immunomodulatory function of FVP-1 towards macrophages and mouse splenocytes, through mediating FVP-1's activation and regulation of the NF-κB/MAPK signaling pathway. These findings indicate the potential of FVP-1 as a natural immunomodulator and approach for improving immune function.
Collapse
Affiliation(s)
- Qiuyi Wang
- National R & D Center for Edible Fungus Processing Technology, Henan University, Kaifeng 475004, China
| | - Senye Wang
- National R & D Center for Edible Fungus Processing Technology, Henan University, Kaifeng 475004, China
| | - Lili Cui
- National R & D Center for Edible Fungus Processing Technology, Henan University, Kaifeng 475004, China; Joint International Research Laboratory of Food & Medicine Resource Function, Kaifeng 475004, China
| | - Yu Zhang
- National R & D Center for Edible Fungus Processing Technology, Henan University, Kaifeng 475004, China
| | - Geoffrey I N Waterhouse
- National R & D Center for Edible Fungus Processing Technology, Henan University, Kaifeng 475004, China; Joint International Research Laboratory of Food & Medicine Resource Function, Kaifeng 475004, China; School of Chemical Sciences, the University of Auckland, Auckland 1142, New Zealand
| | - Dongxiao Sun-Waterhouse
- National R & D Center for Edible Fungus Processing Technology, Henan University, Kaifeng 475004, China; Joint International Research Laboratory of Food & Medicine Resource Function, Kaifeng 475004, China; School of Chemical Sciences, the University of Auckland, Auckland 1142, New Zealand.
| | - Changyang Ma
- National R & D Center for Edible Fungus Processing Technology, Henan University, Kaifeng 475004, China; Joint International Research Laboratory of Food & Medicine Resource Function, Kaifeng 475004, China; Functional Food Engineering Technology Research Center, Kaifeng 475004, China; College of Agriculture, Henan University, Kaifeng 475004, China.
| | - Wenyi Kang
- National R & D Center for Edible Fungus Processing Technology, Henan University, Kaifeng 475004, China; Joint International Research Laboratory of Food & Medicine Resource Function, Kaifeng 475004, China; Functional Food Engineering Technology Research Center, Kaifeng 475004, China; College of Agriculture, Henan University, Kaifeng 475004, China.
| |
Collapse
|
4
|
Qin C, Yan Z, Yu Q, Chen M, Hu T, Wang X, Lei B, Chen Y, Ma K, Deng Z, Cheng S. Exploring the Relationship Between Immune Cells and Scoliosis by Mendelian Randomization, Colocalization Analysis, and SMR. Mediators Inflamm 2025; 2025:8833556. [PMID: 40177401 PMCID: PMC11964722 DOI: 10.1155/mi/8833556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Accepted: 02/27/2025] [Indexed: 04/05/2025] Open
Abstract
Background: Scoliosis is a condition that can have severe consequences for millions of individuals on an annual basis. Current research in this field is increasingly focusing on the role of the immune system in the development of the disease. However, the precise relationship between immunity and scoliosis remains to be fully elucidated. Method: Our investigation involved a comprehensive Mendelian randomization (MR) analysis to explore the potential causal relationship between immune cells and scoliosis. The comprehensive univariable MR analysis encompassed 731 immune cells to explore their relationship with scoliosis. Cochran's Q test, the leave-one-out test, and MR-Egger intercept analysis were used to assess pleiotropy and heterogeneity. We performed multivariable MR analysis to account for potential confounding factors between the immune cells. The colocalization analysis and summary data-based MR (SMR) analysis were utilized to explore relationship between immune cells and cis-eQTL. Results: Our study identified 13 immune cells that were significantly associated with scoliosis by univariable MR, including four risk factors and nine protective factors for scoliosis. In order to reduce confounding between immune cells, multivariable MR was employed, and it was determined that only six immune cell types had independent effects on scoliosis. SERPINH1 shared the same variant with CX3CR1 on CD14- CD16-. FSD1L shared the same variant with CCR2 on CD14- CD16-. SNHG14, SNORA33, NET1, and SNORD100 shared the same variant with HLA DR on CD14+ CD16+ monocyte. Conclusion: Our findings suggested a possible link between immune cells and scoliosis and found the key genes for the immune cell, which provides a new direction for further research. However, the specific underlying mechanisms require further investigation in future experiments.
Collapse
Affiliation(s)
- Chaofan Qin
- Department of Orthopedics, Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Zhengjian Yan
- Department of Orthopedics, Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Qingshuai Yu
- Department of Orthopedics, Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Mingxin Chen
- Department of Orthopedics, Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Tao Hu
- Department of Orthopedics, Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Xin Wang
- Department of Orthopedics, Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Bo Lei
- Department of Orthopedics, Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Yu Chen
- Department of Orthopedics, Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Ke Ma
- Department of Orthopedics, Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Zhongliang Deng
- Department of Orthopedics, Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Si Cheng
- Department of Orthopedics, Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| |
Collapse
|
5
|
Graciliano NG, Goulart MOF, de Oliveira ACM. Impact of Maternal Exposure to SARS-CoV-2 on Immunological Components of Breast Milk. Int J Mol Sci 2025; 26:2600. [PMID: 40141241 PMCID: PMC11942142 DOI: 10.3390/ijms26062600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2025] [Revised: 03/07/2025] [Accepted: 03/09/2025] [Indexed: 03/28/2025] Open
Abstract
COVID-19, caused by SARS-CoV-2, has become a global public health threat. Although no replication-competent virus has been found in breast milk samples, breastfeeding practices during the pandemic were impacted. It is well known that breast milk is adapted to meet the needs of infants, providing the appropriate amounts of nutrients and various bioactive compounds that contribute to the maturation of the immune system and antioxidant protection, safeguarding infants against diseases. While its composition is variable, breast milk contains immune cells, antibodies, and cytokines, which have anti-inflammatory, pro-inflammatory, antiviral, and antibacterial properties that strengthen infant immunity. Since COVID-19 vaccines have not yet been approved for infants under six months of age, newborns rely on the passive transfer of antibodies via the placenta and breast milk to protect them against severe SARS-CoV-2 infection. Several studies that analyzed breast milk samples in the context of COVID-19 have demonstrated that a strong antibody response is induced following maternal infection with SARS-CoV-2. Therefore, this review aims to provide a comprehensive overview of the impact of maternal exposure to SARS-CoV-2 through natural infection and/or vaccination on the immunological composition of breast milk based on the studies conducted on this topic.
Collapse
Affiliation(s)
- Nayara Gomes Graciliano
- Institute of Biological and Health Sciences, Federal University of Alagoas, Maceió 57072-900, Alagoas, Brazil
| | - Marília Oliveira Fonseca Goulart
- Institute of Biological and Health Sciences, Federal University of Alagoas, Maceió 57072-900, Alagoas, Brazil
- Institute of Chemistry and Biotechnology, Federal University of Alagoas, Maceió 57072-900, Alagoas, Brazil
| | - Alane Cabral Menezes de Oliveira
- Institute of Biological and Health Sciences, Federal University of Alagoas, Maceió 57072-900, Alagoas, Brazil
- College of Nutrition, Federal University of Alagoas, Maceió 57072-900, Alagoas, Brazil
| |
Collapse
|
6
|
Cajanding RJM. Implementation of chimeric antigen receptor (CAR) T-cell therapy in the NHS: prospects, promises and pitfalls. BRITISH JOURNAL OF NURSING (MARK ALLEN PUBLISHING) 2025; 34:S20-S30. [PMID: 40063539 DOI: 10.12968/bjon.2024.0056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/13/2025]
Abstract
The approval, introduction, and provision of chimeric antigen receptor (CAR) T-cell therapy in the UK NHS presents a innovative and revolutionary approach in cancer treatment and management. CAR T-cell therapy is a highly specialised and personalised type of immunotherapy that involves reprogramming a patient's immune system by synthetically modifying their T-cells to specifically target and eliminate cancer cells. This therapy offers the potential to cure malignancies that were previously deemed incurable or refractory to conventional chemotherapy. CAR T-cell therapy, however, is associated with significant risks and life-threatening complications, and it entails substantial financial cost. The implementation of CAR T-cell therapy in the NHS marks a new era of personalised medicine, offering a promising approach not only for improving cancer outcomes, but for enhancing survivorship and quality of life among patients with advanced and relapsing haematologic malignancies.
Collapse
|
7
|
Lu X, Xv Y, Hu W, Sun B, Hu H. Targeting CD4+ T cells through gut microbiota: therapeutic potential of traditional Chinese medicine in inflammatory bowel disease. Front Cell Infect Microbiol 2025; 15:1557331. [PMID: 40099014 PMCID: PMC11911530 DOI: 10.3389/fcimb.2025.1557331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Accepted: 02/18/2025] [Indexed: 03/19/2025] Open
Abstract
Inflammatory Bowel Disease (IBD) is an autoimmune disease characterized by chronic relapsing inflammation of the intestinal tract. Gut microbiota (GM) and CD4+T cells are important in the development of IBD. A lot of studies have shown that GM and their metabolites like short-chain fatty acids, bile acids and tryptophan can be involved in the differentiation of CD4+T cells through various mechanisms, which in turn regulate the immune homeostasis of the IBD patients. Therefore, regulating CD4+T cells through GM may be a potential therapeutic direction for the treatment of IBD. Many studies have shown that Traditional Chinese Medicine (TCM) formulas and some herbal extracts can affect CD4+T cell differentiation by regulating GM and its metabolites. In this review, we mainly focus on the role of GM and their metabolites in regulating the differentiation of CD4+T cells and their correlation with IBD. We also summarize the current research progress on the regulation of this process by TCM.
Collapse
Affiliation(s)
- Xingyao Lu
- Department of Gastroenterology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yichuan Xv
- Department of Gastroenterology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Weiye Hu
- Department of Liver Disease, Shanghai Yueyang Integrated Traditional Chinese Medicine and Western Medicine Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Boyun Sun
- Department of Gastroenterology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hongyi Hu
- Department of Gastroenterology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
8
|
Magalhães MI, Azevedo MJ, Castro F, Oliveira MJ, Costa ÂM, Sampaio Maia B. The link between obesity and the gut microbiota and immune system in early-life. Crit Rev Microbiol 2025; 51:264-284. [PMID: 38651972 DOI: 10.1080/1040841x.2024.2342427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Revised: 04/04/2024] [Accepted: 04/06/2024] [Indexed: 04/25/2024]
Abstract
In early-life, the gut microbiota is highly modifiable, being modulated by external factors such as maternal microbiota, mode of delivery, and feeding strategies. The composition of the child's gut microbiota will deeply impact the development and maturation of its immune system, with consequences for future health. As one of the main sources of microorganisms to the child, the mother represents a crucial factor in the establishment of early-life microbiota, impacting the infant's wellbeing. Recent studies have proposed that dysbiotic maternal gut microbiota could be transmitted to the offspring, influencing the development of its immunity, and leading to the development of diseases such as obesity. This paper aims to review recent findings in gut microbiota and immune system interaction in early-life, highlighting the benefits of a balanced gut microbiota in the regulation of the immune system.
Collapse
Affiliation(s)
- Maria Inês Magalhães
- Doctoral Program in Biomedical Sciences, ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
- Nephrology and Infectious Diseases R&D group, i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Tumor and Microenvironment Interactions group, i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- nBTT, NanoBiomaterials for Targeted Therapies group, i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- FMDUP - Faculdade de Medicina Dentária da Universidade do Porto, Porto, Portugal
| | - Maria João Azevedo
- Nephrology and Infectious Diseases R&D group, i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- nBTT, NanoBiomaterials for Targeted Therapies group, i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- FMDUP - Faculdade de Medicina Dentária da Universidade do Porto, Porto, Portugal
- Academic Center for Dentistry Amsterdam (ACTA), Universiteit van Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Flávia Castro
- Tumor and Microenvironment Interactions group, i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Maria José Oliveira
- Tumor and Microenvironment Interactions group, i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Ângela M Costa
- Tumor and Microenvironment Interactions group, i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Benedita Sampaio Maia
- Nephrology and Infectious Diseases R&D group, i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- nBTT, NanoBiomaterials for Targeted Therapies group, i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- FMDUP - Faculdade de Medicina Dentária da Universidade do Porto, Porto, Portugal
| |
Collapse
|
9
|
Čelakovská J, Čermáková E, Boudková P, Andýs C, Krejsek J. Differences in immunological profile in atopic dermatitis patients with and without dupilumab therapy. Immunol Med 2025; 48:33-46. [PMID: 39148480 DOI: 10.1080/25785826.2024.2387882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 07/23/2024] [Indexed: 08/17/2024] Open
Abstract
Our aim is to determine the number of leukocytes, T lymphocytes and B lymphocytes and the expression of activation markers CD200 and CD23 on B lymphocytes in atopic dermatitis (AD) patients (treated and not treated with dupilumab) during the pollen season. We examined 29 patients not treated with dupilumab, 24 patients treated with dupilumab and 40 healthy subjects as a control group. The count of T and B lymphocytes and their subsets were assessed by flow cytometry. The non-parametric Kruskal-Wallis one-factor analysis of variance with post hoc by Dunn's test with Bonferroni's modification was used for statistical processing. Although there was a significant improvement in skin findings in patients treated with dupilumab, the changes in immunological profile show a persistent altered immune response characterized by dysregulation and overactivation of B lymphocytes. Dupilumab therapy leads to normalization of relative T regulatory lymphocytes and total memory B lymphocytes and to decreased count of absolute CD8+ T lymphocytes. Why carry out this study?Studies investigating the immunological profile of atopic dermatitis (AD) patients during the pollen season are rare. There are no studies investigating the count of B lymphocytes (CD5+, CD22+ and CD73+ B lymphocytes) and the expression of activation markers CD23 and CD200 on B lymphocytes and on their subsets during pollen season in AD patients treated and non-treated with dupilumab therapy.What was learned from the study?In atopic dermatitis (AD) patients with and without dupilumab therapy, we confirmed the significantly higher count of absolute neutrophils, absolute monocytes, absolute eosinophils, absolute basophils, non-switched B lymphocytes, transitional B lymphocytes, CD23 memory, naive, non-switched, switched and total CD23 B lymphocytes, the relative count of CD200 memory and CD200 switched B lymphocytes.In dupilumab treated patients, we confirmed the significantly higher count of relative eosinophils, relative CD16+ eosinophils, relative CD200 non-switched B lymphocytes and lower count of absolute CD8+ T lymphocytes. Further studies should focus on investigating the effect of dupilumab on CD8+ T lymphocytes and their subpopulations.In patients without dupilumab therapy, we confirmed the significantly higher count of relative neutrophils, relative T regulatory lymphocytes and total memory B lymphocytes.The changes in the count of CD5+, CD22+ and CD73+ B lymphocytes were not observed during pollen season in both groups of AD patients.
Collapse
Affiliation(s)
- Jarmila Čelakovská
- Department of Dermatology and Venereology, Faculty Hospital and Medical Faculty of Charles University, Hradec Králové, Czech Republic
| | - Eva Čermáková
- Department of Medical Biophysics, Medical Faculty of Charles University, Hradec Králové, Czech republic
| | - Petra Boudková
- Department of Clinical Immunology and Allergy, Faculty Hospital and Medical Faculty of Charles University, Hradec Králové, Czech Republic
| | - Ctirad Andýs
- Department of Clinical Immunology and Allergy, Faculty Hospital and Medical Faculty of Charles University, Hradec Králové, Czech Republic
| | - Jan Krejsek
- Department of Clinical Immunology and Allergy, Faculty Hospital and Medical Faculty of Charles University, Hradec Králové, Czech Republic
| |
Collapse
|
10
|
Jin W, Wang J, Cao H, Shen X, Yang Y, Lv L. Effects of sufentanil on immune response, pain mediators and brain-sparing effect in patients with breast cancer undergoing radical mastectomy. BMC Surg 2025; 25:80. [PMID: 39994609 PMCID: PMC11849331 DOI: 10.1186/s12893-025-02814-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Accepted: 02/14/2025] [Indexed: 02/26/2025] Open
Abstract
OBJECTIVE To investigate the effects of sufentanil on immune response, pain mediators and brain-sparing effect in patients with breast cancer undergoing radical mastectomy. METHODS This study was a single center retrospective cohort study. The 118 study subjects were diagnosed and treated in our hospital from the period of January 2020-October 2022, who planned to undergo radical surgery for breast cancer. According to the different surgical drugs, these subjects were divided into sufentanil group and the control group, with 59 cases each. The visual analog scores (VAS) of patients in two groups were compared at 24 hour and 48 hour after surgery. The immune response indexes (including CD3+, CD4+, CD8+, CD4+/CD8+), pain mediators (β-endorphin, substance P and 5-hydroxytryptophan), brain-sparing effect indexes [arterio-venous oxygen content difference (Da-jvO2), jugular bulb venous saturation (S-jvO2), cerebral oxygen uptake (CEO2) and the Mini Mental State Scale (MMSE)], and brain damage indexes [S100 calcium-binding protein B (S100B) and neuron-specific enolase (NSE)] in two groups were compared. The incidence of adverse reactions in two groups was compared. RESULTS VAS scores were obviously lower in the sufentanil group than the control group at 24 hour and 48 hour postoperatively (P<0.001). Compared with the control group, the sufentanil group had higher CD3+, CD4+, CD4+/CD8+, MMSE scores, and lower content of CD8+, β-Endorphins, substance P, 5-hydroxytryptophan, Da-jvO2, S-jvO2 and CEO2 at 24 hour and 48 hour postoperatively (P<0.05). Patients in the sufentanil group had lower levels of S100B and NSE than the control group on the 1st and 7th day after surgery (P<0.01). The incidence of gastrointestinal reactions, hypertension and chills was significantly lower in the sufentanil group than the control group (P<0.05). CONCLUSION The application of sufentanil in breast cancer radical surgery effectively improved the immune function of the body, reduced pain response, alleviated brain damage, and had a certain brain-sparing effect.
Collapse
Affiliation(s)
- Weicheng Jin
- Department of anesthesiology, The Ninth People's Hospital of Suzhou, TaihuXincheng Town, No. 2666 Ludang RoadWujiang District, Suzhou, 215200, Jiangsu, China
| | - Jie Wang
- Department of anesthesiology, The Ninth People's Hospital of Suzhou, TaihuXincheng Town, No. 2666 Ludang RoadWujiang District, Suzhou, 215200, Jiangsu, China
| | - Hui Cao
- Department of anesthesiology, The Ninth People's Hospital of Suzhou, TaihuXincheng Town, No. 2666 Ludang RoadWujiang District, Suzhou, 215200, Jiangsu, China
| | - Xiaoping Shen
- Department of anesthesiology, The Ninth People's Hospital of Suzhou, TaihuXincheng Town, No. 2666 Ludang RoadWujiang District, Suzhou, 215200, Jiangsu, China
| | - Yang Yang
- Department of anesthesiology, The Ninth People's Hospital of Suzhou, TaihuXincheng Town, No. 2666 Ludang RoadWujiang District, Suzhou, 215200, Jiangsu, China
| | - Lanqing Lv
- Department of anesthesiology, The Ninth People's Hospital of Suzhou, TaihuXincheng Town, No. 2666 Ludang RoadWujiang District, Suzhou, 215200, Jiangsu, China.
| |
Collapse
|
11
|
Bartold M, Ivanovski S. Biological processes and factors involved in soft and hard tissue healing. Periodontol 2000 2025; 97:16-42. [PMID: 38243683 PMCID: PMC11808446 DOI: 10.1111/prd.12546] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Revised: 11/12/2023] [Accepted: 11/23/2023] [Indexed: 01/21/2024]
Abstract
Wound healing is a complex and iterative process involving myriad cellular and biologic processes that are highly regulated to allow satisfactory repair and regeneration of damaged tissues. This review is intended to be an introductory chapter in a volume focusing on the use of platelet concentrates for tissue regeneration. In order to fully appreciate the clinical utility of these preparations, a sound understanding of the processes and factors involved in soft and hard tissue healing. This encompasses an appreciation of the cellular and biological mediators of both soft and hard tissues in general as well as specific consideration of the periodontal tissues. In light of good advances in this basic knowledge, there have been improvements in clinical strategies and therapeutic management of wound repair and regeneration. The use of platelet concentrates for tissue regeneration offers one such strategy and is based on the principles of cellular and biologic principles of wound repair discussed in this review.
Collapse
Affiliation(s)
- Mark Bartold
- University of QueenslandBrisbaneQueenslandAustralia
| | | |
Collapse
|
12
|
Wang Y, Fang X, Liu J, Lv X, Lu K, Lu Y, Jiang Y. PCSK9 in T-cell function and the immune response. Biomark Res 2024; 12:163. [PMID: 39736777 DOI: 10.1186/s40364-024-00712-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 12/19/2024] [Indexed: 01/01/2025] Open
Abstract
Proprotein convertase subtilisin/kexin type 9 (PCSK9) was first reported in 2003 and confirmed to be strongly associated with familial hypercholesterolemia. Small-molecule inhibitors targeting PCSK9 provide an effective and safe method for managing hypercholesterolemia and reducing the cardiovascular risk. In recent years, increasing evidence has indicated other important roles for PCSK9 in inflammation, tumors, and even immune regulation. PCSK9 might be an attractive regulator of T-cell activation and expansion. It might mediate inflammation and regulate other types of immune cells. In this review, we summarize the current advances in the field of PCSK9 and provide a narrative of the biological processes associated with PCSK9. The relationships between PCSK9 and different T cells were investigated in depth. Finally, the signaling pathways associated with PCSK9 and the immune response are also summarized in this review.
Collapse
Affiliation(s)
- Yuying Wang
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Shandong, 250021, China
| | - Xiaosheng Fang
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Shandong, 250021, China
| | - Jiarui Liu
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Shandong, 250021, China
| | - Xiao Lv
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Shandong, 250021, China
| | - Kang Lu
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Shandong, 250021, China
| | - Yingxue Lu
- Department of Nephrology, Shandong Second Provincial General Hospital, Jinan , Shandong, 250021, China
| | - Yujie Jiang
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Shandong, 250021, China.
| |
Collapse
|
13
|
Kalim M, Jing R, Guo W, Xing H, Lu Y. Functional diversity and regulation of IL-9-producing T cells in cancer immunotherapy. Cancer Lett 2024; 606:217306. [PMID: 39426662 PMCID: PMC11675864 DOI: 10.1016/j.canlet.2024.217306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 10/15/2024] [Accepted: 10/17/2024] [Indexed: 10/21/2024]
Abstract
IL-9-producing T cells (T9) regulate immunological responses that affect various cellular biological processes, though their precise function remains fully understood. Previous studies have linked T9 cells to conditions such as allergic disorders, parasitic infection clearance, and various types of cancers. While the functional heterogeneity of IL-9 and T9 cells in cancer development has been documented, these cells present promising therapeutic opportunities for treating solid tumors. This review highlights the roles of IL-9 and T9 cells in cancer progression and treatment responses, focusing on potential discrepancies in IL-9/IL-9R signaling between murine tumors and cancer patients. Additionally, we discuss the regulation of tumor-specific Th9/Tc9 cell differentiation, the therapeutic potential of these cells, and current strategies to enhance their anti-tumor activities.
Collapse
Affiliation(s)
- Muhammad Kalim
- Houston Methodist Cancer Center/Weill Cornell Medicine, Houston, TX, 77030, USA
| | - Rui Jing
- Houston Methodist Cancer Center/Weill Cornell Medicine, Houston, TX, 77030, USA
| | - Wei Guo
- Houston Methodist Cancer Center/Weill Cornell Medicine, Houston, TX, 77030, USA
| | - Hui Xing
- Houston Methodist Cancer Center/Weill Cornell Medicine, Houston, TX, 77030, USA
| | - Yong Lu
- Houston Methodist Cancer Center/Weill Cornell Medicine, Houston, TX, 77030, USA.
| |
Collapse
|
14
|
Gao J, Liu Y, Tao L, Zeng P, Ye G, Zheng Y, Zhang N. Single-cell data revealed the regulatory mechanism of TNK cell heterogeneity in liver metastasis from gastric cancer. Discov Oncol 2024; 15:664. [PMID: 39549183 PMCID: PMC11569111 DOI: 10.1007/s12672-024-01528-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 11/04/2024] [Indexed: 11/18/2024] Open
Abstract
AIM The present work set out to classify cell subpopulations related to liver metastasis from gastric cancer (GC) and the mechanisms of their interactions with other immune cell subpopulations. BACKGROUND GC is characterized by a high degree of heterogeneity and liver metastasis. Exploring the mechanism of liver metastasis of GC from the perspective of heterogeneity of the tumor microenvironment (TME) might help improve the efficacy of GC treatment. OBJECTIVE Based on the cellular subpopulation characteristics of GC with liver metastasis, the regulatory mechanisms contributing to GC progression were analyzed, with special focuses on the roles of signaling pathways, transcription factors (TFs) and ligand-receptor pairs. METHODS The GSE163558 dataset was downloaded from the Gene Expression Omnibus (GEO) database to collect single-cell transcriptomic data of GC patients and their metastasis groups for cell clustering and relevant analyses. Differentially expressed genes (DEGs) in the GC and GC liver metastasis groups were screened and subjected to Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis. SCENIC analysis was used to mine TFs that affected cellular subpopulations during liver metastasis from GC. The relative expression levels of TFs in GC were determined using qRT-PCR. Transwell and wound healing assays were utilized to verify the regulation of the TFs on the migration and invasion of GC cells. Interaction network between the cellular subpopulations was developed applying CellChat. RESULTS Single-cell clustering was performed to group six major cell subpopulations, namely, Myeloid cells, B cells, Mast cells, Epithelial cells, Fibroblasts, and TNK cells, among which the number of TNK cells was significantly increased in the GC liver metastasis group. Differentially enriched pathways of TNK cells between GC and GC liver metastasis groups mainly included IL-17 and Pi3k-Akt signaling pathways. TNK cell subsets could be further categorized into CD8 T cells, Exhausted T cells, NK cells, NKT cells, and Treg cells, with the GC liver metastasis group showing significantly more CD8 T cells and NKT cells. FOS and JUNB were the TFs of TNK cell marker genes that contributed to liver metastasis from GC and the invasion and migration of GC cell lines. Significant differences in immune cell communication ligand-receptor pairs existed between the GC and GC liver metastasis groups. CONCLUSION This study revealed the critical role of TNK cell subsets in GC with liver metastasis applying single-cell transcriptomics analysis. The findings provided an important theoretical basis for developing novel therapies to inhibit liver metastasis from GC.
Collapse
Affiliation(s)
- Jun Gao
- Department of Gastrointestinal Surgery, Jiangxi Province Hospital of Integrated Chinese and Western Medicine, Nanchang, 330003, China
| | - Yujuan Liu
- Department of Emergency, Jiangxi Province Hospital of Integrated Chinese and Western Medicine, Nanchang, 330003, China
| | - Lu Tao
- Department of Emergency, Jiangxi Province Hospital of Integrated Chinese and Western Medicine, Nanchang, 330003, China
| | - Peng Zeng
- Department of Emergency, Jiangxi Province Hospital of Integrated Chinese and Western Medicine, Nanchang, 330003, China
| | - Guiying Ye
- Department of Emergency, Jiangxi Province Hospital of Integrated Chinese and Western Medicine, Nanchang, 330003, China
| | - Ying Zheng
- Department of Emergency, Jiangxi Province Hospital of Integrated Chinese and Western Medicine, Nanchang, 330003, China
| | - Nai Zhang
- Department of Emergency, Jiangxi Province Hospital of Integrated Chinese and Western Medicine, Nanchang, 330003, China.
| |
Collapse
|
15
|
Yang C, Li S, Chen D, Liu D, Yang Y, Guo H, Sun N, Bai X, Li G, Zhang R, Wang T, Zhang L, Peng L, Liu S, Zhang W, Zhao G, Tu X, Tian W. IMM2520, a novel anti-CD47/PD-L1 bispecific antibody for cancer immune therapy. Heliyon 2024; 10:e39858. [PMID: 39553551 PMCID: PMC11564011 DOI: 10.1016/j.heliyon.2024.e39858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 10/17/2024] [Accepted: 10/24/2024] [Indexed: 11/19/2024] Open
Abstract
PD-1/PD-L1 is an important signaling pathway in the adaptive immune system. The CD47/SIRPα signaling pathway is a crucial "do not eat me" signal for innate immunity. This study evaluated the anti-tumor mechanism of IMM2520 in vitro and in vivo. IMM2520 was generated using the "mab-trap" platform. IMM2520 showed high affinity to PD-L1 and relatively lower affinity to CD47, displaying preferential binding to PD-L1 on tumor cells. IMM2520 had the potent ability to inhibit the PD-1/PD-L1 and CD47/SIRPα signaling pathways and killed tumor cells through ADCC and ADCP. Importantly, IMM2520 did not bind to human red blood cells or induce erythrocyte agglutination. IMM2520 demonstrated a tendency to bind to CD47+/PD-L1+ tumor cells, reducing its binding to CD47 single-positive cells. In mouse transplantation models, compared with the first-generation CD47/PD-L1 BsAb (IMM2505), IMM2520 exhibited stronger and dose-dependent antitumor activity. These findings imply that IMM2520 may offer a novel therapeutic alternative for cancer patients.
Collapse
Affiliation(s)
- Chunmei Yang
- Department of R&D, ImmuneOnco Biopharmaceuticals (Shanghai) Inc., Shanghai, 201203, China
| | - Song Li
- Department of R&D, ImmuneOnco Biopharmaceuticals (Shanghai) Inc., Shanghai, 201203, China
| | - Dianze Chen
- Department of R&D, ImmuneOnco Biopharmaceuticals (Shanghai) Inc., Shanghai, 201203, China
| | - Dandan Liu
- Department of R&D, ImmuneOnco Biopharmaceuticals (Shanghai) Inc., Shanghai, 201203, China
| | - Yanan Yang
- Department of R&D, ImmuneOnco Biopharmaceuticals (Shanghai) Inc., Shanghai, 201203, China
| | - Huiqin Guo
- Department of R&D, ImmuneOnco Biopharmaceuticals (Shanghai) Inc., Shanghai, 201203, China
| | - Nana Sun
- Department of R&D, ImmuneOnco Biopharmaceuticals (Shanghai) Inc., Shanghai, 201203, China
| | - Xing Bai
- Department of R&D, ImmuneOnco Biopharmaceuticals (Shanghai) Inc., Shanghai, 201203, China
| | - Guanghui Li
- Department of R&D, ImmuneOnco Biopharmaceuticals (Shanghai) Inc., Shanghai, 201203, China
| | - Ruliang Zhang
- Department of CMC, ImmuneOnco Biopharmaceuticals (Shanghai) Inc., Shanghai, 201203, China
| | - Tianxiang Wang
- Department of CMC, ImmuneOnco Biopharmaceuticals (Shanghai) Inc., Shanghai, 201203, China
| | - Li Zhang
- Department of CMC, ImmuneOnco Biopharmaceuticals (Shanghai) Inc., Shanghai, 201203, China
| | - Liang Peng
- Department of CMC, ImmuneOnco Biopharmaceuticals (Shanghai) Inc., Shanghai, 201203, China
| | - Sijin Liu
- Department of CMC, ImmuneOnco Biopharmaceuticals (Shanghai) Inc., Shanghai, 201203, China
| | - Wei Zhang
- Department of CMC, ImmuneOnco Biopharmaceuticals (Shanghai) Inc., Shanghai, 201203, China
| | - Gui Zhao
- Department of CMC, ImmuneOnco Biopharmaceuticals (Shanghai) Inc., Shanghai, 201203, China
| | - Xiaoping Tu
- Department of CMC, ImmuneOnco Biopharmaceuticals (Shanghai) Inc., Shanghai, 201203, China
| | - Wenzhi Tian
- Department of R&D, ImmuneOnco Biopharmaceuticals (Shanghai) Inc., Shanghai, 201203, China
- Department of CMC, ImmuneOnco Biopharmaceuticals (Shanghai) Inc., Shanghai, 201203, China
| |
Collapse
|
16
|
Wang X, Hu H, Yan G, Zheng B, Luo J, Fan J. Identification and validation of interferon-stimulated gene 15 as a biomarker for dermatomyositis by integrated bioinformatics analysis and machine learning. Front Immunol 2024; 15:1429817. [PMID: 39559355 PMCID: PMC11570269 DOI: 10.3389/fimmu.2024.1429817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 10/14/2024] [Indexed: 11/20/2024] Open
Abstract
Background Dermatomyositis (DM) is an autoimmune disease that primarily affects the skin and muscles. It can lead to increased mortality, particularly when patients develop associated malignancies or experience fatal complications such as pulmonary fibrosis. Identifying reliable biomarkers is essential for the early diagnosis and treatment of DM. This study aims to identify and validate pivotal diagnostic biomarker for DM through integrated bioinformatics analysis and clinical sample validation. Methods Gene expression datasets GSE46239 and GSE142807 from the Gene Expression Omnibus (GEO) database were merged for analysis. Differentially expressed genes (DEGs) were identified and subjected to enrichment analysis. Advanced machine learning methods were utilized to further pinpoint hub genes. Weighted gene co-expression network analysis (WGCNA) was also conducted to discover key gene modules. Subsequently, we derived intersection gene from these methods. The diagnostic performance of the candidate biomarker was evaluated using analysis with dataset GSE128314 and confirmed by immunohistochemistry (IHC) in skin lesion biopsy specimens. The CIBERSORT algorithm was used to analyze immune cell infiltration patterns in DM, then the association between the hub gene and immune cells was investigated. Gene set enrichment analysis (GSEA) was performed to understand the biomarker's biological functions. Finally, the drug-gene interactions were predicted using the DrugRep server. Results Interferon-stimulated gene 15 (ISG15) was identified by intersecting DEGs, advanced machine learning-selected genes and key module genes from WGCNA. ROC analysis showed ISG15 had a high Area under the curve (AUC) of 0.950. IHC findings confirmed uniformly positive expression of ISG15, particularly in perivascular regions and lymphocytes, contrasting with universally negative expression in controls. Further analysis revealed that ISG15 is involved in abnormalities in various immune cells and inflammation-related pathways. We also predicted three drugs targeting ISG15, supported by molecular docking studies. Conclusion Our study identifies ISG15 as a highly specific diagnostic biomarker for DM, ISG15 may be closely related to the pathogenesis of DM, demonstrating promising potential for clinical application.
Collapse
Affiliation(s)
- Xingwang Wang
- Department of Dermatology, General Hospital of Southern Theater Command, Guangzhou, China
| | - Hao Hu
- Department of Radiation Therapy, General Hospital of Southern Theater Command, Guangzhou, China
| | - Guangning Yan
- Department of Pathology, General Hospital of Southern Theater Command, Guangzhou, China
| | - Bo Zheng
- Department of Dermatology, General Hospital of Southern Theater Command, Guangzhou, China
| | - Jinxia Luo
- Department of Pathology, General Hospital of Southern Theater Command, Guangzhou, China
| | - Jianyong Fan
- Department of Dermatology, General Hospital of Southern Theater Command, Guangzhou, China
| |
Collapse
|
17
|
Wang B, Liu W, Zhang M, Li Y, Tang H, Wang Y, Song C, Song B, Tan B. Circ_0001947 encapsulated by small extracellular vesicles promotes gastric cancer progression and anti-PD-1 resistance by modulating CD8 + T cell exhaustion. J Nanobiotechnology 2024; 22:563. [PMID: 39272146 PMCID: PMC11401313 DOI: 10.1186/s12951-024-02826-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 08/31/2024] [Indexed: 09/15/2024] Open
Abstract
BACKGROUND While small extracellular vesicles (sEVs)-derived circular RNAs (circRNAs) have been emerged as significant players in cancer, the function and underlying mechanism of sEVs-derived circRNAs in anti-cancer immunity remain unclear. METHODS Gastric cancer (GC)-derived circRNAs were identified using RNA-seq data from GEO datasets and quantitative reverse transcription polymerase chain reaction (qRT-PCR), RNA immunoprecipitation, dual-luciferase assay, and bioinformatics analysis were performed to investigate the regulatory axis. Transwell assay, wound healing assay, cell counting kit-8 (CCK-8) assay, and xenograft models were used to evaluate its role in GC progression in vivo and in vitro. The delivery of specific circRNAs into sEVs were verified through electron microscopy, nanoparticle tracking analysis (NTA) and fuorescence in situ hybridization (FISH). Flow cytometric analysis and immunohistochemical staining were conducted to find out how specific circRNAs mediated CD8+ T cell exhaustion and resistant to anti-programmed cell death 1 (PD-1) therapy. RESULTS We identified that circ_0001947, packaged by GC-derived sEVs, was obviously elevated in GC and was associated with poor clinical outcome. High circ0001947 level augmented the proliferation, migration, and invasion of GC cells. Mechanistically, circ0001947 sponged miR-661 and miR-671-5p to promote the expression of CD39, which further facilitated CD8+ T cell exhaustion and immune resistance. Conversely, blocking circ_0001947 attenuated CD8+ T cell exhaustion and increased the response to anti-PD-1 therapy. CONCLUSIONS Our study manifested the therapeutic potential of targeting sEVs-transmitted circ_0001947 to prohibit CD8+ T cell exhaustion and immune resistance in GC.
Collapse
Affiliation(s)
- Bingyu Wang
- The Third Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, China
| | - Wenbo Liu
- The Third Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, China
| | - Mingming Zhang
- Hebei Key Laboratory of Metabolic Disease, Shijiazhuang, 050011, China
| | - Yong Li
- The Third Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, China
| | - Hongyue Tang
- Hebei Key Laboratory of Metabolic Disease, Shijiazhuang, 050011, China
| | - Yingying Wang
- The Third Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, China
| | - Chao Song
- The Third Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, China
| | - Buyun Song
- The Third Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, China
| | - Bibo Tan
- The Third Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, China.
| |
Collapse
|
18
|
Xie S, Sun M, Zhang X, Kan C, Shi G, Peng W, Guo J, Wu D, Yin Z, Yang Q, Zhang R. T cell responses in immune-mediated IgA nephropathy. J Leukoc Biol 2024; 116:523-535. [PMID: 38713107 DOI: 10.1093/jleuko/qiae103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 04/08/2024] [Accepted: 04/11/2024] [Indexed: 05/08/2024] Open
Abstract
Immunoglobulin A nephropathy is a complex autoimmune disease with various underlying causes and significant clinical heterogeneity. There are large individual differences in its development, and the etiology and pathogenesis are still poorly understood. While it is known that immunobiological factors play a significant role in the pathophysiology of immunoglobulin A nephropathy, the specific nature of these factors has yet to be fully elucidated. Numerous investigations have verified that CD4+ and CD8+ T lymphocytes are involved in the immunopathogenesis of immunoglobulin A nephropathy. Furthermore, certain data also point to γδT cells' involvement in the pathophysiology of immunoglobulin A nephropathy. By thoroughly examining the mechanisms of action of these T cells in the context of immunoglobulin A nephropathy, this review sheds light on the immunopathogenesis of the disease and its associated factors. The review is intended to provide reference value for the future research in this field and promising treatment clues for clinical patients.
Collapse
Affiliation(s)
- Shimin Xie
- Department of Nephrology, Zhuhai People's Hospital, Zhuhai Clinical Medical College of Jinan University, Kangning Road, Xiangzhou District, Zhuhai, Guangdong, 519000, China
| | - Mengying Sun
- Department of Nephrology, Zhuhai People's Hospital, Zhuhai Clinical Medical College of Jinan University, Kangning Road, Xiangzhou District, Zhuhai, Guangdong, 519000, China
| | - Xiaohan Zhang
- Department of Nephrology, Zhuhai People's Hospital, Zhuhai Clinical Medical College of Jinan University, Kangning Road, Xiangzhou District, Zhuhai, Guangdong, 519000, China
| | - Chao Kan
- Department of Nephrology, Zhuhai People's Hospital, Zhuhai Clinical Medical College of Jinan University, Kangning Road, Xiangzhou District, Zhuhai, Guangdong, 519000, China
| | - Guojuan Shi
- Department of Nephrology, Zhuhai People's Hospital, Zhuhai Clinical Medical College of Jinan University, Kangning Road, Xiangzhou District, Zhuhai, Guangdong, 519000, China
| | - Weixiang Peng
- Department of Nephrology, Zhuhai People's Hospital, Zhuhai Clinical Medical College of Jinan University, Kangning Road, Xiangzhou District, Zhuhai, Guangdong, 519000, China
| | - Junli Guo
- Department of Nephrology, Zhuhai People's Hospital, Zhuhai Clinical Medical College of Jinan University, Kangning Road, Xiangzhou District, Zhuhai, Guangdong, 519000, China
| | - Dantong Wu
- Department of Nephrology, Zhuhai People's Hospital, Zhuhai Clinical Medical College of Jinan University, Kangning Road, Xiangzhou District, Zhuhai, Guangdong, 519000, China
| | - Zhinan Yin
- Biomedical Translational Research Institute, Jinan University, Huangpu Avenue, Tianhe District, Guangzhou, Guangdong, 510632, China
| | - Quanli Yang
- Department of Nephrology, Zhuhai People's Hospital, Zhuhai Clinical Medical College of Jinan University, Kangning Road, Xiangzhou District, Zhuhai, Guangdong, 519000, China
| | - Rui Zhang
- Department of Nephrology, Zhuhai People's Hospital, Zhuhai Clinical Medical College of Jinan University, Kangning Road, Xiangzhou District, Zhuhai, Guangdong, 519000, China
| |
Collapse
|
19
|
Liu X, Shen J, Yan H, Hu J, Liao G, Liu D, Zhou S, Zhang J, Liao J, Guo Z, Li Y, Yang S, Li S, Chen H, Guo Y, Li M, Fan L, Li L, Luo P, Zhao M, Liu Y. Posttransplant complications: molecular mechanisms and therapeutic interventions. MedComm (Beijing) 2024; 5:e669. [PMID: 39224537 PMCID: PMC11366828 DOI: 10.1002/mco2.669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 07/02/2024] [Accepted: 07/08/2024] [Indexed: 09/04/2024] Open
Abstract
Posttransplantation complications pose a major challenge to the long-term survival and quality of life of organ transplant recipients. These complications encompass immune-mediated complications, infectious complications, metabolic complications, and malignancies, with each type influenced by various risk factors and pathological mechanisms. The molecular mechanisms underlying posttransplantation complications involve a complex interplay of immunological, metabolic, and oncogenic processes, including innate and adaptive immune activation, immunosuppressant side effects, and viral reactivation. Here, we provide a comprehensive overview of the clinical features, risk factors, and molecular mechanisms of major posttransplantation complications. We systematically summarize the current understanding of the immunological basis of allograft rejection and graft-versus-host disease, the metabolic dysregulation associated with immunosuppressive agents, and the role of oncogenic viruses in posttransplantation malignancies. Furthermore, we discuss potential prevention and intervention strategies based on these mechanistic insights, highlighting the importance of optimizing immunosuppressive regimens, enhancing infection prophylaxis, and implementing targeted therapies. We also emphasize the need for future research to develop individualized complication control strategies under the guidance of precision medicine, ultimately improving the prognosis and quality of life of transplant recipients.
Collapse
Affiliation(s)
- Xiaoyou Liu
- Department of Organ transplantationThe First Affiliated Hospital, Guangzhou Medical UniversityGuangzhouChina
| | - Junyi Shen
- Department of OncologyZhujiang HospitalSouthern Medical UniversityGuangzhouChina
| | - Hongyan Yan
- Department of Organ transplantationThe First Affiliated Hospital, Guangzhou Medical UniversityGuangzhouChina
| | - Jianmin Hu
- Department of Organ transplantationZhujiang HospitalSouthern Medical UniversityGuangzhouChina
| | - Guorong Liao
- Department of Organ transplantationZhujiang HospitalSouthern Medical UniversityGuangzhouChina
| | - Ding Liu
- Department of Organ transplantationZhujiang HospitalSouthern Medical UniversityGuangzhouChina
| | - Song Zhou
- Department of Organ transplantationZhujiang HospitalSouthern Medical UniversityGuangzhouChina
| | - Jie Zhang
- Department of Organ transplantationThe First Affiliated Hospital, Guangzhou Medical UniversityGuangzhouChina
| | - Jun Liao
- Department of Organ transplantationZhujiang HospitalSouthern Medical UniversityGuangzhouChina
| | - Zefeng Guo
- Department of Organ transplantationZhujiang HospitalSouthern Medical UniversityGuangzhouChina
| | - Yuzhu Li
- Department of Organ transplantationZhujiang HospitalSouthern Medical UniversityGuangzhouChina
| | - Siqiang Yang
- Department of Organ transplantationZhujiang HospitalSouthern Medical UniversityGuangzhouChina
| | - Shichao Li
- Department of Organ transplantationZhujiang HospitalSouthern Medical UniversityGuangzhouChina
| | - Hua Chen
- Department of Organ transplantationZhujiang HospitalSouthern Medical UniversityGuangzhouChina
| | - Ying Guo
- Department of Organ transplantationZhujiang HospitalSouthern Medical UniversityGuangzhouChina
| | - Min Li
- Department of Organ transplantationZhujiang HospitalSouthern Medical UniversityGuangzhouChina
| | - Lipei Fan
- Department of Organ transplantationZhujiang HospitalSouthern Medical UniversityGuangzhouChina
| | - Liuyang Li
- Department of Organ transplantationZhujiang HospitalSouthern Medical UniversityGuangzhouChina
| | - Peng Luo
- Department of OncologyZhujiang HospitalSouthern Medical UniversityGuangzhouChina
| | - Ming Zhao
- Department of Organ transplantationZhujiang HospitalSouthern Medical UniversityGuangzhouChina
| | - Yongguang Liu
- Department of Organ transplantationZhujiang HospitalSouthern Medical UniversityGuangzhouChina
| |
Collapse
|
20
|
Ercilla-Rodríguez P, Sánchez-Díez M, Alegría-Aravena N, Quiroz-Troncoso J, Gavira-O'Neill CE, González-Martos R, Ramírez-Castillejo C. CAR-T lymphocyte-based cell therapies; mechanistic substantiation, applications and biosafety enhancement with suicide genes: new opportunities to melt side effects. Front Immunol 2024; 15:1333150. [PMID: 39091493 PMCID: PMC11291200 DOI: 10.3389/fimmu.2024.1333150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Accepted: 06/14/2024] [Indexed: 08/04/2024] Open
Abstract
Immunotherapy has made significant strides in cancer treatment with strategies like checkpoint blockade antibodies and adoptive T cell transfer. Chimeric antigen receptor T cells (CAR-T) have emerged as a promising approach to combine these strategies and overcome their limitations. This review explores CAR-T cells as a living drug for cancer treatment. CAR-T cells are genetically engineered immune cells designed to target and eliminate tumor cells by recognizing specific antigens. The study involves a comprehensive literature review on CAR-T cell technology, covering structure optimization, generations, manufacturing processes, and gene therapy strategies. It examines CAR-T therapy in haematologic cancers and solid tumors, highlighting challenges and proposing a suicide gene-based mechanism to enhance safety. The results show significant advancements in CAR-T technology, particularly in structure optimization and generation. The manufacturing process has improved for broader clinical application. However, a series of inherent challenges and side effects still need to be addressed. In conclusion, CAR-T cells hold great promise for cancer treatment, but ongoing research is crucial to improve efficacy and safety for oncology patients. The proposed suicide gene-based mechanism offers a potential solution to mitigate side effects including cytokine release syndrome (the most common toxic side effect of CAR-T therapy) and the associated neurotoxicity.
Collapse
MESH Headings
- Humans
- Immunotherapy, Adoptive/adverse effects
- Immunotherapy, Adoptive/methods
- Receptors, Chimeric Antigen/genetics
- Receptors, Chimeric Antigen/immunology
- Genes, Transgenic, Suicide
- Neoplasms/therapy
- Neoplasms/immunology
- Neoplasms/genetics
- T-Lymphocytes/immunology
- Animals
- Genetic Therapy/adverse effects
- Genetic Therapy/methods
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
Collapse
Affiliation(s)
| | - Marta Sánchez-Díez
- ETSIAAB, Universidad Politécnica de Madrid, Madrid, Spain
- Laboratorio Cancer Stem Cell, HST group, Centro de Tecnología Biomédica, Universidad Politécnica de Madrid, Madrid, Spain
| | - Nicolás Alegría-Aravena
- Grupo de Biología y Producción de Cérvidos, Instituto de Desarrollo Regional, Universidad de Castilla-La Mancha, Albacete, Spain
- Asociación Española Contra el Cáncer (AECC)-Fundación Científica AECC, Albacete, Spain
| | - Josefa Quiroz-Troncoso
- ETSIAAB, Universidad Politécnica de Madrid, Madrid, Spain
- Laboratorio Cancer Stem Cell, HST group, Centro de Tecnología Biomédica, Universidad Politécnica de Madrid, Madrid, Spain
| | - Clara E. Gavira-O'Neill
- Laboratorio Cancer Stem Cell, HST group, Centro de Tecnología Biomédica, Universidad Politécnica de Madrid, Madrid, Spain
- Sección de Oncología, Instituto de Investigación Sanitaria San Carlos, Madrid, Spain
| | - Raquel González-Martos
- ETSIAAB, Universidad Politécnica de Madrid, Madrid, Spain
- Laboratorio Cancer Stem Cell, HST group, Centro de Tecnología Biomédica, Universidad Politécnica de Madrid, Madrid, Spain
| | - Carmen Ramírez-Castillejo
- ETSIAAB, Universidad Politécnica de Madrid, Madrid, Spain
- Laboratorio Cancer Stem Cell, HST group, Centro de Tecnología Biomédica, Universidad Politécnica de Madrid, Madrid, Spain
- Sección de Oncología, Instituto de Investigación Sanitaria San Carlos, Madrid, Spain
| |
Collapse
|
21
|
Zhang L, Zhu L, Che P, Sun X, Guo Y, Gao M, Wang J. Cytotoxic T Lymphocytes, Tc17 Cells, Th1 Cells, and ThGM Cells are Increased in the Blood and Ectopic Endometrium of Patients With Adenomyosis. Am J Reprod Immunol 2024; 92:e13901. [PMID: 39042523 DOI: 10.1111/aji.13901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 06/16/2024] [Accepted: 06/28/2024] [Indexed: 07/25/2024] Open
Abstract
PROBLEM Adenomyosis (AM) is associated with immune response and inflammation. However, the role of T cell subsets in AM development has not been thoroughly understood. METHOD OF STUDY Patients with focal or diffuse AM were recruited. Serum cytokines were quantified by enzyme-linked immunosorbent assay (ELISA). Different T cell subsets in the blood and ectopic endometrium were determined by flow cytometry. RESULTS Serum interleukin-6 (IL-6) and macrophage-colony-stimulating factor (GM-CSF) were increased in patients with focal or diffuse AM before focused ultrasound ablation surgery (FUAS), but not after FUAS. Compared with the healthy control, the frequencies of CD8+ interferon-gamma (IFN-γ)-expressing cytotoxic T lymphocytes (CTLs), interleukin-17A (IL-17A)-expressing Tc17 cells, CD4+ T helper 1 (Th1) cells, and GM-CSF-expressing T helper (ThGM) cells were up-regulated in the blood of patients with AM, especially those with diffuse AM. However, these changes were eradicated after FUAS. Meanwhile, the frequencies of these T cell subsets were positively correlated with the CA-125 level. Furthermore, these T cell subsets were also increased in ectopic endometrium. CONCLUSIONS Our study delineates for the first time the presence of CTLs, Tc17 cells, Th1, and ThGM cells in the blood and ectopic endometrium in AM. The results imply that T cell response might impact AM development.
Collapse
Affiliation(s)
- Li Zhang
- Department of Gynecology and Obstetrics, Affiliated Renhe Hospital of China Three Gorges University, Yichang, Hubei, China
| | - Lei Zhu
- Emergency Center, Affiliated Renhe Hospital of China Three Gorges University, Yichang, Hubei, China
| | - Pengfei Che
- Department of Ultrasound, Affiliated Renhe Hospital of China Three Gorges University, Yichang, Hubei, China
| | - Xiaoyan Sun
- Department of Gynecology and Obstetrics, Xingshan County People's Hospital, Yichang, Hubei, China
| | - Yupeng Guo
- Department of Interventional Radiology, Affiliated Renhe Hospital of China Three Gorges University, Yichang, Hubei, China
| | - Mingjie Gao
- Department of Oncology, Affiliated Renhe Hospital of China Three Gorges University, Yichang, Hubei, China
| | - Junjie Wang
- Department of Gynecology and Obstetrics, Affiliated Renhe Hospital of China Three Gorges University, Yichang, Hubei, China
| |
Collapse
|
22
|
Li C, Hong W, Reuben A, Wang L, Maitra A, Zhang J, Cheng C. TimiGP-Response: the pan-cancer immune landscape associated with response to immunotherapy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.21.600089. [PMID: 38979334 PMCID: PMC11230183 DOI: 10.1101/2024.06.21.600089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Accumulating evidence suggests that the tumor immune microenvironment (TIME) significantly influences the response to immunotherapy, yet this complex relationship remains elusive. To address this issue, we developed TimiGP-Response (TIME Illustration based on Gene Pairing designed for immunotherapy Response), a computational framework leveraging single-cell and bulk transcriptomic data, along with response information, to construct cell-cell interaction networks associated with responders and estimate the role of immune cells in treatment response. This framework was showcased in triple-negative breast cancer treated with immune checkpoint inhibitors targeting the PD-1:PD-L1 interaction, and orthogonally validated with imaging mass cytometry. As a result, we identified CD8+ GZMB+ T cells associated with responders and its interaction with regulatory T cells emerged as a potential feature for selecting patients who may benefit from these therapies. Subsequently, we analyzed 3,410 patients with seven cancer types (melanoma, non-small cell lung cancer, renal cell carcinoma, metastatic urothelial carcinoma, hepatocellular carcinoma, breast cancer, and esophageal cancer) treated with various immunotherapies and combination therapies, as well as several chemo- and targeted therapies as controls. Using TimiGP-Response, we depicted the pan-cancer immune landscape associated with immunotherapy response at different resolutions. At the TIME level, CD8 T cells and CD4 memory T cells were associated with responders, while anti-inflammatory (M2) macrophages and mast cells were linked to non-responders across most cancer types and datasets. Given that T cells are the primary targets of these immunotherapies and our TIME analysis highlights their importance in response to treatment, we portrayed the pan-caner landscape on 40 T cell subtypes. Notably, CD8+ and CD4+ GZMK+ effector memory T cells emerged as crucial across all cancer types and treatments, while IL-17-producing CD8+ T cells were top candidates associated with immunotherapy non-responders. In summary, this study provides a computational method to study the association between TIME and response across the pan-cancer immune landscape, offering resources and insights into immune cell interactions and their impact on treatment efficacy.
Collapse
Affiliation(s)
- Chenyang Li
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center UTHealth Houston, Houston, TX 77030, USA
| | - Wei Hong
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Alexandre Reuben
- Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center UTHealth Houston, Houston, TX 77030, USA
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Linghua Wang
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center UTHealth Houston, Houston, TX 77030, USA
| | - Anirban Maitra
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Sheikh Ahmed Center for Pancreatic Cancer Research, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jianjun Zhang
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center UTHealth Houston, Houston, TX 77030, USA
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Lung Cancer Genomics Program, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Lung Cancer Interception Program, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Chao Cheng
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
- The Institute for Clinical and Translational Research, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
23
|
Sinkarevs S, Strumfs B, Volkova S, Strumfa I. Tumour Microenvironment: The General Principles of Pathogenesis and Implications in Diffuse Large B Cell Lymphoma. Cells 2024; 13:1057. [PMID: 38920685 PMCID: PMC11201569 DOI: 10.3390/cells13121057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 06/10/2024] [Accepted: 06/12/2024] [Indexed: 06/27/2024] Open
Abstract
Diffuse large B cell lymphoma (DLBCL) is the most common type of non-Hodgkin lymphoma worldwide, constituting around 30-40% of all cases. Almost 60% of patients develop relapse of refractory DLBCL. Among the reasons for the therapy failure, tumour microenvironment (TME) components could be involved, including tumour-associated macrophages (TAMs), myeloid-derived suppressor cells (MDSCs), tumour-associated neutrophils (TANs), cancer-associated fibroblasts (CAFs), and different subtypes of cytotoxic CD8+ cells and T regulatory cells, which show complex interactions with tumour cells. Understanding of the TME can provide new therapeutic options for patients with DLBCL and improve their prognosis and overall survival. This review provides essentials of the latest understanding of tumour microenvironment elements and discusses their role in tumour progression and immune suppression mechanisms which result in poor prognosis for patients with DLBCL. In addition, we point out important markers for the diagnostic purposes and highlight novel therapeutic targets.
Collapse
Affiliation(s)
| | | | | | - Ilze Strumfa
- Department of Pathology, Riga Stradins University, 16 Dzirciema Street, LV-1007 Riga, Latvia
| |
Collapse
|
24
|
Yang Y, Bai Q, Liu F, Zhang S, Tang W, Liu L, Xing Z, Wang H, Zhang C, Yang Y, Fan H. Establishment of the Diagnostic Signature of Ferroptosis Genes in Multiple Sclerosis. Biochem Genet 2024:10.1007/s10528-024-10832-3. [PMID: 38886317 DOI: 10.1007/s10528-024-10832-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 05/08/2024] [Indexed: 06/20/2024]
Abstract
Ferroptosis is a novel form of membrane-dependent cell death that differs from other cell death modalities such as necrosis, apoptosis, and autophagy. Multiple sclerosis (MS) is a chronic inflammatory disease of the central nervous system primarily affecting brain and spinal cord neurons. Although the pathogenesis of these two conditions may seem unrelated, recent studies have indicated a connection between ferroptosis and multiple sclerosis. In fact, ferroptosis plays a significant role in the development of MS, as evidenced by the presence of elevated iron levels and iron metabolism abnormalities in the brains, spinal cords, and other neurons of MS patients. These abnormalities disrupt iron homeostasis within cells, leading to the occurrence of ferroptosis. However, there is currently a lack of research on the diagnostic value of ferroptosis-related genes in multiple sclerosis. In this study, we employed bioinformatics methods to identify ferroptosis-related genes (ATM, GSK3B, HMGCR, KLF2, MAPK1, NFE2L1, NRAS, PCBP1, PIK3CA, RPL8, VDAC3) associated with the diagnosis of multiple sclerosis and constructed a diagnostic model. The results demonstrated that the diagnostic model accurately identified the patients' condition. Subsequently, subgroup analysis was performed based on the expression levels of ferroptosis-related genes, dividing patients into high and low expression groups. The results showed differences in immune function and immune cell infiltration between the two groups. Our study not only confirms the correlation between ferroptosis and multiple sclerosis but also demonstrates the diagnostic value of ferroptosis-related genes in the disease. This provides guidance for clinical practice and direction for further mechanistic research.
Collapse
Affiliation(s)
- Yang Yang
- Office of Research & Innovation, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, 471003, China
| | - Qianqian Bai
- Office of Research & Innovation, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, 471003, China
| | - Fangfei Liu
- Office of Research & Innovation, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, 471003, China
| | - Shumin Zhang
- Office of Research & Innovation, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, 471003, China
| | - Wenchao Tang
- Office of Research & Innovation, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, 471003, China
| | - Ling Liu
- Office of Research & Innovation, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, 471003, China
| | - Zhehua Xing
- Department of Trauma Center, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, 471003, China
| | - Hao Wang
- Department of Trauma Center, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, 471003, China
| | - Chi Zhang
- Department of Trauma Center, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, 471003, China
| | - Yanhui Yang
- Department of Trauma Center, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, 471003, China.
| | - Hua Fan
- Office of Research & Innovation, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, 471003, China.
| |
Collapse
|
25
|
Yan C, Du W, Kirkwood KL, Wang Y, Zhou W, Li Z, Tian Y, Lin S, Zheng L, Al-Aroomi MA, Gao J, Jiang S, Sun C, Liu F. CCR7 affects the tumor microenvironment by regulating the activation of naïve CD8 + T cells to promote the proliferation of oral squamous cell carcinoma. Transl Oncol 2024; 44:101924. [PMID: 38430712 PMCID: PMC10920962 DOI: 10.1016/j.tranon.2024.101924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 02/18/2024] [Accepted: 02/25/2024] [Indexed: 03/05/2024] Open
Abstract
BACKGROUND Head and neck cancer is the sixth most common malignancy worldwide, and oral squamous cell carcinoma (OSCC) is the most common head and neck cancer, being one of the leading causes of cancer morbidity and mortality worldwide. CC Chemokine receptor 7(CCR7) is a multifunctional G protein-coupled trans-membrane chemokine that affects immune cell chemotaxis, migration, and cancer progression through its interaction with its ligands C-C motif chemokine ligand 19(CCL19) and C-C motif chemokine ligand 21(CCL21). Numerous studies have demonstrated the involvement of CCR7 in the malignant progression of a variety of cancers, reflecting the pro-cancer properties of CCR7. The Cancer Genome Atlas data suggests CCR7 has elevated expression in oral cancer. Specifically, CCR7 expression in tumor microenvironment (TME) may regulate the ability of some immune cells to engage in anti-tumor immune responses. Since CD8+ T cells have become a key immunotherapeutic target, the role of CCR7 in antitumor immune response of naïve CD8+ T cells in TME has not been thoroughly investigated. METHODS A CCR7 knockout mouse model was constructed, and the mechanism of ccr7 on the regulation of tumor microenvironment by naïve CD8+ T cells was verified under the guidance of single-cell RNA sequencing combined with in vivo animal experiments and in vitro cell experiments. RESULTS CCR7 is knocked out with impaired tumor growth and altered CD8+ T cell profiles, revealing the importance of this protein in OSCC. CONCLUSIONS Inhibition of CCR7 enhances CD8+ T cell activation, proliferation, and anti-tumor function, suggesting its potential as a therapeutic target.
Collapse
Affiliation(s)
- Cong Yan
- Department of Oral Maxillofacial-Head and Neck Surgery, School of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, 117 Nanjing North Road, Heping District, Shenyang, Liaoning 110000, PR China
| | - Weidong Du
- Department of Oral Maxillofacial-Head and Neck Surgery, School of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, 117 Nanjing North Road, Heping District, Shenyang, Liaoning 110000, PR China
| | - Keith L Kirkwood
- Department of Oral Biology, School of Dental Medicine, University at Buffalo, Buffalo, NY 14214-8006, USA
| | - Yao Wang
- Department of Oral Maxillofacial-Head and Neck Surgery, School of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, 117 Nanjing North Road, Heping District, Shenyang, Liaoning 110000, PR China
| | - Wanhang Zhou
- Department of Oral Maxillofacial-Head and Neck Surgery, School of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, 117 Nanjing North Road, Heping District, Shenyang, Liaoning 110000, PR China
| | - Zhenning Li
- Department of Oral Maxillofacial-Head and Neck Surgery, School of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, 117 Nanjing North Road, Heping District, Shenyang, Liaoning 110000, PR China
| | - Yuan Tian
- Department of Oral Maxillofacial-Head and Neck Surgery, School of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, 117 Nanjing North Road, Heping District, Shenyang, Liaoning 110000, PR China
| | - Shanfeng Lin
- Department of Oral Maxillofacial-Head and Neck Surgery, School of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, 117 Nanjing North Road, Heping District, Shenyang, Liaoning 110000, PR China
| | - Li Zheng
- Department of Oral Maxillofacial-Head and Neck Surgery, School of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, 117 Nanjing North Road, Heping District, Shenyang, Liaoning 110000, PR China
| | - Maged Ali Al-Aroomi
- Department of Oral Maxillofacial-Head and Neck Surgery, School of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, 117 Nanjing North Road, Heping District, Shenyang, Liaoning 110000, PR China
| | - Jiaxing Gao
- Department of Oral Maxillofacial-Head and Neck Surgery, School of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, 117 Nanjing North Road, Heping District, Shenyang, Liaoning 110000, PR China
| | - Sheng Jiang
- Department of Oral Maxillofacial-Head and Neck Surgery, School of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, 117 Nanjing North Road, Heping District, Shenyang, Liaoning 110000, PR China
| | - Changfu Sun
- Department of Oral Maxillofacial-Head and Neck Surgery, School of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, 117 Nanjing North Road, Heping District, Shenyang, Liaoning 110000, PR China
| | - Fayu Liu
- Department of Oral Maxillofacial-Head and Neck Surgery, School of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, 117 Nanjing North Road, Heping District, Shenyang, Liaoning 110000, PR China.
| |
Collapse
|
26
|
Fang Y, Li X, Tian R. Unlocking Glioblastoma Vulnerabilities with CRISPR-Based Genetic Screening. Int J Mol Sci 2024; 25:5702. [PMID: 38891890 PMCID: PMC11171782 DOI: 10.3390/ijms25115702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 05/17/2024] [Accepted: 05/22/2024] [Indexed: 06/21/2024] Open
Abstract
Glioblastoma (GBM) is the most common malignant brain tumor in adults. Despite advancements in treatment, the prognosis for patients with GBM remains poor due to its aggressive nature and resistance to therapy. CRISPR-based genetic screening has emerged as a powerful tool for identifying genes crucial for tumor progression and treatment resistance, offering promising targets for tumor therapy. In this review, we provide an overview of the recent advancements in CRISPR-based genetic screening approaches and their applications in GBM. We highlight how these approaches have been used to uncover the genetic determinants of GBM progression and responsiveness to various therapies. Furthermore, we discuss the ongoing challenges and future directions of CRISPR-based screening methods in advancing GBM research.
Collapse
Affiliation(s)
- Yitong Fang
- Department of Medical Neuroscience, School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China; (Y.F.); (X.L.)
- Key University Laboratory of Metabolism and Health of Guangdong, Southern University of Science and Technology, Shenzhen 518055, China
| | - Xing Li
- Department of Medical Neuroscience, School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China; (Y.F.); (X.L.)
- Key University Laboratory of Metabolism and Health of Guangdong, Southern University of Science and Technology, Shenzhen 518055, China
| | - Ruilin Tian
- Department of Medical Neuroscience, School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China; (Y.F.); (X.L.)
- Key University Laboratory of Metabolism and Health of Guangdong, Southern University of Science and Technology, Shenzhen 518055, China
| |
Collapse
|
27
|
Lee AR, Wilson KR, Clarke M, Engel S, Tscharke DC, Gebhardt T, Bedoui S, Bachem A. GPR41 and GPR43 regulate CD8 + T cell priming during herpes simplex virus type 1 infection. Front Immunol 2024; 15:1332588. [PMID: 38524121 PMCID: PMC10957577 DOI: 10.3389/fimmu.2024.1332588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 02/13/2024] [Indexed: 03/26/2024] Open
Abstract
Naïve CD8+ T cells need to undergo a complex and coordinated differentiation program to gain the capacity to control virus infections. This not only involves the acquisition of effector functions, but also regulates the development of a subset of effector CD8+ T cells into long-lived and protective memory cells. Microbiota-derived metabolites have recently gained interest for their influence on T cells, but much remains unclear about their role in CD8+ T cell differentiation. In this study, we investigated the role of the G protein-coupled receptors (GPR)41 and GPR43 that can bind microbiota-derived short chain fatty acids (SCFAs) in CD8+ T cell priming following epicutaneous herpes simplex virus type 1 (HSV-1) infection. We found that HSV-specific CD8+ T cells in GPR41/43-deficient mice were impaired in the antigen-elicited production of interferon-gamma (IFN-γ), tumour necrosis factor-alpha (TNF-α), granzyme B and perforin, and failed to differentiate effectively into memory precursors. The defect in controlling HSV-1 at the site of infection could be restored when GPR41 and GPR43 were expressed exclusively by HSV-specific CD8+ T cells. Our findings therefore highlight roles for GPR41 and GPR43 in CD8+ T cell differentiation, emphasising the importance of metabolite sensing in fine-tuning anti-viral CD8+ T cell priming.
Collapse
Affiliation(s)
- Ariane Renita Lee
- Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, VIC, Australia
| | - Kayla Roberta Wilson
- Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, VIC, Australia
| | - Michele Clarke
- Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, VIC, Australia
| | - Sven Engel
- Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, VIC, Australia
| | - David C. Tscharke
- John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
| | - Thomas Gebhardt
- Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, VIC, Australia
| | - Sammy Bedoui
- Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, VIC, Australia
| | - Annabell Bachem
- Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
28
|
Peng Y, Zhang X, Tang Y, He S, Rao G, Chen Q, Xue Y, Jin H, Liu S, Zhou Z, Xiang Y. Role of autoreactive Tc17 cells in the pathogenesis of experimental autoimmune encephalomyelitis. NEUROPROTECTION 2024; 2:49-59. [DOI: 10.1002/nep3.38] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 01/16/2024] [Indexed: 07/04/2024]
Abstract
AbstractBackgroundThe pathogenesis of multiple sclerosis (MS) and experimental autoimmune encephalomyelitis (EAE—an animal model of MS) is primarily mediated by T cells. However, recent studies have only focused on interleukin (IL)‐17‐secreting CD4+ T‐helper cells, also known as Th17 cells. This study aimed to compare Th17 cells and IL‐17‐secreting CD8+ T‐cytotoxic cells (Tc17) in the context of MS/EAE.MethodsFemale C57BL/6 mice were immunized with myelin oligodendrocyte glycoprotein peptides 35–55 (MOG35–55), pertussis toxin, and complete Freund's adjuvant to establish the EAE animal model. T cells were isolated from the spleen (12–14 days postimmunization). CD4+ and CD8+ T cells were purified using isolation kit and then differentiated into Th17 and Tc17, respectively, using MOG35–55 and IL‐23. The secretion levels of interferon‐γ (IFN‐γ) and IL‐17 were measured via enzyme‐linked immunosorbent assay using cultured CD4+ and CD8+ T cell supernatants. The pathogenicity of Tc17 and Th17 cells was assessed through adoptive transfer (tEAE), with the clinical course assessed using an EAE score (0–5). Hematoxylin and eosin as well as Luxol fast blue staining were used to examine the spinal cord. Purified CD8+ CD3+ and CD4+ CD3+ cells differentiated into Tc17 and Th17 cells, respectively, were stimulated with MOG35–55 peptide for proliferation assays.ResultsThe results showed that Tc17 cells (15,951 ± 1985 vs. 55,709 ± 4196 cpm; p < 0.050) exhibited a weaker response to highest dose (20 μg/mL) MOG35–55 than Th17 cells. However, this response was not dependent on Th17 cells. After the 48 h stimulation, at the highest dose (20 μg/mL) of MOG35–55. Tc17 cells secreted lower levels of IFN‐γ (280.00 ± 15.00 vs. 556.67 ± 15.28 pg/mL, p < 0.050) and IL‐17 (102.67 ± 5.86 pg/mL vs. 288.33 ± 12.58 pg/mL; p < 0.050) than Th17 cells. Similar patterns were observed for IFN‐γ secretion at 96 and 144 h. Furthermore, Tc17 cell‐induced tEAE mice exhibited similar EAE scores to Th17 cell‐induced tEAE mice and also showed similar inflammation and demyelination.ConclusionThe degree of pathogenicity of Tc17 cells in EAE is lower than that of Th17 cells. Future investigation on different immune cells and EAE models is warranted to determine the mechanisms underlying MS.
Collapse
Affiliation(s)
- Yong Peng
- Department of Neurology Affiliated First Hospital of Hunan Traditional Chinese Medical College Zhuzhou Hunan China
- Department of Neurology The Third Affiliated Hospital of Hunan University of Chinese Medicine Zhuzhou Hunan China
| | - Xiuli Zhang
- Science and Technology Innovation Center Hunan University of Chinese Medicine Changsha Hunan China
| | - Yandan Tang
- Department of Neurology Affiliated First Hospital of Hunan Traditional Chinese Medical College Zhuzhou Hunan China
- Department of Neurology The Third Affiliated Hospital of Hunan University of Chinese Medicine Zhuzhou Hunan China
| | - Shunqing He
- Department of Neurology Affiliated First Hospital of Hunan Traditional Chinese Medical College Zhuzhou Hunan China
- Department of Neurology The Third Affiliated Hospital of Hunan University of Chinese Medicine Zhuzhou Hunan China
| | - Guilan Rao
- Department of Neurology Affiliated First Hospital of Hunan Traditional Chinese Medical College Zhuzhou Hunan China
- Department of Neurology The Third Affiliated Hospital of Hunan University of Chinese Medicine Zhuzhou Hunan China
| | - Quan Chen
- Department of Neurology Affiliated First Hospital of Hunan Traditional Chinese Medical College Zhuzhou Hunan China
- Department of Neurology The Third Affiliated Hospital of Hunan University of Chinese Medicine Zhuzhou Hunan China
| | - Yahui Xue
- Department of Neurology Affiliated First Hospital of Hunan Traditional Chinese Medical College Zhuzhou Hunan China
- Department of Neurology The Third Affiliated Hospital of Hunan University of Chinese Medicine Zhuzhou Hunan China
| | - Hong Jin
- Department of Neurology Affiliated First Hospital of Hunan Traditional Chinese Medical College Zhuzhou Hunan China
- Department of Neurology The Third Affiliated Hospital of Hunan University of Chinese Medicine Zhuzhou Hunan China
| | - Shu Liu
- Department of Neurology Affiliated First Hospital of Hunan Traditional Chinese Medical College Zhuzhou Hunan China
- Department of Neurology The Third Affiliated Hospital of Hunan University of Chinese Medicine Zhuzhou Hunan China
| | - Ziyang Zhou
- Science and Technology Innovation Center Hunan University of Chinese Medicine Changsha Hunan China
| | - Yun Xiang
- Science and Technology Innovation Center Hunan University of Chinese Medicine Changsha Hunan China
| |
Collapse
|
29
|
Chu L, Wang C, Zhou H. Inflammation mechanism and anti-inflammatory therapy of dry eye. Front Med (Lausanne) 2024; 11:1307682. [PMID: 38420354 PMCID: PMC10899709 DOI: 10.3389/fmed.2024.1307682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 01/14/2024] [Indexed: 03/02/2024] Open
Abstract
Dry eye is a widespread chronic inflammatory disease that causes fatigue, tingling, burning, and other symptoms. Dry eye is attributed to rheumatic diseases, diabetes, hormone disorders, and contact lenses, which activate inflammatory pathways: mitogen-activated protein kinases (MAPK) and nuclear factor-B (NF-κB), promote macrophage inflammatory cell and T cell activation, and inflammation factors. Clinicians use a combination of anti-inflammatory drugs to manage different symptoms of dry eye; some of these anti-inflammatory drugs are being developed. This review introduces the dry eye inflammation mechanisms and the involved inflammatory factors. We also elucidate the anti-inflammatory drug mechanism and the detection limits.
Collapse
Affiliation(s)
- Liyuan Chu
- Department of Ophthalmology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Caiming Wang
- Department of Ophthalmology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Hongyan Zhou
- Department of Ophthalmology, China-Japan Union Hospital of Jilin University, Changchun, China
| |
Collapse
|
30
|
Saad HA, Baz A, Riad M, Eraky ME, El-Taher A, Farid MI, Sharaf K, Said HEM, Ibrahim LA. Tumor microenvironment and immune system preservation in early-stage breast cancer: routes for early recurrence after mastectomy and treatment for lobular and ductal forms of disease. BMC Immunol 2024; 25:9. [PMID: 38273260 PMCID: PMC10809557 DOI: 10.1186/s12865-023-00591-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 12/07/2023] [Indexed: 01/27/2024] Open
Abstract
BACKGROUND Intra-ductal cancer (IDC) is the most common type of breast cancer, with intra-lobular cancer (ILC) coming in second. Surgery is the primary treatment for early stage breast cancer. There are now irrefutable data demonstrating that the immune context of breast tumors can influence growth and metastasis. Adjuvant chemotherapy may be administered in patients who are at a high risk of recurrence. Our goal was to identify the processes underlying both types of early local recurrences. METHODS This was a case-control observational study. Within 2 years of receiving adjuvant taxan and anthracycline-based chemotherapy, as well as modified radical mastectomy (MRM), early stage IDC and ILC recurred. Vimentin, α-smooth muscle actin (SMA), platelet-derived growth factor (PDGF), matrix metalloproteinase (MMP1), and clustered differentiation (CD95) were investigated. RESULTS Of the samples in the ductal type group, 25 showed local recurrence, and 25 did not. Six individuals in the lobular-type group did not experience recurrence, whereas seven did. Vimentin (p = 0.000 and 0.021), PDGF (p = 0.000 and 0.002), and CD95 (p = 0.000 and 0.045) expressions were significantly different in ductal and lobular carcinoma types, respectively. Measurement of ductal type was the sole significant difference found in MMP1 (p = 0.000) and α-SMA (p = 0.000). α-SMA and CD95 were two variables that helped the recurrence mechanism in the ductal type according to the pathway analysis. In contrast, the CD95 route is a recurrent mechanism for the lobular form. CONCLUSIONS While the immune system plays a larger role in ILC, the tumor microenvironment and immune system both influence the recurrence of IDC. According to this study, improving the immune system may be a viable cancer treatment option.
Collapse
Affiliation(s)
- Hassan A Saad
- Surgical Department, Faculty of Medicine, Zagazig University, Zagazig City, 44661, Egypt.
| | - Azza Baz
- Surgical Department, Alahrar Teaching Hospital, Zagazig University, Zagazig City, 55971, Egypt
| | - Mohamed Riad
- Surgical Department, Faculty of Medicine, Zagazig University, Zagazig City, 44661, Egypt
| | - Mohamed E Eraky
- Surgical Department, Faculty of Medicine, Zagazig University, Zagazig City, 44661, Egypt
| | - Ahmed El-Taher
- Surgical Department, Faculty of Medicine, Zagazig University, Zagazig City, 44661, Egypt
| | - Mohamed I Farid
- Surgical Department, Faculty of Medicine, Zagazig University, Zagazig City, 44661, Egypt
| | - Khaled Sharaf
- Surgical Department, Faculty of Medicine, Zagazig University, Zagazig City, 44661, Egypt
| | - Huda E M Said
- Clinical Pathology Department, Faculty of Medicine, Zagazig University, Zagazig City, 55971, Egypt
| | - Lotfy A Ibrahim
- Surgical Department, AlAzhar University, Nasr City, Cairo, 55888, Egypt
| |
Collapse
|
31
|
Shen W, Wang C, Jiang J, He Y, Liang Q, Hu K. Targeted delivery of herpes simplex virus glycoprotein D to CD169 + macrophages using ganglioside liposomes alleviates herpes simplex keratitis in mice. J Control Release 2024; 365:208-218. [PMID: 37981051 DOI: 10.1016/j.jconrel.2023.11.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 10/12/2023] [Accepted: 11/13/2023] [Indexed: 11/21/2023]
Abstract
Herpes simplex keratitis (HSK) is a common blinding corneal disease caused by herpes simplex virus type 1 (HSV-1) infection. Antiviral drugs and corticosteroids haven't shown adequate therapeutic efficacy. During the early stage of HSV-1 infection, macrophages serve as the first line of defense. In particular, CD169+ macrophages play an important role in phagocytosis and antigen presentation. Therefore, we constructed GM-gD-lip, a ganglioside GM1 liposome vaccine encapsulating HSV-1 glycoprotein D and targeting CD169+ macrophages. After subconjunctival injection of the vaccine, we evaluated the survival rate and ocular surface lesions of the HSK mice, as well as the virus levels in the tear fluid, corneas, and trigeminal ganglia. We discovered that GM-gD-lip reduced HSV-1 viral load and alleviated the clinical severity of HSK. The GM-gD-lip also increased the number of corneal infiltrating macrophages, especially CD169+ macrophages, and polarized them toward M1. Furthermore, the number of dendritic cells (DCs) and CD8+ T cells in the ocular draining lymph nodes was significantly increased. These findings demonstrated that GM-gD-lip polarized CD169+ macrophages toward M1 to eliminate the virus while cross-presenting antigens to CD8+ T cells via DCs to activate adaptive immunity, ultimately attenuating the severity of HSK. The use of GM-gD-lip as an immunotherapeutic method for the treatment of HSK has significant implications.
Collapse
Affiliation(s)
- Wenhao Shen
- Department of Ophthalmology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Rd,Nanjing, Jiangsu, China.
| | - Chenchen Wang
- The Eye Hospital of Wenzhou Medical University, Wenzhou Medical University, 618 Fengqi East Rd, Hangzhou, Zhejiang, China.
| | - Jiaxuan Jiang
- Department of Ophthalmology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Rd,Nanjing, Jiangsu, China.
| | - Yun He
- Department of Ophthalmology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Rd,Nanjing, Jiangsu, China.
| | - Qi Liang
- Department of Ophthalmology, Sir Run Run Shaw Hospital, Zhejiang University School of medicine, 3 Qingchun East Road, Hangzhou, Zhejiang, China.
| | - Kai Hu
- Department of Ophthalmology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Rd,Nanjing, Jiangsu, China.
| |
Collapse
|
32
|
Neyens D, Hirsch T, Abdel Aziz Issa Abdel Hadi A, Dauguet N, Vanhaver C, Bayard A, Wildmann C, Luyckx M, Squifflet JL, D’Hondt Q, Duhamel C, Huaux A, Montiel V, Dechamps M, van der Bruggen P. HELIOS-expressing human CD8 T cells exhibit limited effector functions. Front Immunol 2023; 14:1308539. [PMID: 38187391 PMCID: PMC10770868 DOI: 10.3389/fimmu.2023.1308539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 11/15/2023] [Indexed: 01/09/2024] Open
Abstract
Introduction The transcription factor HELIOS is primarily known for its expression in CD4 regulatory T cells, both in humans and mice. In mice, HELIOS is found in exhausted CD8 T cells. However, information on human HELIOS+ CD8 T cells is limited and conflicting. Methods In this study, we characterized by flow cytometry and transcriptomic analyses human HELIOS+ CD8 T cells. Results These T cells primarily consist of memory cells and constitute approximately 21% of blood CD8 T cells. In comparison with memory HELIOS- T-BEThigh CD8 T cells that displayed robust effector functions, the memory HELIOS+ T-BEThigh CD8 T cells produce lower amounts of IFN-γ and TNF-α and have a lower cytotoxic potential. We wondered if these cells participate in the immune response against viral antigens, but did not find HELIOS+ cells among CD8 T cells recognizing CMV peptides presented by HLA-A2 and HLA-B7. However, we found HELIOS+ CD8 T cells that recognize a CMV peptide presented by MHC class Ib molecule HLA-E. Additionally, a portion of HELIOS+ CD8 T cells is characterized by the expression of CD161, often used as a surface marker for identifying TC17 cells. These CD8 T cells express TH17/TC17-related genes encoding RORgt, RORa, PLZF, and CCL20. Discussion Our findings emphasize that HELIOS is expressed across various CD8 T cell populations, highlighting its significance beyond its role as a transcription factor for Treg or exhausted murine CD8 T cells. The significance of the connection between HELIOS and HLA-E restriction is yet to be understood.
Collapse
Affiliation(s)
- Damien Neyens
- De Duve Institute, Université Catholique de Louvain, Brussels, Belgium
| | - Thibault Hirsch
- De Duve Institute, Université Catholique de Louvain, Brussels, Belgium
| | | | - Nicolas Dauguet
- De Duve Institute, Université Catholique de Louvain, Brussels, Belgium
| | | | - Alexandre Bayard
- De Duve Institute, Université Catholique de Louvain, Brussels, Belgium
| | - Claude Wildmann
- De Duve Institute, Université Catholique de Louvain, Brussels, Belgium
| | - Mathieu Luyckx
- De Duve Institute, Université Catholique de Louvain, Brussels, Belgium
- Département de gynécologie, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Jean-Luc Squifflet
- De Duve Institute, Université Catholique de Louvain, Brussels, Belgium
- Département de gynécologie, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Quentin D’Hondt
- De Duve Institute, Université Catholique de Louvain, Brussels, Belgium
| | - Céline Duhamel
- De Duve Institute, Université Catholique de Louvain, Brussels, Belgium
| | - Antoine Huaux
- De Duve Institute, Université Catholique de Louvain, Brussels, Belgium
| | - Virginie Montiel
- Unité de soins intensifs, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Mélanie Dechamps
- Unité de soins intensifs, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Pierre van der Bruggen
- De Duve Institute, Université Catholique de Louvain, Brussels, Belgium
- Walloon Excellence in Life Sciences and Biotechnology (WELBIO), Wavre, Belgium
| |
Collapse
|
33
|
Blanc-Durand F, Clemence Wei Xian L, Tan DSP. Targeting the immune microenvironment for ovarian cancer therapy. Front Immunol 2023; 14:1328651. [PMID: 38164130 PMCID: PMC10757966 DOI: 10.3389/fimmu.2023.1328651] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 12/05/2023] [Indexed: 01/03/2024] Open
Abstract
Ovarian cancer (OC) is an aggressive malignancy characterized by a complex immunosuppressive tumor microenvironment (TME). Immune checkpoint inhibitors have emerged as a breakthrough in cancer therapy by reactivating the antitumor immune response suppressed by tumor cells. However, in the case of OC, these inhibitors have failed to demonstrate significant improvements in patient outcomes, and existing biomarkers have not yet identified promising subgroups. Consequently, there remains a pressing need to understand the interplay between OC tumor cells and their surrounding microenvironment to develop effective immunotherapeutic approaches. This review aims to provide an overview of the OC TME and explore its potential as a therapeutic strategy. Tumor-infiltrating lymphocytes (TILs) are major actors in OC TME. Evidence has been accumulating regarding the spontaneous TILS response against OC antigens. Activated T-helpers secrete a wide range of inflammatory cytokines with a supportive action on cytotoxic T-cells. Simultaneously, mature B-cells are recruited and play a significant antitumor role through opsonization of target antigens and T-cell recruitment. Macrophages also form an important subset of innate immunity (M1-macrophages) while participating in the immune-stimulation context. Finally, OC has shown to engage a significant natural-killer-cells immune response, exerting direct cytotoxicity without prior sensitization. Despite this initial cytotoxicity, OC cells develop various strategies to induce an immune-tolerant state. To this end, multiple immunosuppressive molecules are secreted to impair cytotoxic cells, recruit regulatory cells, alter antigen presentation, and effectively evade immune response. Consequently, OC TME is predominantly infiltrated by immunosuppressive cells such as FOXP3+ regulatory T-cells, M2-polarized macrophages and myeloid-derived suppressor cells. Despite this strong immunosuppressive state, PD-1/PD-L1 inhibitors have failed to improve outcomes. Beyond PD-1/PD-L1, OC expresses multiple other immune checkpoints that contribute to immune evasion, and each representing potential immune targets. Novel immunotherapies are attempting to overcome the immunosuppressive state and induce specific immune responses using antibodies adoptive cell therapy or vaccines. Overall, the OC TME presents both opportunities and obstacles. Immunotherapeutic approaches continue to show promise, and next-generation inhibitors offer exciting opportunities. However, tailoring therapies to individual immune characteristics will be critical for the success of these treatments.
Collapse
Affiliation(s)
- Felix Blanc-Durand
- Department of Haematology-Oncology, National University Cancer Institute, Singapore (NCIS), National University Hospital, Singapore, Singapore
- Yong Loo Lin School of Medicine and Cancer Science Institute (CSI), National University of Singapore (NUS), Singapore, Singapore
| | - Lai Clemence Wei Xian
- Department of Haematology-Oncology, National University Cancer Institute, Singapore (NCIS), National University Hospital, Singapore, Singapore
- Yong Loo Lin School of Medicine and Cancer Science Institute (CSI), National University of Singapore (NUS), Singapore, Singapore
| | - David S. P. Tan
- Department of Haematology-Oncology, National University Cancer Institute, Singapore (NCIS), National University Hospital, Singapore, Singapore
- Yong Loo Lin School of Medicine, National University Centre for Cancer Research (N2CR) and Cancer Science Institute (CSI), National University of Singapore, Singapore, Singapore
| |
Collapse
|
34
|
Jiang H, Fan W. Research progress on CD8+ T cell immune regulation in allogenic transplantation. Transpl Immunol 2023; 81:101945. [PMID: 37871888 DOI: 10.1016/j.trim.2023.101945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 10/16/2023] [Accepted: 10/19/2023] [Indexed: 10/25/2023]
Abstract
With advances in tissue typing, organ preservation techniques, and clinical surgery, organ transplantation has gained popularity as a treatment option for various end-stage diseases. Allogeneic transplantation has been widely adopted and extensively researched in clinical practice. Despite significant breakthroughs and progress in immunosuppression, this procedure is still associated with several adverse reactions and complications. Therefore, there is a continuing need to explore new immunological approaches to provide fresh insights and guidance for clinical transplantation. CD8+ T cells, traditionally known for their cytotoxic function and their ability to recognize transplanted organs as "non-self" entities, display cytotoxicity. However, recent studies have unveiled that CD8+ T cells have various subtypes and functions that extend beyond conventional cytotoxicity. These CD8+ T cell subtypes include Effector CD8+ T cells, Memory CD8+ T cells, and CD8Treg cells. This review examines the immune regulatory mechanisms of CD8+ T cells in allogeneic transplantation and discusses the potential applications of CD8+ T cells in treating tumors in transplant recipients who are receiving immunosuppressive therapy. These findings offer theoretical guidance for reducing post-transplant rejection reactions and improving adverse prognoses, offering new hope for improved clinical survival rate.
Collapse
Affiliation(s)
- Haowen Jiang
- Institute of Urology, The Third Medical Center of Chinese PLA General Hospital, Beijing 100039, China
| | - Wenmei Fan
- Institute of Urology, The Third Medical Center of Chinese PLA General Hospital, Beijing 100039, China.
| |
Collapse
|
35
|
Yilmaz I, Tavukcuoglu E, Horzum U, Yilmaz KB, Akinci M, Gulcelik MA, Oral HB, Esendagli G. Immune checkpoint status and exhaustion-related phenotypes of CD8 + T cells from the tumor-draining regional lymph nodes in breast cancer. Cancer Med 2023; 12:22196-22205. [PMID: 38069525 PMCID: PMC10757146 DOI: 10.1002/cam4.6802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 10/31/2023] [Accepted: 11/27/2023] [Indexed: 12/31/2023] Open
Abstract
BACKGROUND Functional status of T cells determines the responsiveness of cancer patients to immunotherapeutic interventions. Even though T cell-mediated immunity is inaugurated in the tumor-adjacent lymph nodes, peripheral blood has been routinely sampled for testing the immunological assays. The purpose of this study is to determine the immune checkpoint molecule expression and the exhaustion-related phenotype of cytotoxic T cells in the regional lymph nodes from breast cancer patients. PATIENTS AND METHODS Multicolor immunophenotyping was used to determine the expression of PD-1, TIM-3, LAG3, CTLA-4, CCR7, CD45RO, CD127, CD25, CXCR5, and ICOS molecules on CD3+ CD4- CD56- CD8+ cytotoxic T cells freshly obtained from the lymph nodes and the peripheral blood samples of the breast cancer patients. The results were assessed together with the clinical data. RESULTS A population of cytotoxic T cells was noted with high PD-1 and CXCR5 expression in the lymph nodes of the breast cancer patients. Co-expression of PD-1, CXCR5, TIM-3, and ICOS indicated a follicular helper T cell (Tfh)-like, exhaustion-related immunophenotype in these cytotoxic T cells. Only a minor population with CTLA-4 and LAG3 expression was noted. The PD-1+ CXCR5+ cytotoxic T cells largely displayed CD45RO+ CCR7+ central memory markers. The amount of CXCR5-expressing PD-1- cytotoxic T cells was elevated in the lymph nodes of the patients. CONCLUSION The regional lymph nodes of breast cancer patients harbor Tfh-like exhausted cytotoxic T lymphocytes with high PD-1 and TIM-3 checkpoint molecule expression. The immunological conditions in the regional lymph nodes should be implicated for immune checkpoint immunotherapy (ICI) of cancer.
Collapse
Affiliation(s)
- Izel Yilmaz
- Department of Medical Immunology, Institute of Health SciencesBursa Uludag UniversityBursaTurkey
- Department of Basic OncologyHacettepe University Cancer InstituteAnkaraTurkey
| | - Ece Tavukcuoglu
- Department of Basic OncologyHacettepe University Cancer InstituteAnkaraTurkey
| | - Utku Horzum
- Department of Basic OncologyHacettepe University Cancer InstituteAnkaraTurkey
| | - Kerim Bora Yilmaz
- Department of General Surgery, Gulhane Training and Research HospitalUniversity of Health SciencesAnkaraTurkey
- Department of Medical and Surgical ResearchHacettepe University Institute of Health SciencesAnkaraTurkey
| | - Melih Akinci
- Department of General Surgery, Gulhane Training and Research HospitalUniversity of Health SciencesAnkaraTurkey
| | - Mehmet Ali Gulcelik
- Department of General Surgery, Gulhane Training and Research HospitalUniversity of Health SciencesAnkaraTurkey
| | - Haluk Barbaros Oral
- Department of Immunology, Faculty of MedicineBursa Uludag UniversityBursaTurkey
| | - Gunes Esendagli
- Department of Basic OncologyHacettepe University Cancer InstituteAnkaraTurkey
- Department of Medical and Surgical ResearchHacettepe University Institute of Health SciencesAnkaraTurkey
| |
Collapse
|
36
|
Semeniuk-Wojtaś A, Modzelewska M, Poddębniak-Strama K, Kołaczyńska S, Lubas A, Górnicka B, Jakieła A, Stec R. CD4, CD20 and PD-L1 as Markers of Recurrence in Non-Muscle-Invasive Bladder Cancer. Cancers (Basel) 2023; 15:5529. [PMID: 38067231 PMCID: PMC10705362 DOI: 10.3390/cancers15235529] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 11/13/2023] [Accepted: 11/15/2023] [Indexed: 06/30/2024] Open
Abstract
INTRODUCTION A tumor microenvironment plays an important role in bladder cancer development and in treatment response. PURPOSE The aim of the study was to assess how the components of the microenvironment affect tumor recurrence and to find the potential biomarkers for immunotherapy in NMIBC. METHODS The study group consisted of 55 patients with primary NMIBC. Immunohistochemistry was performed on sections of primary papillary urothelial carcinoma of the bladder. Cox proportional hazard multiple regression analysis was performed to characterize tumors with the highest probability of an unfavorable outcome. RESULTS Multivariate analysis confirmed that the CD4 (p = 0.001), CD20 (p = 0.008) and PD-L1 expressed on tumor cells (p = 0.01) were independently associated with the risk of recurrence of bladder cancer. Patients with weak CD4+ cell infiltration (<4.6%) and severe CD20+ infiltration (>10%) belong to the group with a lower risk of recurrence. The cancer in this group also frequently recurs after 12 months (p = 0.0005). CONCLUSIONS The evaluation of CD4+ and CD20+ cells in the tumor microenvironment, in addition to PD-L1 on tumor cells, facilitates the determination of a group of patients with a low risk of recurrence.
Collapse
Affiliation(s)
| | | | | | - Sylwia Kołaczyńska
- Oncology Department, 4 Military Clinical Hospital with a Polyclinic, 53-114 Wroclaw, Poland
| | - Arkadiusz Lubas
- Department of Internal Medicine, Nephrology and Dialysis, Military Institute of Medicine—National Research Institute, 04-141 Warsaw, Poland
| | - Barbara Górnicka
- Pathomorphology Department, Medical University of Warsaw, 02-091 Warsaw, Poland
| | - Anna Jakieła
- Oncology Department, 4 Military Clinical Hospital with a Polyclinic, 53-114 Wroclaw, Poland
| | - Rafał Stec
- Oncology Department, Medical University of Warsaw, 02-091 Warsaw, Poland
| |
Collapse
|
37
|
Zhang Z, Duan Z, Cui Y. CD8 + T cells in brain injury and neurodegeneration. Front Cell Neurosci 2023; 17:1281763. [PMID: 38077952 PMCID: PMC10702747 DOI: 10.3389/fncel.2023.1281763] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 10/30/2023] [Indexed: 02/19/2024] Open
Abstract
The interaction between the peripheral immune system and the brain is increasingly being recognized as an important layer of neuroimmune regulation and plays vital roles in brain homeostasis as well as neurological disorders. As an important population of T-cell lymphocytes, the roles of CD8+ T cells in infectious diseases and tumor immunity have been well established. Recently, increasing number of complex functions of CD8+ T cells in brain disorders have been revealed. However, an advanced summary and discussion of the functions and mechanisms of CD8+ T cells in brain injury and neurodegeneration are still lacking. Here, we described the differentiation and function of CD8+ T cells, reviewed the involvement of CD8+ T cells in the regulation of brain injury including stroke and traumatic brain injury and neurodegenerative diseases, such as Alzheimer's disease (AD) and Parkinson's disease (PD), and discussed therapeutic prospects and future study goals. Understanding these processes will promote the investigation of T-cell immunity in brain disorders and provide new intervention strategies for the treatment of brain injury and neurodegeneration.
Collapse
Affiliation(s)
- Zhaolong Zhang
- Department of Interventional Radiology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Zhongying Duan
- Institute of Neuroregeneration and Neurorehabilitation, Qingdao University, Qingdao, Shandong, China
- Qingdao Medical College, Qingdao University, Qingdao, China
| | - Yu Cui
- Institute of Neuroregeneration and Neurorehabilitation, Qingdao University, Qingdao, Shandong, China
- Qingdao Medical College, Qingdao University, Qingdao, China
| |
Collapse
|
38
|
Koh CH, Lee S, Kwak M, Kim BS, Chung Y. CD8 T-cell subsets: heterogeneity, functions, and therapeutic potential. Exp Mol Med 2023; 55:2287-2299. [PMID: 37907738 PMCID: PMC10689838 DOI: 10.1038/s12276-023-01105-x] [Citation(s) in RCA: 68] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 08/11/2023] [Accepted: 08/12/2023] [Indexed: 11/02/2023] Open
Abstract
CD8 T cells play crucial roles in immune surveillance and defense against infections and cancer. After encountering antigenic stimulation, naïve CD8 T cells differentiate and acquire effector functions, enabling them to eliminate infected or malignant cells. Traditionally, cytotoxic T cells, characterized by their ability to produce effector cytokines and release cytotoxic granules to directly kill target cells, have been recognized as the constituents of the predominant effector T-cell subset. However, emerging evidence suggests distinct subsets of effector CD8 T cells that each exhibit unique effector functions and therapeutic potential. This review highlights recent advancements in our understanding of CD8 T-cell subsets and the contributions of these cells to various disease pathologies. Understanding the diverse roles and functions of effector CD8 T-cell subsets is crucial to discern the complex dynamics of immune responses in different disease settings. Furthermore, the development of immunotherapeutic approaches that specifically target and regulate the function of distinct CD8 T-cell subsets holds great promise for precision medicine.
Collapse
Affiliation(s)
- Choong-Hyun Koh
- Laboratory of Immune Regulation, Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Suyoung Lee
- Laboratory of Immune Regulation, Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826, Republic of Korea
- BK21 Plus Program, College of Pharmacy, Seoul National University, Seoul, 08826, Republic of Korea
| | - Minkyeong Kwak
- Laboratory of Immune Regulation, Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826, Republic of Korea
- BK21 Plus Program, College of Pharmacy, Seoul National University, Seoul, 08826, Republic of Korea
| | - Byung-Seok Kim
- Division of Life Sciences, College of Life Science and Bioengineering, Incheon National University, Incheon, 22012, Republic of Korea
| | - Yeonseok Chung
- Laboratory of Immune Regulation, Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826, Republic of Korea.
- BK21 Plus Program, College of Pharmacy, Seoul National University, Seoul, 08826, Republic of Korea.
- Wide River Institute of Immunology, Seoul National University, Hongcheon, Gangwon, 25159, Republic of Korea.
| |
Collapse
|
39
|
Kudryavtsev I, Benevolenskaya S, Serebriakova M, Grigor'yeva I, Kuvardin E, Rubinstein A, Golovkin A, Kalinina O, Zaikova E, Lapin S, Maslyanskiy A. Circulating CD8+ T Cell Subsets in Primary Sjögren's Syndrome. Biomedicines 2023; 11:2778. [PMID: 37893153 PMCID: PMC10604770 DOI: 10.3390/biomedicines11102778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 09/05/2023] [Accepted: 09/07/2023] [Indexed: 10/29/2023] Open
Abstract
Currently, multiple studies have indicated that CD8+ T lymphocytes play a role in causing damage to the exocrine glands through acinar injury in primary Sjögren's syndrome (pSS). The aim of this research was to assess the imbalance of circulating CD8+ T cell subsets. We analyzed blood samples from 34 pSS patients and 34 healthy individuals as controls. We used flow cytometry to enumerate CD8+ T cell maturation stages, using as markers CD62L, CD28, CD27, CD4, CD8, CD3, CD45RA and CD45. For immunophenotyping of 'polarized' CD8+ T cell subsets, we used the following monoclonal antibodies: CXCR5, CCR6, CXCR3 and CCR4. The findings revealed that both the relative and absolute numbers of 'naïve' CD8+ T cells were higher in pSS patients compared to the healthy volunteers. Conversely, the proportions of effector memory CD8+ T cells were notably lower. Furthermore, our data suggested that among patients with pSS, the levels of cytotoxic Tc1 CD8+ T cells were reduced, while the frequencies of regulatory cytokine-producing Tc2 and Tc17 CD8+ T cells were significantly elevated. Simultaneously, the Tc1 cell subsets displayed a negative correlation with immunoglobulin G, rheumatoid factor, the Schirmer test and unstimulated saliva flow. On the other hand, the Tc2 cell subsets exhibited a positive correlation with these parameters. In summary, our study indicated that immune dysfunction within CD8+ T cells, including alterations in Tc1 cells, plays a significant role in the development of pSS.
Collapse
Affiliation(s)
- Igor Kudryavtsev
- Federal State Budgetary Institution "Almazov National Medical Research Centre" of the Ministry of Health of the Russian Federation, St. Petersburg 197341, Russia
| | - Stanislava Benevolenskaya
- Federal State Budgetary Institution "Almazov National Medical Research Centre" of the Ministry of Health of the Russian Federation, St. Petersburg 197341, Russia
| | - Maria Serebriakova
- Federal State Budgetary Institution "Almazov National Medical Research Centre" of the Ministry of Health of the Russian Federation, St. Petersburg 197341, Russia
| | - Irina Grigor'yeva
- Federal State Budgetary Institution "Almazov National Medical Research Centre" of the Ministry of Health of the Russian Federation, St. Petersburg 197341, Russia
| | - Evgeniy Kuvardin
- Federal State Budgetary Institution "Almazov National Medical Research Centre" of the Ministry of Health of the Russian Federation, St. Petersburg 197341, Russia
| | - Artem Rubinstein
- Federal State Budgetary Institution "Almazov National Medical Research Centre" of the Ministry of Health of the Russian Federation, St. Petersburg 197341, Russia
| | - Alexey Golovkin
- Federal State Budgetary Institution "Almazov National Medical Research Centre" of the Ministry of Health of the Russian Federation, St. Petersburg 197341, Russia
| | - Olga Kalinina
- Federal State Budgetary Institution "Almazov National Medical Research Centre" of the Ministry of Health of the Russian Federation, St. Petersburg 197341, Russia
| | - Ekaterina Zaikova
- Federal State Budgetary Institution "Almazov National Medical Research Centre" of the Ministry of Health of the Russian Federation, St. Petersburg 197341, Russia
| | - Sergey Lapin
- Federal State Budgetary Educational Institution of Higher Education Academician I.P. Pavlov First St. Petersburg State Medical University of the Ministry of Healthcare of Russian Federation, St. Petersburg 197022, Russia
| | - Alexey Maslyanskiy
- Federal State Budgetary Institution "Almazov National Medical Research Centre" of the Ministry of Health of the Russian Federation, St. Petersburg 197341, Russia
| |
Collapse
|
40
|
Avery D, Morandini L, Gabriec M, Sheakley L, Peralta M, Donahue HJ, Martin RK, Olivares-Navarrete R. Contribution of αβ T cells to macrophage polarization and MSC recruitment and proliferation on titanium implants. Acta Biomater 2023; 169:605-624. [PMID: 37532133 PMCID: PMC10528595 DOI: 10.1016/j.actbio.2023.07.052] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 07/20/2023] [Accepted: 07/26/2023] [Indexed: 08/04/2023]
Abstract
Physiochemical cues like topography and wettability can impact the inflammatory response and tissue integration after biomaterial implantation. T cells are essential for immunomodulation of innate immune cells and play an important role in the host response to biomaterial implantation. This study aimed to understand how CD4+ and CD8+ T cell subsets, members of the αβ T cell family, polarize in response to smooth, rough, or rough-hydrophilic titanium (Ti) implants and whether their presence modulates immune cell crosstalk and mesenchymal stem cell (MSC) recruitment following biomaterial implantation. Post-implantation in mice, we found that CD4+ and CD8+ T cell subsets polarized differentially in response to modified Ti surfaces. Additionally, mice lacking αβ T cells had significantly more pro-inflammatory macrophages, fewer anti-inflammatory macrophages, and reduced MSC recruitment in response to modified Ti post-implantation than αβ T cell -competent mice. Our results demonstrate that T cell activation plays a significant role during the inflammatory response to implanted biomaterials, contributing to macrophage polarization and MSC recruitment and proliferation, and the absence of αβ T cells compromises new bone formation at the implantation site. STATEMENT OF SIGNIFICANCE: T cells are essential for immunomodulation and play an important role in the host response to biomaterial implantation. Our results demonstrate that T cells actively participate during the inflammatory response to implanted biomaterials, controlling macrophage phenotype and recruitment of MSCs to the implantation site.
Collapse
Affiliation(s)
- Derek Avery
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, VA, United States
| | - Lais Morandini
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, VA, United States
| | - Melissa Gabriec
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, VA, United States
| | - Luke Sheakley
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, VA, United States
| | - Matthieu Peralta
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, VA, United States
| | - Henry J Donahue
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, VA, United States
| | - Rebecca K Martin
- Department of Microbiology and Immunology, School of Medicine, Virginia Commonwealth University, Richmond, VA, United States
| | - Rene Olivares-Navarrete
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, VA, United States.
| |
Collapse
|
41
|
Kudryavtsev I, Zinchenko Y, Serebriakova M, Akisheva T, Rubinstein A, Savchenko A, Borisov A, Belenjuk V, Malkova A, Yablonskiy P, Kudlay D, Starshinova A. A Key Role of CD8+ T Cells in Controlling of Tuberculosis Infection. Diagnostics (Basel) 2023; 13:2961. [PMID: 37761328 PMCID: PMC10528134 DOI: 10.3390/diagnostics13182961] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 08/30/2023] [Accepted: 09/13/2023] [Indexed: 09/29/2023] Open
Abstract
The main role in the control of tuberculosis infection is played by macrophages and Th1 and CD8+ T cells. The study aimed to identify the most diagnostically significant CD8+ T cell subsets in tuberculosis patients. METHODS Peripheral blood samples from patients with clinical, radiological, and bacteriologically confirmed pulmonary tuberculosis (TB, n = 32) and healthy subjects (HC, n = 31) were collected and analyzed using 10-color flow cytometry. RESULTS The frequency of the EM4 CD3+CD8+ cells was reduced in the peripheral blood of patients with pulmonary tuberculosis, while the relative and absolute number of EM1 CD3+CD8+ cells increased compared to the control group. CD57 expression was reduced in patients with pulmonary tuberculosis on EM1, EM2, and pE1 CD3+CD8+ cells, whereas the EM3 cells had a high level of CD57 expression. The relative and absolute number of Tc2 (CCR6-CXCR3-) cells in peripheral blood in patients with pulmonary tuberculosis was increased, while the frequency of Tc1 (CCR6-CXCR3+) was decreased, compared to healthy donors. CONCLUSIONS Patients with pulmonary tuberculosis have an abnormal CD3+CD8+ cell profile and demonstrate their impaired maturation and functional activity.
Collapse
Affiliation(s)
- Igor Kudryavtsev
- Institution of Experimental Medicine, Department of Immunology, 197376 St-Petersburg, Russia; (I.K.); (M.S.); (T.A.); (A.R.)
- Almazov National Medical Research Centre, 197341 St-Petersburg, Russia
| | - Yulia Zinchenko
- Research Institute of Phthisiopulmonology, 191036 St-Petersburg, Russia; (Y.Z.); (P.Y.)
| | - Maria Serebriakova
- Institution of Experimental Medicine, Department of Immunology, 197376 St-Petersburg, Russia; (I.K.); (M.S.); (T.A.); (A.R.)
| | - Tatiana Akisheva
- Institution of Experimental Medicine, Department of Immunology, 197376 St-Petersburg, Russia; (I.K.); (M.S.); (T.A.); (A.R.)
| | - Artem Rubinstein
- Institution of Experimental Medicine, Department of Immunology, 197376 St-Petersburg, Russia; (I.K.); (M.S.); (T.A.); (A.R.)
| | - Andrei Savchenko
- Federal Research Center «Krasnoyarsk Science Center» of the Siberian Branch of the Russian Academy of Sciences, Scientific Research Institute of Medical Problems of the North, 660022 Krasnoyarsk, Russia; (A.S.); (A.B.); (V.B.)
| | - Alexandr Borisov
- Federal Research Center «Krasnoyarsk Science Center» of the Siberian Branch of the Russian Academy of Sciences, Scientific Research Institute of Medical Problems of the North, 660022 Krasnoyarsk, Russia; (A.S.); (A.B.); (V.B.)
| | - Vasilij Belenjuk
- Federal Research Center «Krasnoyarsk Science Center» of the Siberian Branch of the Russian Academy of Sciences, Scientific Research Institute of Medical Problems of the North, 660022 Krasnoyarsk, Russia; (A.S.); (A.B.); (V.B.)
| | - Anna Malkova
- Department of Molecular Biology, Faculty of Natural Sciences, Ariel University, Ariel 40700, Israel;
| | - Piotr Yablonskiy
- Research Institute of Phthisiopulmonology, 191036 St-Petersburg, Russia; (Y.Z.); (P.Y.)
- St. Petersburg Research Institute of Phthisiopulmonology, Ligovskii Prospect, 2–4, 191036 St-Petersburg, Russia
| | - Dmitry Kudlay
- Department of Pharmacology, I.M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia;
- NRC Institute of Immunology FMBA of Russia, 115552 Moscow, Russia
| | - Anna Starshinova
- Almazov National Medical Research Centre, 197341 St-Petersburg, Russia
| |
Collapse
|
42
|
Amin J, Gee C, Stowell K, Coulthard D, Boche D. T Lymphocytes and Their Potential Role in Dementia with Lewy Bodies. Cells 2023; 12:2283. [PMID: 37759503 PMCID: PMC10528562 DOI: 10.3390/cells12182283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 09/08/2023] [Accepted: 09/12/2023] [Indexed: 09/29/2023] Open
Abstract
Dementia with Lewy bodies (DLB) is the second most common neurodegenerative cause of dementia. People with DLB have an inferior prognosis compared to Alzheimer's disease (AD), but the diseases overlap in their neuropathology and clinical syndrome. It is imperative that we enhance our understanding of the aetiology and pathogenesis of DLB. The impact of peripheral inflammation on the brain in dementia has been increasingly explored in recent years, with T lymphocyte recruitment into brain parenchyma identified in AD and Parkinson's disease. There is now a growing range of literature emerging on the potential role of innate and adaptive immune cells in DLB, including T lymphocytes. In this review, we examine the profile of T lymphocytes in DLB, focusing on studies of post-mortem brain tissue, cerebrospinal fluid, and the blood compartment. We present an integrated viewpoint on the results of these studies by proposing how changes to the T lymphocyte profile in the brain and periphery may relate to each other. Improving our understanding of T lymphocytes in DLB has the potential to guide the development of disease-modifying treatments.
Collapse
Affiliation(s)
- Jay Amin
- Clinical Neurosciences, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton SO17 1BJ, UK
- Memory Assessment and Research Centre, Tom Rudd Unit, Moorgreen Hospital, Southern Health NHS Foundation Trust, Southampton SO30 3JB, UK
| | - Claire Gee
- Clinical Neurosciences, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton SO17 1BJ, UK
- Memory Assessment and Research Centre, Tom Rudd Unit, Moorgreen Hospital, Southern Health NHS Foundation Trust, Southampton SO30 3JB, UK
| | - Kiran Stowell
- Clinical Neurosciences, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton SO17 1BJ, UK
| | - Daisy Coulthard
- Clinical Neurosciences, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton SO17 1BJ, UK
| | - Delphine Boche
- Clinical Neurosciences, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton SO17 1BJ, UK
| |
Collapse
|
43
|
Yuan N, Pan HH, Liang YS, Hu HL, Zhai CL, Wang B. Identification of prognostic and diagnostic signatures for cancer and acute myocardial infarction: multi-omics approaches for deciphering heterogeneity to enhance patient management. Front Pharmacol 2023; 14:1249145. [PMID: 37781709 PMCID: PMC10539594 DOI: 10.3389/fphar.2023.1249145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 09/04/2023] [Indexed: 10/03/2023] Open
Abstract
Patients diagnosed with cancer face an increased risk of cardiovascular events in the short term, while those experiencing acute myocardial infarction (AMI) have a higher incidence of cancer. Given limitations in clinical resources, identifying shared biomarkers offers a cost-effective approach to risk assessment by minimizing the need for multiple tests and screenings. Hence, it is crucial to identify common biomarkers for both cancer survival and AMI prediction. Our study suggests that monocyte-derived biomarkers, specifically WEE1, PYHIN1, SEC61A2, and HAL, hold potential as predictors for cancer prognosis and AMI. We employed a novel formula to analyze mRNA levels in clinical samples from patients with AMI and cancer, resulting in the development of a new risk score based on expression profiles. By categorizing patients into high-risk and low-risk groups based on the median risk score, we observed significantly poorer overall survival among high-risk patients in cancer cohorts using Kaplan-Meier analysis. Furthermore, calibration curves, decision curve analysis (DCA), and clinical impact curve analyses provided additional evidence supporting the robust diagnostic capacity of the risk score for AMI. Noteworthy is the shared activation of the Notch Signaling pathway, which may shed light on common high-risk factors underlying both AMI and cancer. Additionally, we validated the differential expression of these genes in cell lines and clinical samples, respectively, reinforcing their potential as meaningful biomarkers. In conclusion, our study demonstrates the promise of mRNA levels as biomarkers and emphasizes the significance of further research for validation and refinement.
Collapse
Affiliation(s)
- Na Yuan
- The First Hospital of Jiaxing Affiliated Hospitial of Jiaxing University, Jiaxing, Zhejiang, China
| | - Hai-Hua Pan
- The First Hospital of Jiaxing Affiliated Hospitial of Jiaxing University, Jiaxing, Zhejiang, China
| | - Yan-Shan Liang
- Affiliated Dongguan Hospital, Southern Medical University, Dongguan, Guangdong, China
| | - Hui-Lin Hu
- The First Hospital of Jiaxing Affiliated Hospitial of Jiaxing University, Jiaxing, Zhejiang, China
| | - Chang-Lin Zhai
- The First Hospital of Jiaxing Affiliated Hospitial of Jiaxing University, Jiaxing, Zhejiang, China
| | - Bo Wang
- The Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| |
Collapse
|
44
|
Mahajan S, Alexander A, Koenig Z, Saba N, Prasanphanich N, Hildeman DA, Chougnet CA, DeFranco E, Andorf S, Tilburgs T. Antigen-specific decidual CD8+ T cells include distinct effector memory and tissue-resident memory cells. JCI Insight 2023; 8:e171806. [PMID: 37681414 PMCID: PMC10544202 DOI: 10.1172/jci.insight.171806] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 07/25/2023] [Indexed: 09/09/2023] Open
Abstract
Maternal decidual CD8+ T cells must integrate the antithetical demands of providing immunity to infection while maintaining immune tolerance for fetal and placental antigens. Human decidual CD8+ T cells were shown to be highly differentiated memory T cells with mixed signatures of dysfunction, activation, and effector function. However, no information is present on how specificity for microbial or fetal antigens relates to their function or dysfunction. In addition, a key question, whether decidual CD8+ T cells include unique tissue-resident memory T cells (Trm) or also effector memory T cell (Tem) types shared with peripheral blood populations, is unknown. Here, high-dimensional flow cytometry of decidual and blood CD8+ T cells identified 2 Tem populations shared in blood and decidua and 9 functionally distinct Trm clusters uniquely found in decidua. Interestingly, fetus- and virus-specific decidual CD8+ Trm cells had similar features of inhibition and cytotoxicity, with no significant differences in their expression of activation, inhibitory, and cytotoxic molecules, suggesting that not all fetus-specific CD8+ T cell responses are suppressed at the maternal-fetal interface. Understanding how decidual CD8+ T cell specificity relates to their function and tissue residency is crucial in advancing understanding of their contribution to placental inflammation and control of congenital infections.
Collapse
Affiliation(s)
- Shweta Mahajan
- Immunology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- Division of Immunobiology
| | - Aria Alexander
- Immunology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- Division of Immunobiology
| | - Zachary Koenig
- Immunology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- Division of Immunobiology
| | | | - Nina Prasanphanich
- Division of Immunobiology
- Division of Infectious disease, Cincinnati Children’s Hospital, Cincinnati, Ohio, USA
| | | | | | - Emily DeFranco
- Department of Obstetrics and Gynecology, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Sandra Andorf
- Division of Biomedical Informatics, and
- Department of Pediatrics, and
- Division of Allergy & Immunology, and
| | - Tamara Tilburgs
- Division of Immunobiology
- Department of Pediatrics, and
- Center for Inflammation and Tolerance, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| |
Collapse
|
45
|
Ramirez F, Zambrano A, Hennis R, Holland N, Lakshmanaswamy R, Chacon J. Sending a Message: Use of mRNA Vaccines to Target the Tumor Immune Microenvironment. Vaccines (Basel) 2023; 11:1465. [PMID: 37766141 PMCID: PMC10534833 DOI: 10.3390/vaccines11091465] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 08/25/2023] [Accepted: 09/05/2023] [Indexed: 09/29/2023] Open
Abstract
While cancer immunotherapies have become central to treatment, challenges associated with the ability of tumors to evade the immune system remain significant obstacles. At the heart of this issue is the tumor immune microenvironment, the complex interplay of the tumor microenvironment and the immune response. Recent advances in mRNA cancer vaccines represent major progress towards overcoming some of the challenges posed by deleterious components of the tumor immune microenvironment. Indeed, major breakthroughs in mRNA vaccine technology, such as the use of replacement nucleotides and lipid nanoparticle delivery, led to the vital success of mRNA vaccine technology in fighting COVID-19. This has in turn generated massive additional interest and investment in the platform. In this review, we detail recent research in the nature of the tumor immune microenvironment and in mRNA cancer vaccines and discuss applications by which mRNA cancer vaccines, often in combination with various adjuvants, represent major areas of potential in overcoming tumor immune microenvironment-imposed obstacles. To this end, we also review current mRNA cancer vaccine clinical trials.
Collapse
Affiliation(s)
- Fabiola Ramirez
- Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA; (F.R.); (A.Z.); (R.H.); (N.H.); (R.L.)
| | - Angelica Zambrano
- Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA; (F.R.); (A.Z.); (R.H.); (N.H.); (R.L.)
| | - Robert Hennis
- Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA; (F.R.); (A.Z.); (R.H.); (N.H.); (R.L.)
| | - Nathan Holland
- Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA; (F.R.); (A.Z.); (R.H.); (N.H.); (R.L.)
| | - Rajkumar Lakshmanaswamy
- Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA; (F.R.); (A.Z.); (R.H.); (N.H.); (R.L.)
- L. Frederick Francis Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA
| | - Jessica Chacon
- Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA; (F.R.); (A.Z.); (R.H.); (N.H.); (R.L.)
| |
Collapse
|
46
|
Chen S, Wang Q, Wang H, Xia S. Endoplasmic reticulum stress in T cell-mediated diseases. Scand J Immunol 2023; 98:e13307. [PMID: 38441291 DOI: 10.1111/sji.13307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 05/23/2023] [Accepted: 06/18/2023] [Indexed: 03/07/2024]
Abstract
T cells synthesize a large number of proteins during their development, activation, and differentiation. The build-up of misfolded and unfolded proteins in the endoplasmic reticulum, however, causes endoplasmic reticulum (ER) stress. Thus, T cells can maintain ER homeostasis via endoplasmic reticulum-associated degradation, unfolded protein response, and autophagy. In T cell-mediated diseases, such as rheumatoid arthritis, systemic lupus erythematosus, Sjogren's syndrome, type 1 diabetes and vitiligo, ER stress caused by changes in the internal microenvironment can cause disease progression by affecting T cell homeostasis. This review discusses ER stress in T cell formation, activation, differentiation, and T cell-mediated illnesses, and may offer new perspectives on the involvement of T cells in autoimmune disorders and cancer.
Collapse
Affiliation(s)
- Shaodan Chen
- Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Qiulei Wang
- Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Hui Wang
- Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Sheng Xia
- Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang, China
| |
Collapse
|
47
|
Hipp AV, Bengsch B, Globig AM. Friend or Foe - Tc17 cell generation and current evidence for their importance in human disease. DISCOVERY IMMUNOLOGY 2023; 2:kyad010. [PMID: 38567057 PMCID: PMC10917240 DOI: 10.1093/discim/kyad010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 06/12/2023] [Accepted: 07/19/2023] [Indexed: 04/04/2024]
Abstract
The term Tc17 cells refers to interleukin 17 (IL-17)-producing CD8+ T cells. While IL-17 is an important mediator of mucosal defense, it is also centrally involved in driving the inflammatory response in immune-mediated diseases, such as psoriasis, multiple sclerosis, and inflammatory bowel disease. In this review, we aim to gather the current knowledge on the phenotypic and transcriptional profile, the in vitro and in vivo generation of Tc17 cells, and the evidence pointing towards a relevant role of Tc17 cells in human diseases such as infectious diseases, cancer, and immune-mediated diseases.
Collapse
Affiliation(s)
- Anna Veronika Hipp
- Clinic for Internal Medicine II, Gastroenterology, Hepatology, Endocrinology, and Infectious Diseases, University Medical Center Freiburg, Faculty of Medicine, Freiburg, Germany
| | - Bertram Bengsch
- Clinic for Internal Medicine II, Gastroenterology, Hepatology, Endocrinology, and Infectious Diseases, University Medical Center Freiburg, Faculty of Medicine, Freiburg, Germany
| | - Anna-Maria Globig
- Clinic for Internal Medicine II, Gastroenterology, Hepatology, Endocrinology, and Infectious Diseases, University Medical Center Freiburg, Faculty of Medicine, Freiburg, Germany
| |
Collapse
|
48
|
Yang Y, Li J, Jing C, Zhai Y, Bai Z, Yang Y, Deng W. Inhibition of neuroactive ligand-receptor interaction pathway can enhance immunotherapy response in colon cancer: an in silico study. Expert Rev Anticancer Ther 2023; 23:1205-1215. [PMID: 37555253 DOI: 10.1080/14737140.2023.2245567] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 07/17/2023] [Accepted: 07/19/2023] [Indexed: 08/10/2023]
Abstract
BACKGROUND The potential mechanism underlying the association between Homologous recombination deficiency (HRD) and immunotherapy in colon cancer has not been investigated. METHODS The exon sequencing data and transcriptome data of 456 colon adenocarcinoma (COAD) patients were obtained from the TCGA database. Pathway activity score was calculated by GSVA methods and engaged in further survival analysis. The prognostic value of the candidate pathways was validated in an external GEO cohort and an immunotherapy cohort. RESULTS Patients with high HRD were associated with poor prognosis, lower tumor mutation burden and microsatellite instability, higher fraction genome alteration, and less sensitivity to immunotherapy in COAD. And then, the neuroactive ligand-receptor interaction pathway was over-activated in high-HRD tumors and associated with immunosuppression in colon cancer with high HRD. Besides, the pathway was associated with prognosis and immunotherapy response in COAD. Moreover, genes in this pathway such as LTB4R2 can be used as a novel target for therapy development in colon cancer. CONCLUSION Our study not only revealed the potential mechanism of HRD and the function of the neuroactive ligand-receptor interaction pathway in colon cancer but also provided new clues for the improvement of immunotherapy response in colon cancer.
Collapse
Affiliation(s)
- Yun Yang
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Jun Li
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Chao Jing
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Yuhao Zhai
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Zhigang Bai
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research & National Clinical Research Center for Digestive Diseases, Beijing, China
| | - Yingchi Yang
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research & National Clinical Research Center for Digestive Diseases, Beijing, China
| | - Wei Deng
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research & National Clinical Research Center for Digestive Diseases, Beijing, China
| |
Collapse
|
49
|
Wang Z, Zhang T, Jia F, Ge C, He Y, Tian Y, Wang W, Yang G, Huang H, Wang J, Shi C, Yang W, Cao X, Zeng Y, Wang N, Qian A, Wang C, Jiang Y. Homologous Sequential Immunization Using Salmonella Oral Administration Followed by an Intranasal Boost with Ferritin-Based Nanoparticles Enhanced the Humoral Immune Response against H1N1 Influenza Virus. Microbiol Spectr 2023; 11:e0010223. [PMID: 37154735 PMCID: PMC10269571 DOI: 10.1128/spectrum.00102-23] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Accepted: 04/17/2023] [Indexed: 05/10/2023] Open
Abstract
The influenza virus continues to pose a great threat to public health due to the frequent variations in RNA viruses. Vaccines targeting conserved epitopes, such as the extracellular domain of the transmembrane protein M2 (M2e), a nucleoprotein, and the stem region of hemagglutinin proteins, have been developed, but more efficient strategies, such as nanoparticle-based vaccines, are still urgently needed. However, the labor-intensive in vitro purification of nanoparticles is still necessary, which could hinder the application of nanoparticles in the veterinary field in the future. To overcome this limitation, we used regulated lysis Salmonella as an oral vector with which to deliver three copies of M2e (3M2e-H1N1)-ferritin nanoparticles in situ and evaluated the immune response. Then, sequential immunization using Salmonella-delivered nanoparticles followed by an intranasal boost with purified nanoparticles was performed to further improve the efficiency. Compared with 3M2e monomer administration, Salmonella-delivered in situ nanoparticles significantly increased the cellular immune response. Additionally, the results of sequential immunization showed that the intranasal boost with purified nanoparticles dramatically stimulated the activation of lung CD11b dendritic cells (DCs) and elevated the levels of effector memory T (TEM) cells in both spleen and lung tissues as well as those of CD4 and CD8 tissue-resident memory T (TRM) cells in the lungs. The increased production of mucosal IgG and IgA antibody titers was also observed, resulting in further improvements to protection against a virus challenge, compared with the pure oral immunization group. Salmonella-delivered in situ nanoparticles efficiently increased the cellular immune response, compared with the monomer, and sequential immunization further improved the systemic immune response, as shown by the activation of DCs, the production of TEM cells and TRM cells, and the mucosal immune response, thereby providing us with a novel strategy by which to apply nanoparticle-based vaccines in the future. IMPORTANCE Salmonella-delivered in situ nanoparticle platforms may provide novel nanoparticle vaccines for oral administration, which would be beneficial for veterinary applications. The combination of administering Salmonella-vectored, self-assembled nanoparticles and an intranasal boost with purified nanoparticles significantly increased the production of effector memory T cells and lung resident memory T cells, thereby providing partial protection against an influenza virus challenge. This novel strategy could open a novel avenue for the application of nanoparticle vaccines for veterinary purposes.
Collapse
Affiliation(s)
- Zhannan Wang
- College of Animal Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Tongyu Zhang
- College of Animal Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Futing Jia
- College of Animal Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Chongbo Ge
- College of Animal Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Yingkai He
- College of Animal Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Yawen Tian
- College of Animal Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Wenfeng Wang
- College of Animal Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Guilian Yang
- College of Animal Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Haibin Huang
- College of Animal Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Jianzhong Wang
- College of Animal Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Chunwei Shi
- College of Animal Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Wentao Yang
- College of Animal Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Xin Cao
- College of Animal Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Yan Zeng
- College of Animal Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Nan Wang
- College of Animal Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Aidong Qian
- College of Animal Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Chunfeng Wang
- College of Animal Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Yanlong Jiang
- College of Animal Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, China
| |
Collapse
|
50
|
De Simoni O, Dal Santo L, Scarpa M, Munari G, Spolverato YC, Scapinello A, Lonardi S, Soldà C, Bergamo F, Fantin A, Bardini R, Pilati P, Fassan M, Gruppo M. Role of Immune Microenvironment in Pancreatic Ductal Adenocarcinoma: Could It Be Considered a Predictor of Prognosis? Curr Oncol 2023; 30:5515-5528. [PMID: 37366900 PMCID: PMC10296875 DOI: 10.3390/curroncol30060417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 05/23/2023] [Accepted: 06/06/2023] [Indexed: 06/28/2023] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is characterized by a highly immunosuppressive tumor microenvironment (TME). The aim of this study is to determine the potential significant TME immune markers of long-term survival. METHODS We retrospectively included patients with a diagnosis of resectable PDAC having undergone upfront surgery. Immunohistochemical (IHC) staining using tissue microarray for PD-L1, CD3, CD4, CD8, FOXP3, CD20, iNOS and CD163 was performed in order to characterize the TME. The primary endpoint was long-term survival, defined as the Overall Survival > 24 months from surgery. RESULTS A total of 38 consecutive patients were included, and 14 (36%) of them were long-term survivors. Long-term survivors showed a higher density of CD8+ lymphocytes intra- and peri-acinar (p = 0.08), and a higher CD8/FOXP3 intra- and peri-tumoral ratio (p = 0.05). A low density of intra- and peri-tumoral FOXP3 infiltration is a good predictor of long-term survival (p = 0.04). A significant association of the low density of intra- and peri-tumoral tumor-associated macrophages (TAMs) iNOS+ with long-term survival was detected (p = 0.04). CONCLUSIONS Despite the retrospective nature and small sample size, our study showed that the high infiltration of CD8+ lymphocytes and low infiltration of FOXP3+ and TAMs iNOS+ are predictors of good prognosis. A preoperative assessment of these potential immune markers could be useful and determinant in the staging process and in PDAC management.
Collapse
Affiliation(s)
- Ottavia De Simoni
- Surgical Oncology of Digestive Tract Unit, Veneto Institute of Oncology (IOV-IRCCS), 35128 Padua, Italy; (P.P.); (M.G.)
| | - Luca Dal Santo
- Pathology Unit, Department of Medicine, University of Padova, 35128 Padua, Italy; (L.D.S.); (M.F.)
| | - Marco Scarpa
- Chirurgia Generale 3, Azienda Ospedale Università Padova, 35128 Padua, Italy;
| | - Giada Munari
- Veneto Institute of Oncology (IOV-IRCCS), 35128 Padua, Italy;
| | | | - Antonio Scapinello
- Anatomy and Pathological Histology Unit, Veneto Institute of Oncology (IOV-IRCCS), 35128 Padua, Italy;
| | - Sara Lonardi
- Unit of Medical Oncology 3, Veneto Institute of Oncology (IOV-IRCCS), 35128 Padua, Italy;
| | - Caterina Soldà
- Unit of Medical Oncology 1, Veneto Institute of Oncology (IOV-IRCCS), 35128 Padua, Italy; (C.S.); (F.B.)
| | - Francesca Bergamo
- Unit of Medical Oncology 1, Veneto Institute of Oncology (IOV-IRCCS), 35128 Padua, Italy; (C.S.); (F.B.)
| | - Alberto Fantin
- Gastroenterology Unit, Veneto Institute of Oncology (IOV-IRCCS), 35128 Padua, Italy;
| | - Romeo Bardini
- General Surgery Unit, Azienda Ospedaliera di Padova, 35128 Padua, Italy; (Y.C.S.); (R.B.)
| | - Pierluigi Pilati
- Surgical Oncology of Digestive Tract Unit, Veneto Institute of Oncology (IOV-IRCCS), 35128 Padua, Italy; (P.P.); (M.G.)
| | - Matteo Fassan
- Pathology Unit, Department of Medicine, University of Padova, 35128 Padua, Italy; (L.D.S.); (M.F.)
- Veneto Institute of Oncology (IOV-IRCCS), 35128 Padua, Italy;
| | - Mario Gruppo
- Surgical Oncology of Digestive Tract Unit, Veneto Institute of Oncology (IOV-IRCCS), 35128 Padua, Italy; (P.P.); (M.G.)
| |
Collapse
|