1
|
Su S, Yang B, Liu H, Yin W, Chen S, Du G. Four genes shared between rheumatoid arthritis and cervical cancer are associated with cervical cancer prognosis. Sci Rep 2025; 15:15884. [PMID: 40335617 PMCID: PMC12059032 DOI: 10.1038/s41598-025-01177-x] [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: 01/08/2025] [Accepted: 05/05/2025] [Indexed: 05/09/2025] Open
Abstract
Rheumatoid arthritis (RA) and cervical cancer (CC) are major global health challenges, yet the molecular connections between these two conditions remain poorly understood. To bridge this gap, our study employs bioinformatics approaches to explore shared genetic pathways and potential biomarkers. We started by identifying differentially expressed genes in RA and CC and then applied WGCNA to detect functionally related gene clusters using gene expression data from the GEO database. Additionally, we constructed protein-protein interaction (PPI) networks and examined the role of the immune microenvironment. To assess the prognostic relevance of key genes in CC, we leveraged survival data from TCGA. Our analysis identified 55 key genes common to RA and CC, with four-CXCL1, CXCL13, ZWINT, and PTTG1-emerging as significant. ROC curve validation confirmed their diagnostic potential, and a model incorporating these genes was associated with poorer prognosis in CC. Among them, CXCL1 stood out as especially crucial. Our findings suggest a potential link between chronic inflammation, immune dysregulation, and chemokine-related pathways in RA patients, which may contribute to an increased susceptibility to CC.
Collapse
Affiliation(s)
- Shuqiong Su
- Department of Oncology, Guangzhou Royallee Hospital, Guangzhou, China
| | - Bo Yang
- Department of Oncology, The People's Hospital of Yichun City, Yichun, China
| | - Huixia Liu
- Department of Oncology, Guangzhou Royallee Hospital, Guangzhou, China
| | - Wenyao Yin
- Department of Cardiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.
| | - Shuo Chen
- The First Clinical Medicine College, Sun Yat-Sen University, Guangzhou, China
| | - Ge Du
- Department of Oncology, Foresea Life Insurance Guangzhou General Hospital, Guangzhou, China
| |
Collapse
|
2
|
Wang C, Zhang Y, Liu T, Mi Z, Shi P, Wang Z, Li W, Wang H, Liu H, Zhang F. Insufficient CXCL13 secretion in leprosy foamy macrophages attenuates lymphocyte recruitment and antimicrobial protein production. Front Immunol 2025; 16:1541954. [PMID: 40264781 PMCID: PMC12011874 DOI: 10.3389/fimmu.2025.1541954] [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: 12/09/2024] [Accepted: 03/21/2025] [Indexed: 04/24/2025] Open
Abstract
Background Pathogens trigger metabolic reprogramming, leading to the formation of foamy macrophages (FMs). This process provides a favorable environment for bacterial proliferation and enables bacteria to evade immune killing. Objective To elucidate the mechanisms by which pathogens escape immune surveillance and elimination via the formation of FMs. Methods We constructed a FM model using monocyte-derived macrophages (MDMs) that were incubated with oxidized low-density lipoprotein (oxLDL). Subsequently, we employed bulk RNA-sequencing (bulk RNA-seq) to comprehensively analyze the immune responses in MDMs and FMs against Mycobacterium leprae (M. leprae) infection in samples from 10 healthy individuals. Results We found that CXCL13, a component of the cytokine-cytokine receptor interaction pathway, was specifically upregulated in M. leprae infected MDMs, when compared with M. leprae infected FMs. Significantly, further functional analyses revealed that in vitro treatment with CXCL13 could enhance the expression of CXCR5, thereby promoting lymphocyte migration and secretion of antimicrobial proteins. Additionally, NLRP12 was found to be specifically and highly expressed in the NOD-like receptor signaling pathway, which was enriched in infected FMs. In macrophages, M. leprae infection increased CXCL13 expression via NF-κB signal pathway. Conversely, in FMs, mycobacteria induced upregulation of CXCL13 was suppressed by NLRP12 through the inhibition of p52 factor expression. Conclusion In conclusion, the NLRP12/NF-κB/CXCL13 axis is crucial for the immune response of FMs after mycobacterial infection. These findings contribute to a deeper understanding of the pathological mechanisms of mycobacterial infection.
Collapse
Affiliation(s)
- Chuan Wang
- Hospital for Skin Diseases, Shandong First Medical University, Jinan, Shandong, China
- Shandong Provincial Institute of Dermatology and Venereology, Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Yuan Zhang
- Hospital for Skin Diseases, Shandong First Medical University, Jinan, Shandong, China
- Shandong Provincial Institute of Dermatology and Venereology, Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Tingting Liu
- Hospital for Skin Diseases, Shandong First Medical University, Jinan, Shandong, China
- Shandong Provincial Institute of Dermatology and Venereology, Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Zihao Mi
- Hospital for Skin Diseases, Shandong First Medical University, Jinan, Shandong, China
- Shandong Provincial Institute of Dermatology and Venereology, Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Peidian Shi
- Hospital for Skin Diseases, Shandong First Medical University, Jinan, Shandong, China
- Shandong Provincial Institute of Dermatology and Venereology, Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Zhenzhen Wang
- Hospital for Skin Diseases, Shandong First Medical University, Jinan, Shandong, China
- Shandong Provincial Institute of Dermatology and Venereology, Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Wenchao Li
- Hospital for Skin Diseases, Shandong First Medical University, Jinan, Shandong, China
- Shandong Provincial Institute of Dermatology and Venereology, Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Honglei Wang
- Hospital for Skin Diseases, Shandong First Medical University, Jinan, Shandong, China
- Shandong Provincial Institute of Dermatology and Venereology, Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Hong Liu
- Hospital for Skin Diseases, Shandong First Medical University, Jinan, Shandong, China
- Shandong Provincial Institute of Dermatology and Venereology, Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Furen Zhang
- Hospital for Skin Diseases, Shandong First Medical University, Jinan, Shandong, China
- Shandong Provincial Institute of Dermatology and Venereology, Shandong Academy of Medical Sciences, Jinan, Shandong, China
| |
Collapse
|
3
|
Xin M, Wang A, Ji M, Wu J, Jiang B, Shi M, Song L, Xin Z. Molecular Biology and Functions of T Follicular Helper Cells in Cancer and Immunotherapy. Immune Netw 2025; 25:e7. [PMID: 40342840 PMCID: PMC12056291 DOI: 10.4110/in.2025.25.e7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 12/22/2024] [Accepted: 01/08/2025] [Indexed: 05/11/2025] Open
Abstract
T follicular helper (Tfh) cells are integral to the germinal center (GC) response and the development of potent humoral immunity. By priming B cells, Tfh cells can initiate both extrafollicular and GC-dependent Ab responses. The dynamic physical interactions between Tfh and B cells constitute the primary platform for Tfh cells to provide essential "help" factors to B cells, as well as for reciprocal signaling from B cells to sustain the helper state of Tfh cells. In recent years, significant advancements have been made in understanding the diverse roles of Tfh cells across various diseases, particularly in cancer. Notably, beyond the classical GC-Tfh cells, it is increasingly recognized that the Tfh cell phenotype is highly heterogeneous and dynamic, which adds complexity to their roles in disease contexts. This review aims to encapsulate progress in Tfh cell biology, with a focus on their role in cancer and immunotherapy.
Collapse
Affiliation(s)
- Mengyuan Xin
- Department of Thoracic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, China
- Medical Science and Technology Innovation Center, Shandong First Medical University, Jinan 250117, China
| | - Antuo Wang
- Department of Thoracic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, China
- Medical Science and Technology Innovation Center, Shandong First Medical University, Jinan 250117, China
| | - Minghao Ji
- Department of Thoracic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, China
| | - Jingru Wu
- Department of Thoracic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, China
| | - Bin Jiang
- Department of Thoracic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, China
| | - Mo Shi
- Department of Thoracic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, China
| | - Liang Song
- Department of Thoracic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, China
| | - Zhongwei Xin
- Department of Thoracic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, China
- Medical Science and Technology Innovation Center, Shandong First Medical University, Jinan 250117, China
| |
Collapse
|
4
|
Mitchell JL, Buranapraditkun S, Gantner P, Takata H, Dietze K, N'guessan KF, Pollara J, Nohara J, Muir R, Kroon E, Pinyakorn S, Tulmethakaan N, Manasnayakorn S, Chottanapund S, Thantiworasit P, Prueksakaew P, Ratnaratorn N, Puttamaswin S, Nuntapinit B, Fox L, Haddad EK, Paquin-Proulx D, Phanuphak P, Sacdalan CP, Phanuphak N, Ananworanich J, Hsu D, Vasan S, Ferrari G, Chomont N, Trautmann L. Activation of CXCR3 + Tfh cells and B cells in lymph nodes during acute HIV-1 infection correlates with HIV-specific antibody development. J Virol 2025; 99:e0153224. [PMID: 39932316 PMCID: PMC11915809 DOI: 10.1128/jvi.01532-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 01/17/2025] [Indexed: 03/19/2025] Open
Abstract
Lymph node T follicular helper (Tfh) cells and germinal center (GC) B cells are critical to generate potent antibodies but are rarely possible to study in humans. To understand how Tfh/GC B-cell interactions during acute HIV-1 infection (AHI) impact the generation of HIV-specific antibodies, we performed a unique cross-sectional analysis of inguinal lymph node biopsies taken prior to antiretroviral therapy (ART) initiation in AHI. Although total Tfh and GC B cell frequencies did not change during AHI, increased frequencies of proliferating Th1-like CXCR3+ Tfh, CXCR3+ non-GC B cells, and total CXCR3+ GC B cells correlated with gp120-specific IgG antibody levels in AHI. Frequencies of proliferating CXCR3+ Tfh in AHI also correlated with gp120-specific IgG antibody levels after 48 weeks of ART, antibody-dependent cellular cytotoxicity, antibody-dependent cellular phagocytosis, and increased antibody binding to infected cells after ART. Importantly, while beneficial for antibody development, CXCR3+ Tfh cells were also infected by HIV-1 at higher frequencies than their CXCR3- counterparts and may contribute to the initial dissemination of HIV-1 in follicles. Together, these data suggest that activation of CXCR3+ Tfh cells is associated with induction of the germinal center response and subsequent antibody development, making these cells an important target for future therapeutic interventions. IMPORTANCE Early initiation of antiretroviral therapy (ART) is important to limit the seeding of the long-lasting HIV-1 reservoir; however, it also precludes the development of HIV-specific antibodies that can help control the virus if ART is stopped. Antibody development occurs within germinal centers in the lymph node and requires activation of both antigen-specific B cells and T follicular helper cells (Tfh), a specialized CD4+ cell that provides B cell help. To understand how early ART initiation may prohibit antibody development, we analyzed the frequencies and activation status of Tfh and B cells in lymph node biopsies collected in the different stages of acute HIV-1 infection. Our data suggest that decreased antibody development after early ART initiation may be due to limited germinal center development at the time of treatment and that new interventions that target activation of CXCR3+ Tfh may be beneficial to increase long-term HIV-specific antibody levels.
Collapse
Affiliation(s)
- Julie L. Mitchell
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland, USA
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Supranee Buranapraditkun
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland, USA
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
- Department of Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Center of Excellence in Vaccine Research and Development, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Thai Pediatric Gastroenterology, Hepatology and Immunology (TPGHAI) Research Unit, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Pierre Gantner
- Centre de Recherche du CHUM (CRCHUM) and Department of Microbiology, Infectiology and Immunology, Université de Montréal, Montreal, Québec, Canada
| | - Hiroshi Takata
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland, USA
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Kenneth Dietze
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland, USA
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Kombo F. N'guessan
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland, USA
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Justin Pollara
- Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA
| | - Junsuke Nohara
- Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA
| | - Roshell Muir
- Division of Infectious Diseases & HIV Medicine, Department of Medicine, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA
| | | | - Suteeraporn Pinyakorn
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland, USA
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | | | - Sopark Manasnayakorn
- Department of Surgery, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | | | - Pattarawat Thantiworasit
- Center of Excellence in Vaccine Research and Development, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | | | | | | | - Bessara Nuntapinit
- Armed Forces Research Institute of Medical Sciences in Bangkok, Bangkok, Thailand
| | - Lawrence Fox
- Division of AIDS, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Elias K. Haddad
- Division of Infectious Diseases & HIV Medicine, Department of Medicine, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA
| | - Dominic Paquin-Proulx
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland, USA
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | | | - Carlo P. Sacdalan
- SEARCH Research Foundation, Bangkok, Thailand
- Research Affairs, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | | | - Jintanat Ananworanich
- Department of Global Health, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands
- Amsterdam Institute for Global Health and Development, Amsterdam, the Netherlands
| | - Denise Hsu
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland, USA
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Sandhya Vasan
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland, USA
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Guido Ferrari
- Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA
| | - Nicolas Chomont
- Centre de Recherche du CHUM (CRCHUM) and Department of Microbiology, Infectiology and Immunology, Université de Montréal, Montreal, Québec, Canada
| | - Lydie Trautmann
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland, USA
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - on behalf of RV254 and RV304 Study Groups
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland, USA
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
- Department of Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Center of Excellence in Vaccine Research and Development, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Thai Pediatric Gastroenterology, Hepatology and Immunology (TPGHAI) Research Unit, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Centre de Recherche du CHUM (CRCHUM) and Department of Microbiology, Infectiology and Immunology, Université de Montréal, Montreal, Québec, Canada
- Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA
- Division of Infectious Diseases & HIV Medicine, Department of Medicine, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA
- SEARCH Research Foundation, Bangkok, Thailand
- Department of Surgery, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Armed Forces Research Institute of Medical Sciences in Bangkok, Bangkok, Thailand
- Division of AIDS, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
- Research Affairs, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Institute of HIV Research and Innovation (IHRI), Bangkok, Thailand
- Department of Global Health, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands
- Amsterdam Institute for Global Health and Development, Amsterdam, the Netherlands
| |
Collapse
|
5
|
Yildirim D, Erden A, Fanouriakis A, Koc Kanik H, Babaoglu H, Kardas RC, Kaya B, Vasi I, Duran R, Karadeniz H, Kucuk H, Goker B, Ozturk MA, Tufan A. Serum CXCL-13 levels are associated with active neurological involvement in patients with systemic lupus erythematosus. Lupus 2025; 34:281-291. [PMID: 39918038 DOI: 10.1177/09612033251319405] [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] [Indexed: 03/16/2025]
Abstract
Background: Systemic lupus erythematosus (SLE) is an autoimmune disease with diverse systemic manifestations, including neuropsychiatric involvement (NPSLE), which can vary in severity and prognosis. Diagnosing NPSLE remains challenging, necessitating reliable diagnostic markers. CXCL13, a B-cell chemokine implicated in SLE, has garnered attention for its potential role in NPSLE.Purpose: This study aimed to assess serum CXCL-13 levels in NPSLE patients compared to SLE patients without neuropsychiatric symptoms and healthy controls.Research Design: All study groups were studied CXCL-13 levels from blood samples.Study Sample: One hundred twenty-five participants were categorized into four groups: SLE patients with active NPSLE (n = 6), SLE patients with inactive NPSLE (n = 26), SLE patients without NPSLE (n = 71), and healthy controls (n = 22).Data Collection and Analyses: Serum samples were collected at the time of enrollment and CXCL-13 levels were analysed by Enzyme Linked ImmunoSorbent Assay (ELISA) method.Results: Results indicated significantly elevated CXCL-13 levels in active NPSLE patients compared to other SLE patient groups and healthy controls (p < 0.001 for all). Patients with SLE, including those with inactive NPSLE or no history of NPSLE, had statistically significantly higher serum CXCL-13 levels compared to the control group (p < 0.001). Additionally, serum CXCL-13 levels positively correlated with disease activity assessed by the Systemic Lupus Erythematosus Disease Activity Index (SLEDAI).Conclusion: This study underscores the association between serum CXCL-13 levels and neuropsychiatric involvement in SLE, as well as their correlation with disease activity. Moreover, previous research suggesting a link between CXCL-13 levels and clinical activity in SLE further supports its potential as a diagnostic marker for NPSLE. Nevertheless, further investigations are warranted to validate the utility of CXCL-13 as a diagnostic tool for NPSLE and its role in disease management.
Collapse
Affiliation(s)
- Derya Yildirim
- Department of Rheumatology, Gazi University, Ankara, Turkey
| | | | - Antonis Fanouriakis
- Rheumatology and Clinical Immunology Unit, 4th Department of Internal Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | | | - Hakan Babaoglu
- Clinic of Rheumatology, Ankara City Hospital, Ankara, Turkey
| | | | - Burcugul Kaya
- Department of Rheumatology, Gazi University, Ankara, Turkey
| | - Ibrahim Vasi
- Department of Rheumatology, Gazi University, Ankara, Turkey
| | - Rahime Duran
- Department of Rheumatology, Gazi University, Ankara, Turkey
| | | | - Hamit Kucuk
- Department of Rheumatology, Gazi University, Ankara, Turkey
| | - Berna Goker
- Department of Rheumatology, Gazi University, Ankara, Turkey
| | | | | |
Collapse
|
6
|
Chen Y, Liu J, Ren B, Zhou Z, He Y, Li F, Jin M, Liu L, Wang X, Shen H. Validation of DNA methylation and transcriptional characteristics in CCL5 and CXCL8 genes in autoimmune thyroiditis with varying iodine levels. Sci Rep 2025; 15:6006. [PMID: 39972165 PMCID: PMC11840062 DOI: 10.1038/s41598-025-90499-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Accepted: 02/13/2025] [Indexed: 02/21/2025] Open
Abstract
AIT (autoimmune thyroiditis) is a complex disease influenced by genetic and environmental factors as well as immune dysregulation. Epigenetics has unveiled potential connections among environmental factors, gene expression and thyroid autoimmunity. Among epigenetic modifications, DNA methylation is the first discovered and the most extensively-studied. Investigations both domestically and internationally indicate that iodine supplementation in areas with either excessive or insufficient iodine levels increases the incidence of AIT. Chemokines also play a crucial role in the pathogenesis of AIT. Therefore, does iodine influence the DNA methylation of chemokine genes of patients with AIT, and what are the potential mechanisms involved?. Healthy controls and patients with AIT were matched at the ratio of 1:1 according to age, sex, BMI and residential address, and a total of 176 patients with AIT together with 176 controls were included from regions with varying iodine levels. DNA methylation and mRNA expression levels were analyzed in whole blood using MethylTarget and qRT-PCR methods. At the same time, the GSE138198 and GSE54958 datasets were downloaded from GEO to obtain transcriptional datasets of thyroid tissues from patients with AIT. AIT patients had lower DNA methylation levels in CCL5_2 and CXCL8_1 target regions than controls, while the mRNA expression of CCL5 and CXCL8 genes was significantly higher. A negative correlation was found between the DNA methylation of CCL5_2 and its CpG sites as well as CCL5 gene expression. Higher CCL5 mRNA expression was validated in the thyroid tissues of patients with AIT using GSE datasets. DNA methylation differences at different iodine levels were mainly observed in CCL5_1, CCL5_2, CXCL8_1 and CXCR5_1. CXCL8_1 showed a positive correlation with UIC (urinary iodine concentration). This study demonstrates an association between the DNA methylation status of CCL5 and CXCL8 genes and AIT. The DNA methylation level of the CCL5 gene can serve as an epigenetic marker and biological indicator for AIT. Additionally, long-term iodine deficiency supplementation has a more pronounced impact on the DNA methylation levels of CCL5 and CXCL8 genes.
Collapse
Affiliation(s)
- Yun Chen
- Disorders Control, Centre for Endemic Disease Control, Chinese Centre for Disease Control and Prevention, Harbin Medical University, Harbin City, 150081, Heilongjiang Province, People's Republic of China
- Key Laboratory of Etiology and Epidemiology, National Health Commission & Education Bureau of Heilongjiang Province, Harbin Medical University, Harbin, China
- Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health, Harbin Medical University, Harbin, China
| | - Jinjin Liu
- Disorders Control, Centre for Endemic Disease Control, Chinese Centre for Disease Control and Prevention, Harbin Medical University, Harbin City, 150081, Heilongjiang Province, People's Republic of China
- Key Laboratory of Etiology and Epidemiology, National Health Commission & Education Bureau of Heilongjiang Province, Harbin Medical University, Harbin, China
- Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health, Harbin Medical University, Harbin, China
| | - Bingxuan Ren
- Disorders Control, Centre for Endemic Disease Control, Chinese Centre for Disease Control and Prevention, Harbin Medical University, Harbin City, 150081, Heilongjiang Province, People's Republic of China
- Key Laboratory of Etiology and Epidemiology, National Health Commission & Education Bureau of Heilongjiang Province, Harbin Medical University, Harbin, China
- Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health, Harbin Medical University, Harbin, China
| | - Zheng Zhou
- Disorders Control, Centre for Endemic Disease Control, Chinese Centre for Disease Control and Prevention, Harbin Medical University, Harbin City, 150081, Heilongjiang Province, People's Republic of China
- Key Laboratory of Etiology and Epidemiology, National Health Commission & Education Bureau of Heilongjiang Province, Harbin Medical University, Harbin, China
- Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health, Harbin Medical University, Harbin, China
| | - Yanhong He
- Disorders Control, Centre for Endemic Disease Control, Chinese Centre for Disease Control and Prevention, Harbin Medical University, Harbin City, 150081, Heilongjiang Province, People's Republic of China
- Key Laboratory of Etiology and Epidemiology, National Health Commission & Education Bureau of Heilongjiang Province, Harbin Medical University, Harbin, China
- Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health, Harbin Medical University, Harbin, China
| | - Fan Li
- Control Infection Department, Xi'an First Hospital, Xi'an, People's Republic of China
| | - Meihui Jin
- Disorders Control, Centre for Endemic Disease Control, Chinese Centre for Disease Control and Prevention, Harbin Medical University, Harbin City, 150081, Heilongjiang Province, People's Republic of China
- Key Laboratory of Etiology and Epidemiology, National Health Commission & Education Bureau of Heilongjiang Province, Harbin Medical University, Harbin, China
- Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health, Harbin Medical University, Harbin, China
| | - Lixiang Liu
- Disorders Control, Centre for Endemic Disease Control, Chinese Centre for Disease Control and Prevention, Harbin Medical University, Harbin City, 150081, Heilongjiang Province, People's Republic of China
- Key Laboratory of Etiology and Epidemiology, National Health Commission & Education Bureau of Heilongjiang Province, Harbin Medical University, Harbin, China
- Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health, Harbin Medical University, Harbin, China
| | - Xuebing Wang
- Disorders Control, Centre for Endemic Disease Control, Chinese Centre for Disease Control and Prevention, Harbin Medical University, Harbin City, 150081, Heilongjiang Province, People's Republic of China
- Key Laboratory of Etiology and Epidemiology, National Health Commission & Education Bureau of Heilongjiang Province, Harbin Medical University, Harbin, China
- Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health, Harbin Medical University, Harbin, China
| | - Hongmei Shen
- Disorders Control, Centre for Endemic Disease Control, Chinese Centre for Disease Control and Prevention, Harbin Medical University, Harbin City, 150081, Heilongjiang Province, People's Republic of China.
- Key Laboratory of Etiology and Epidemiology, National Health Commission & Education Bureau of Heilongjiang Province, Harbin Medical University, Harbin, China.
- Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health, Harbin Medical University, Harbin, China.
| |
Collapse
|
7
|
Zhou Y, Hubscher CH. Biomarker expression level changes within rectal gut-associated lymphoid tissues in spinal cord-injured rats. Immunohorizons 2025; 9:vlaf002. [PMID: 40048710 PMCID: PMC11884801 DOI: 10.1093/immhor/vlaf002] [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: 01/31/2025] [Indexed: 03/09/2025] Open
Abstract
Neurogenic bowel dysfunction (NBD) is common after spinal cord injury (SCI). Gut-associated lymphoid tissue (GALT), an organized structure within the mucosal immune system, is important for the maintenance of gut homeostasis and body health and serves as the first line barrier/defense against diet antigens, commensal microbiota, pathogens, and toxins in mucosal areas. The current study examined gene expression levels along six segments of anorectal tissue using real-time polymerase chain reaction (RT-PCR) in uninjured rats (28-day sham surgical controls) and at both 28- and 42-days post-T9 contusion injury. Consistent with our previous report of functional regional differences in the ano-rectum, we demonstrate the existence of GALTs located primarily within the segment at 3-4.5 cm from the rectal dentate line (termed rectal GALTs-rGALTs) in shams with upregulated gene expression levels of multiple biomarkers, including B cell and T cell-related genes, major histocompatibility complex (MHC) class II molecules, and germinal center (GC)-related genes, which was further confirmed by histologic examination. In the same rectal tissue segment following T9 SCI, inflammation-related genes were upregulated at 28 days post-injury (DPI) indicating that microbial infection and inflammation of rGALTs modified structure and function of rGALTs, while at 42 DPI rGALTs exhibited resolution of inflammation and impaired structure/function for extrafollicular B cell responses. Taken together, our data suggest that rGALTs exists in rat rectum for homeostasis of gut microbiota/barrier. SCI induces microbial infection and inflammation in rectal tissues containing rGALTs, which could contribute to development of SCI-related gut microbiome dysbiosis, NBD, and systemic diseases.
Collapse
Affiliation(s)
- Yun Zhou
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, KY, United States
- Kentucky Spinal Cord Injury Research Center, Louisville, KY, United States
| | - Charles H Hubscher
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, KY, United States
- Kentucky Spinal Cord Injury Research Center, Louisville, KY, United States
| |
Collapse
|
8
|
Tolentino M, Pace F, Perantie DC, Mikesell R, Huecker J, Chahin S, Ghezzi L, Piccio L, Cross AH. Cerebrospinal fluid biomarkers as predictors of multiple sclerosis severity. Mult Scler Relat Disord 2025; 94:106268. [PMID: 39832432 DOI: 10.1016/j.msard.2025.106268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 12/16/2024] [Accepted: 01/10/2025] [Indexed: 01/22/2025]
Abstract
BACKGROUND Prognostic biomarkers at multiple sclerosis (MS) onset to predict disease severity may help guide initial therapy selection for people with MS. Over 20 disease-modifying treatments (DMTs) of varying levels of risk and efficacy now exist. The ability to predict MS severity would help to identify those patients at higher risk where a highly effective, but potentially risky, therapy would be optimal. The goal of this project was to determine if cerebrospinal fluid (CSF) soluble markers obtained near time of diagnosis can predict disease severity in people with relapsing remitting MS (RRMS). METHODS We identified 42 RRMS subjects with 4 or more years of clinical follow-up at our center, 8 subjects with other inflammatory neurological diseases (OIND), and 4 subjects with non-inflammatory neurological diseases (NIND) who had donated CSF samples collected for disease diagnosis. This study evaluated soluble CSF biomarkers chosen to reflect neuroinflammation (chemokine ligand 13 - CXCL13), microglia activity (soluble triggering receptor expressed on myeloid cells 2 - sTREM2), demyelination (myelin basic protein -MBP), axon injury and loss (neurofilament light, heavy, and intermediate chains - NFL, NFH, internexin-alpha - INT-α) and neuronal loss (parvalbumin - PVALB) to determine whether any of these CSF factors might predict future MS disease severity. The main outcome measure was MS Severity Score (MSSS), which takes into account disability accumulation (expanded disability status scale - EDSS) and duration of disease. EDSS at last clinical visit was a secondary outcome measure. Univariate and multivariable regression models were used for analysis. Spearman correlations were performed to evaluate correlation between laboratory and clinical variables. RESULTS Forty-two RRMS patients with mean 9.4 years follow-up since lumbar puncture (LP) contributed data. Higher NFH, NFL, and sTREM2 each predicted worse MSSS using both univariate and multivariable regression models. Older age at the time of LP predicted worse MSSS both in the univariate and multivariable models. NFL correlated with NFH, and both were positively correlated with sTREM2 and CXCL13. In the combined OIND and NIND comparator group, NFH correlated with both NFL and CXCL13. CONCLUSION These data support that CSF sTREM2, NFH, and NFL are predictors of MSSS, a measure of MS disease aggressiveness. This study adds to a growing literature implicating microglial activity and axonal injury in MS progression, starting from early stages of the disease.
Collapse
Affiliation(s)
- Miguel Tolentino
- Department of Neurology, School of Medicine, Washington University in St. Louis, 660 South Euclid Avenue, St Louis, MO 63110, USA
| | - Francesca Pace
- Department of Neurology, School of Medicine, Washington University in St. Louis, 660 South Euclid Avenue, St Louis, MO 63110, USA
| | - Dana C Perantie
- Department of Neurology, School of Medicine, Washington University in St. Louis, 660 South Euclid Avenue, St Louis, MO 63110, USA
| | - Robert Mikesell
- Department of Neurology, School of Medicine, Washington University in St. Louis, 660 South Euclid Avenue, St Louis, MO 63110, USA
| | - Julia Huecker
- Center for Biostatistics and Data Science, School of Medicine, Washington University in St. Louis, 660 South Euclid Avenue, St Louis, MO 63110, USA
| | - Salim Chahin
- Department of Neurology, School of Medicine, Washington University in St. Louis, 660 South Euclid Avenue, St Louis, MO 63110, USA
| | - Laura Ghezzi
- Department of Neurology, School of Medicine, Washington University in St. Louis, 660 South Euclid Avenue, St Louis, MO 63110, USA
| | - Laura Piccio
- Department of Neurology, School of Medicine, Washington University in St. Louis, 660 South Euclid Avenue, St Louis, MO 63110, USA
| | - Anne H Cross
- Department of Neurology, School of Medicine, Washington University in St. Louis, 660 South Euclid Avenue, St Louis, MO 63110, USA.
| |
Collapse
|
9
|
Gribonika I, Band VI, Chi L, Perez-Chaparro PJ, Link VM, Ansaldo E, Oguz C, Bousbaine D, Fischbach MA, Belkaid Y. Skin autonomous antibody production regulates host-microbiota interactions. Nature 2025; 638:1043-1053. [PMID: 39662506 PMCID: PMC11864984 DOI: 10.1038/s41586-024-08376-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: 12/10/2023] [Accepted: 11/08/2024] [Indexed: 12/13/2024]
Abstract
The microbiota colonizes each barrier site and broadly controls host physiology1. However, when uncontrolled, microbial colonists can also promote inflammation and induce systemic infection2. The unique strategies used at each barrier tissue to control the coexistence of the host with its microbiota remain largely elusive. Here we uncover that, in the skin, host-microbiota symbiosis depends on the ability of the skin to act as an autonomous lymphoid organ. Notably, an encounter with a new skin commensal promotes two parallel responses, both under the control of Langerhans cells. On one hand, skin commensals induce the formation of classical germinal centres in the lymph node associated with immunoglobulin G1 (IgG1) and IgG3 antibody responses. On the other hand, microbial colonization also leads to the development of tertiary lymphoid organs in the skin that can locally sustain IgG2b and IgG2c responses. These phenomena are supported by the ability of regulatory T cells to convert into T follicular helper cells. Skin autonomous production of antibodies is sufficient to control local microbial biomass, as well as subsequent systemic infection with the same microorganism. Collectively, these results reveal a compartmentalization of humoral responses to the microbiota allowing for control of both microbial symbiosis and potential pathogenesis.
Collapse
Affiliation(s)
- Inta Gribonika
- Metaorganism Immunity Section, Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
| | - Victor I Band
- Metaorganism Immunity Section, Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Liang Chi
- Metaorganism Immunity Section, Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Paula Juliana Perez-Chaparro
- NIAID Microbiome Program, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Verena M Link
- Metaorganism Immunity Section, Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Eduard Ansaldo
- Metaorganism Immunity Section, Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Cihan Oguz
- Integrated Data Sciences Section, Research Technologies Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Djenet Bousbaine
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | | | - Yasmine Belkaid
- Metaorganism Immunity Section, Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
- NIAID Microbiome Program, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
- Metaorganism Immunity Laboratory, Immunology Laboratory, Pasteur Institute, Paris, France.
| |
Collapse
|
10
|
Charmetant X, Rigault G, Chen CC, Kaminski H, Visentin J, Taton B, Marseres G, Mathias V, Koenig A, Barba T, Merville P, Graff-Dubois S, Morelon E, Déchanet-Merville J, Dubois V, Duong van Huyen JP, Couzi L, Thaunat O. γδ T Cells' Role in Donor-Specific Antibody Generation: Insights From Transplant Recipients and Experimental Models. Transpl Int 2025; 38:12859. [PMID: 39944220 PMCID: PMC11815947 DOI: 10.3389/ti.2025.12859] [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: 02/16/2024] [Accepted: 01/15/2025] [Indexed: 05/09/2025]
Abstract
The generation of donor-specific antibodies (DSA) requires that alloreactive B cells receive help from follicular helper T (TFH) cells. Recent works have suggested that γδ T cells could contribute to T cell-dependent humoral responses, leading us to investigate their role in DSA generation. Analysis of a cohort of 331 kidney transplant recipients found no relation between the number of circulating γδ T cells and the risk to develop DSA. Coculture models demonstrated that activated γδ T cells were unable to promote the differentiation of B cells into plasma cells, ruling out that they can be "surrogate" TFH. In line with this, γδ T cells preferentially localized outside the B cell follicles, in the T cell area of lymph nodes, suggesting that they could instead act as "antigen-presenting cell" (APC) to prime αβ TFH. This hypothesis was proven wrong since γδ T cells failed to acquire APC functions in vitro. These findings were validated in vivo by the demonstration that following transplantation with an allogeneic Balb/c (H2d) heart, wild-type and TCRδKO C57BL/6 (H2b) mice developed similar DSA responses, whereas TCRαKO recipients did not develop DSA. We concluded that the generation of DSA is unfazed by the absence of γδ T cells.
Collapse
Affiliation(s)
- Xavier Charmetant
- Centre International de Recherche en Infectiologie, INSERM U1111, Université Claude Bernard Lyon I, CNRS UMR5308, Ecole Normale Supérieure de Lyon, Univ. Lyon, Lyon, France
- Department of Transplantation, Nephrology and Clinical Immunology, Edouard Herriot Hospital, Hospices Civils de Lyon, Lyon, France
- Lyon-Est Faculty of Medicine, Claude Bernard University (Lyon 1), Villeurbanne, France
| | - Guillaume Rigault
- Centre International de Recherche en Infectiologie, INSERM U1111, Université Claude Bernard Lyon I, CNRS UMR5308, Ecole Normale Supérieure de Lyon, Univ. Lyon, Lyon, France
| | - Chien-Chia Chen
- Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan
| | - Hannah Kaminski
- Department of Nephrology, Transplantation, Dialysis and Apheresis, Bordeaux University Hospital, Bordeaux, France
- ImmunoConcEpT, CNRS, Université de Bordeaux, UMR 5164, Bordeaux, France
| | - Jonathan Visentin
- ImmunoConcEpT, CNRS, Université de Bordeaux, UMR 5164, Bordeaux, France
- Laboratory of Immunology et Immunogenetics, Pellegrin Hospital, Bordeaux, France
| | - Benjamin Taton
- Department of Nephrology, Transplantation, Dialysis and Apheresis, Bordeaux University Hospital, Bordeaux, France
| | - Gabriel Marseres
- ImmunoConcEpT, CNRS, Université de Bordeaux, UMR 5164, Bordeaux, France
| | - Virginie Mathias
- French National Blood Service (EFS), HLA Laboratory, Décines, France
| | - Alice Koenig
- Centre International de Recherche en Infectiologie, INSERM U1111, Université Claude Bernard Lyon I, CNRS UMR5308, Ecole Normale Supérieure de Lyon, Univ. Lyon, Lyon, France
- Department of Transplantation, Nephrology and Clinical Immunology, Edouard Herriot Hospital, Hospices Civils de Lyon, Lyon, France
- Lyon-Est Faculty of Medicine, Claude Bernard University (Lyon 1), Villeurbanne, France
| | - Thomas Barba
- Centre International de Recherche en Infectiologie, INSERM U1111, Université Claude Bernard Lyon I, CNRS UMR5308, Ecole Normale Supérieure de Lyon, Univ. Lyon, Lyon, France
- Department of Transplantation, Nephrology and Clinical Immunology, Edouard Herriot Hospital, Hospices Civils de Lyon, Lyon, France
- Department of Internal Medicine, Edouard Herriot Hospital, Hospices Civils de Lyon, Lyon, France
| | - Pierre Merville
- Department of Nephrology, Transplantation, Dialysis and Apheresis, Bordeaux University Hospital, Bordeaux, France
- ImmunoConcEpT, CNRS, Université de Bordeaux, UMR 5164, Bordeaux, France
| | - Stéphanie Graff-Dubois
- Sorbonne Université, INSERM, UMRS 959, Immunology-Immunopathology-Immunotherapy (i3), Paris, France
| | - Emmanuel Morelon
- Centre International de Recherche en Infectiologie, INSERM U1111, Université Claude Bernard Lyon I, CNRS UMR5308, Ecole Normale Supérieure de Lyon, Univ. Lyon, Lyon, France
- Department of Transplantation, Nephrology and Clinical Immunology, Edouard Herriot Hospital, Hospices Civils de Lyon, Lyon, France
- Lyon-Est Faculty of Medicine, Claude Bernard University (Lyon 1), Villeurbanne, France
| | | | - Valérie Dubois
- French National Blood Service (EFS), HLA Laboratory, Décines, France
| | | | - Lionel Couzi
- Department of Nephrology, Transplantation, Dialysis and Apheresis, Bordeaux University Hospital, Bordeaux, France
- ImmunoConcEpT, CNRS, Université de Bordeaux, UMR 5164, Bordeaux, France
| | - Olivier Thaunat
- Centre International de Recherche en Infectiologie, INSERM U1111, Université Claude Bernard Lyon I, CNRS UMR5308, Ecole Normale Supérieure de Lyon, Univ. Lyon, Lyon, France
- Department of Transplantation, Nephrology and Clinical Immunology, Edouard Herriot Hospital, Hospices Civils de Lyon, Lyon, France
- Lyon-Est Faculty of Medicine, Claude Bernard University (Lyon 1), Villeurbanne, France
| |
Collapse
|
11
|
Han D, Jeong BK, Hong JM, Seo JH, Lee G, Kim K, Hong C, Lee H, Cha SM, Kim JH, Park T, Gong G, Lee HJ. Optimal chemokine receptors for enhancing immune cell trafficking in adoptive cell therapy. Immunol Res 2025; 73:36. [PMID: 39815137 DOI: 10.1007/s12026-024-09560-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: 09/30/2024] [Accepted: 11/13/2024] [Indexed: 01/18/2025]
Abstract
Recently, a strategy involving the engineering of chemokine receptors on immune cells was developed to optimize adoptive cell therapy (ACT) for solid tumors. Given the variability in chemokine secretion among different tumor types, identifying and modulating the appropriate chemokine receptors is crucial. In this study, we utilized extensive RNA sequencing data from both tumor tissues from The Cancer Genome Atlas and normal tissues from Genotype-Tissue Expression to investigate the expression profiles of chemokines. Through analysis, we identified eight chemokine receptors associated with increased chemokine levels in tumor tissues compared to normal tissues, making them promising candidates for enhancing ACT. Subsequent examination of tumor-infiltrating lymphocytes and chimeric antigen receptor-T cells revealed that five out of the eight candidate chemokine receptors did not exhibit elevated expression in T cells. Among five candidates, we demonstrated that CXCR5 is a particularly promising candidate for enhancing cell migration without compromising cell viability or cytotoxicity. In conclusion, our study underscores the potential of five chemokine receptors (CCR6, CCR9, CXCR1, CXCR5, and XCR1) as valuable targets for modulating ACT to enhance cell trafficking and potentially improve cancer therapy outcomes.
Collapse
Affiliation(s)
- DoYeon Han
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-Ro, 43-Gil, Songpa-Gu, Seoul, 05505, Korea
- Biomedical Sciences, Asan Medical Institute of Convergence Science and Technology, University of Ulsan College of Medicine, Seoul, South Korea
| | - Byung-Kwan Jeong
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-Ro, 43-Gil, Songpa-Gu, Seoul, 05505, Korea
| | - Jong Moo Hong
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-Ro, 43-Gil, Songpa-Gu, Seoul, 05505, Korea
- Biomedical Sciences, Asan Medical Institute of Convergence Science and Technology, University of Ulsan College of Medicine, Seoul, South Korea
| | - Jeong-Han Seo
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-Ro, 43-Gil, Songpa-Gu, Seoul, 05505, Korea
- NeogenTC Corp, Seoul, Korea
- Biomedical Sciences, Asan Medical Institute of Convergence Science and Technology, University of Ulsan College of Medicine, Seoul, South Korea
| | - GunHee Lee
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-Ro, 43-Gil, Songpa-Gu, Seoul, 05505, Korea
| | | | | | | | - Su Min Cha
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-Ro, 43-Gil, Songpa-Gu, Seoul, 05505, Korea
- Biomedical Sciences, Asan Medical Institute of Convergence Science and Technology, University of Ulsan College of Medicine, Seoul, South Korea
| | - Jong Hyeok Kim
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-Ro, 43-Gil, Songpa-Gu, Seoul, 05505, Korea
- Biomedical Sciences, Asan Medical Institute of Convergence Science and Technology, University of Ulsan College of Medicine, Seoul, South Korea
| | - Taehyun Park
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-Ro, 43-Gil, Songpa-Gu, Seoul, 05505, Korea
- Biomedical Sciences, Asan Medical Institute of Convergence Science and Technology, University of Ulsan College of Medicine, Seoul, South Korea
| | - Gyungyub Gong
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-Ro, 43-Gil, Songpa-Gu, Seoul, 05505, Korea.
| | - Hee Jin Lee
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-Ro, 43-Gil, Songpa-Gu, Seoul, 05505, Korea.
- NeogenTC Corp, Seoul, Korea.
| |
Collapse
|
12
|
Pachner AR, Pike S, Smith AD, Gilli F. CXCL13 as a Biomarker: Background and Utility in Multiple Sclerosis. Biomolecules 2024; 14:1541. [PMID: 39766248 PMCID: PMC11673926 DOI: 10.3390/biom14121541] [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: 11/03/2024] [Revised: 11/25/2024] [Accepted: 11/29/2024] [Indexed: 01/11/2025] Open
Abstract
CXCL13 is a chemokine which is upregulated within the CNS in multiple sclerosis, Lyme neuroborreliosis, and other inflammatory diseases and is increasingly clinically useful as a biomarker. This review provides background for understanding its function in the immune system and its relationship to ectopic lymphoid follicles. Also reviewed are its utility in multiple sclerosis and Lyme neuroborreliosis and potential problems in its measurement. CXCL13 has the potential to be an exceptionally useful biomarker in a range of inflammatory diseases.
Collapse
Affiliation(s)
- Andrew R. Pachner
- Geisel School of Medicine at Dartmouth, Dartmouth-Hitchcock Medical Center, Lebanon, NH 03756, USA (A.D.S.); (F.G.)
| | | | | | | |
Collapse
|
13
|
Barakat R, Chatterjee J, Mu R, Qi X, Gu X, Smirnov I, Cobb O, Gao K, Barnes A, Kipnis J, Gutmann DH. Human single cell RNA-sequencing reveals a targetable CD8 + exhausted T cell population that maintains mouse low-grade glioma growth. Nat Commun 2024; 15:10312. [PMID: 39609412 PMCID: PMC11605098 DOI: 10.1038/s41467-024-54569-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 11/14/2024] [Indexed: 11/30/2024] Open
Abstract
In solid cancers, T cells typically function as cytotoxic effectors to limit tumor growth, prompting therapies that capitalize upon this antineoplastic property (immune checkpoint inhibition; ICI). Unfortunately, ICI treatments have been largely ineffective for high-grade brain tumors (gliomas; HGGs). Leveraging several single-cell RNA sequencing datasets, we report greater CD8+ exhausted T cells in human pediatric low-grade gliomas (LGGs) relative to adult and pediatric HGGs. Using several preclinical mouse LGG models (Nf1-OPG mice), we show that these PD1+/TIGIT+ CD8+ exhausted T cells are restricted to the tumor tissue, where they express paracrine factors necessary for OPG growth. Importantly, ICI treatments with α-PD1 and α-TIGIT antibodies attenuate Nf1-OPG tumor proliferation through suppression of two cytokine (Ccl4 and TGFβ)-mediated mechanisms, rather than by T cell-mediated cytotoxicity, as well as suppress monocyte-controlled T cell chemotaxis. Collectively, these findings establish a previously unrecognized function for CD8+ exhausted T cells as specialized regulators of LGG maintenance.
Collapse
Affiliation(s)
- Rasha Barakat
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Jit Chatterjee
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Rui Mu
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Xuanhe Qi
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Xingxing Gu
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Igor Smirnov
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Olivia Cobb
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Karen Gao
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Angelica Barnes
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Jonathan Kipnis
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - David H Gutmann
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, 63110, USA.
| |
Collapse
|
14
|
Gu X, Li D, Wu P, Zhang C, Cui X, Shang D, Ma R, Liu J, Sun N, He J. Revisiting the CXCL13/CXCR5 axis in the tumor microenvironment in the era of single-cell omics: Implications for immunotherapy. Cancer Lett 2024; 605:217278. [PMID: 39332588 DOI: 10.1016/j.canlet.2024.217278] [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: 07/15/2024] [Revised: 09/22/2024] [Accepted: 09/23/2024] [Indexed: 09/29/2024]
Abstract
As one of the important members of the family of chemokines and their receptors, the CXCL13/CXCR5 axis is involved in follicle formation in normal lymphoid tissues and the establishment of somatic cavity immunity under physiological conditions, as well as being associated with a wide range of infectious, autoimmune, and tumoral diseases. Here in this review, we focus on its role in tumors. Traditional studies have found the axis to be both pro- and anti-tumorigenic, involving a variety of immune cells, including the tumor cells themselves and those in the tumor microenvironment (TME), and the prognostic significance of this axis is clinical context-dependent. With the development of techniques at the single-cell level, we were able to explain in detail the status of the CXCL13/CXCR5 axis in the TME based on real clinical samples and found that it involves a range of crucial intrinsic anti-tumor immune processes in the TME and is therefore important in tumor immunotherapy. We summarize the cellular subsets, physiological functions, and prognostic significance associated with this axis in the most promising immune checkpoint inhibitor (ICI) therapies of the day and summarize possible therapeutic ideas based on this axis. As with any TME study, the most important takeaway is that the complexity of the CXCL13/CXCR5 axis in TME suggests the importance of personalized therapy in tumor therapy.
Collapse
Affiliation(s)
- Xuanyu Gu
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China; 4+4 Medical Doctor Program, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Dongyu Li
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China; 4+4 Medical Doctor Program, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Peng Wu
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Chaoqi Zhang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Xinyu Cui
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China; 4+4 Medical Doctor Program, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Dexin Shang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China; 4+4 Medical Doctor Program, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Ruijie Ma
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Jingjing Liu
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Nan Sun
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Jie He
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
15
|
Sun G, Liu Y. Tertiary lymphoid structures in ovarian cancer. Front Immunol 2024; 15:1465516. [PMID: 39569184 PMCID: PMC11576424 DOI: 10.3389/fimmu.2024.1465516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 10/22/2024] [Indexed: 11/22/2024] Open
Abstract
Ovarian cancer (OC) is a significant cause of cancer-related mortality in women worldwide. Despite advances in treatment modalities, including surgery and chemotherapy, the overall prognosis for OC patients remains poor, particularly for patients with advanced or recurrent disease. Immunotherapy, particularly immune checkpoint blockade (ICB), has revolutionized cancer treatment in various malignancies but has shown limited efficacy in treating OC, which is primarily attributed to the immunologically. Tertiary lymphoid structures (TLSs), which are ectopic aggregates of immune cells, have emerged as potential mediators of antitumor immunity. This review explores the composition, formation, and induction of tumor associated TLS (TA-TLS) in OC, along with their role and therapeutic implications in disease development and treatment. By elucidating the roles TA-TLSs and their cellular compositions played in OC microenvironment, novel therapeutic targets may be identified to overcome immune suppression and enhance immunotherapy efficacy in ovarian cancer.
Collapse
Affiliation(s)
- Guojuan Sun
- The Ward Section of Home Overseas Doctors, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yi Liu
- Department of Gynaecology and Obstetrics, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
16
|
Zheng K, Chen M, Xu X, Li P, Yin C, Wang J, Liu B. Chemokine CXCL13-CXCR5 signaling in neuroinflammation and pathogenesis of chronic pain and neurological diseases. Cell Mol Biol Lett 2024; 29:134. [PMID: 39472796 PMCID: PMC11523778 DOI: 10.1186/s11658-024-00653-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 10/16/2024] [Indexed: 11/02/2024] Open
Abstract
Chronic pain dramatically affects life qualities of the sufferers. It has posed a heavy burden to both patients and the health care system. However, the current treatments for chronic pain are usually insufficient and cause many unwanted side effects. Chemokine C-X-C motif ligand 13 (CXCL13), formerly recognized as a B cell chemokine, binds with the cognate receptor CXCR5, a G-protein-coupled receptor (GPCR), to participate in immune cell recruitments and immune modulations. Recent studies further demonstrated that CXCL13-CXCR5 signaling is implicated in chronic pain via promoting neuroimmune interaction and neuroinflammation in the sensory system. In addition, some latest work also pointed out the involvement of CXCL13-CXCR5 in the pathogenesis of certain neurological diseases, including ischemic stroke and amyotrophic lateral sclerosis. Therefore, we aim to outline the recent findings in regard to the involvement of CXCL13-CXCR5 signaling in chronic pain as well as certain neurological diseases, with the focus on how this chemokine signaling contributes to the pathogenesis of these neurological diseases via regulating neuroimmune interaction and neuroinflammation. Strategies that can specifically target CXCL13-CXCR5 signaling in distinct locations may provide new therapeutic options for these neurological diseases.
Collapse
Affiliation(s)
- Kaige Zheng
- Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Zhejiang Chinese Medical University, Hangzhou, China
| | - Muyan Chen
- Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xingjianyuan Xu
- Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Zhejiang Chinese Medical University, Hangzhou, China
| | - Peiyi Li
- Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Zhejiang Chinese Medical University, Hangzhou, China
| | - Chengyu Yin
- Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jie Wang
- Department of Rehabilitation in Traditional Chinese Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China.
| | - Boyi Liu
- Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Zhejiang Chinese Medical University, Hangzhou, China.
| |
Collapse
|
17
|
Sun B, Gan C, Tang Y, Xu Q, Wang K, Zhu F. Identification and validation of a prognostic model based on three TLS-Related genes in oral squamous cell carcinoma. Cancer Cell Int 2024; 24:350. [PMID: 39462422 PMCID: PMC11515094 DOI: 10.1186/s12935-024-03543-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Accepted: 10/21/2024] [Indexed: 10/29/2024] Open
Abstract
BACKGROUND The tertiary lymphoid structures (TLSs) have an immunomodulatory function and have a positive impact on the survival outcomes of patients with oral squamous cell carcinoma (OSCC). However, there is a lack of standard approaches for quantifying TLSs and prognostic models using TLS-related genes (TLSRGs). These limitations limit the widespread use of TLSs in clinical practice. METHODS A convolutional neural network was used to automatically detect and quantify TLSs in HE-stained whole slide images. By employing bioinformatics and diverse statistical methods, this research created a prognostic model using TCGA cohorts and explored the connection between this model and immune infiltration. The expression levels of three TLSRGs in clinical specimens were detected by immunohistochemistry. To facilitate the assessment of individual prognostic outcomes, we further constructed a nomogram based on the risk score and other clinical factors. RESULTS TLSs were found to be an independent predictor of both overall survival (OS) and disease-free survival in OSCC patients. A larger proportion of the TLS area represented a better prognosis. After analysis, we identified 69 differentially expressed TLSRGs and selected three pivotal TLSRGs to construct the risk score model. This model emerged as a standalone predictor for OS and exhibited close associations with CD4 + T cells, CD8 + T cells, and macrophages. Immunohistochemistry revealed high expression levels of CCR7 and CXCR5 in TLS + OSCC samples, while CD86 was highly expressed in TLS- OSCC samples. The nomogram demonstrates excellent predictive ability for overall survival in OSCC patients. CONCLUSIONS This is the first prognostic nomogram based on TLSRGs, that can effectively predict survival outcomes and contribute to individual treatment strategies for OSCC patients.
Collapse
Affiliation(s)
- Bincan Sun
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya Hospital, Central South University, Changsha, Hunan, P. R. China
- Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Changsha, Hunan, P. R. China
| | - Chengwen Gan
- Department of Oral Maxillofacial Surgery, Hainan General Hospital, Haikou, Hainan, P. R. China
| | - Yan Tang
- Department of Nursing, The Second Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, P. R. China
| | - Qian Xu
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya Hospital, Central South University, Changsha, Hunan, P. R. China
- Research Center of Oral and Maxillofacial Tumor, Xiangya Hospital, Central South University, Changsha, Hunan, P. R. China
- Institute of Oral Cancer and Precancerous Lesions, Central South University, Changsha, Hunan, P. R. China
| | - Kai Wang
- Department of Oral and Maxillofacial Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan, P. R. China
| | - Feiya Zhu
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya Hospital, Central South University, Changsha, Hunan, P. R. China.
- Research Center of Oral and Maxillofacial Tumor, Xiangya Hospital, Central South University, Changsha, Hunan, P. R. China.
- Institute of Oral Cancer and Precancerous Lesions, Central South University, Changsha, Hunan, P. R. China.
- National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Peking University School and Hospital of Stomatology, Beijing, P. R. China.
| |
Collapse
|
18
|
Hui L, Li Y, Huang MK, Jiang YM, Liu T. CXCL13: a common target for immune-mediated inflammatory diseases. Clin Exp Med 2024; 24:244. [PMID: 39443356 PMCID: PMC11499446 DOI: 10.1007/s10238-024-01508-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 10/16/2024] [Indexed: 10/25/2024]
Abstract
CXCL13 is a chemokine that plays an important role in the regulation and development of secondary lymphoid organs. CXCL13 is also involved in the regulation of pathological processes, particularly inflammatory responses, of many diseases. The function of CXCL13 varies depending on the condition of the host. In a healthy condition, CXCL13 is mainly secreted by mouse stromal cells or human follicular helper T cells, whereas in diseases conditions, they are produced by human peripheral helper T cells and macrophages in non-lymphoid tissues; this is termed ectopic expression of CXCL13. Ectopic CXCL13 expression is involved in the pathogenesis of various immune-mediated inflammatory diseases as it regulates the migration of B lymphocytes, T lymphocytes, and other immune cells in inflammatory sites as well as influences the expression of inflammatory factors. Additionally, ectopic expression of CXCL13 plays a key role in ectopic lymphoid organ formation. In this review, we focused on the sources of CXCL13 in different conditions and its regulatory mechanisms in immune-mediated inflammatory diseases, providing novel ideas for further research on targeting CXCL13 for the treatment of immune-mediated inflammatory diseases.
Collapse
Affiliation(s)
- Lu Hui
- Department of Laboratory Medicine, West China Second University Hospital, and Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, Sichuan University, No. 20, Section 3, Renmin Road South, Chengdu, 610041, Sichuan, People's Republic of China
| | - Ye Li
- Department of Laboratory Medicine, West China Second University Hospital, and Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, Sichuan University, No. 20, Section 3, Renmin Road South, Chengdu, 610041, Sichuan, People's Republic of China
| | - Meng-Ke Huang
- Department of Laboratory Medicine, West China Second University Hospital, and Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, Sichuan University, No. 20, Section 3, Renmin Road South, Chengdu, 610041, Sichuan, People's Republic of China
| | - Yong-Mei Jiang
- Department of Laboratory Medicine, West China Second University Hospital, and Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, Sichuan University, No. 20, Section 3, Renmin Road South, Chengdu, 610041, Sichuan, People's Republic of China.
| | - Ting Liu
- Department of Laboratory Medicine, West China Second University Hospital, and Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, Sichuan University, No. 20, Section 3, Renmin Road South, Chengdu, 610041, Sichuan, People's Republic of China.
- State Key Laboratory of Biotherapy and Cancer Center/National Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, People's Republic of China.
| |
Collapse
|
19
|
Wang F, Guo B, Jia Z, Jing Z, Wang Q, Li M, Lu B, Liang W, Hu W, Fu X. The Role of CXCR3 in Nervous System-Related Diseases. Mediators Inflamm 2024; 2024:8347647. [PMID: 39429695 PMCID: PMC11488998 DOI: 10.1155/2024/8347647] [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: 01/18/2024] [Revised: 09/12/2024] [Accepted: 09/19/2024] [Indexed: 10/22/2024] Open
Abstract
Inflammatory chemokines are a group of G-protein receptor ligands characterized by conserved cysteine residues, which can be divided into four main subfamilies: CC, CXC, XC, and CX3C. The C-X-C chemokine receptor (CXCR) 3 and its ligands, C-X-C chemokine ligands (CXCLs), are widely expressed in both the peripheral nervous system (PNS) and central nervous system (CNS). This comprehensive literature review aims to examine the functions and pathways of CXCR3 and its ligands in nervous system-related diseases. In summary, while the related pathways and the expression levels of CXCR3 and its ligands are varied among different cells in PNS and CNS, the MPAK pathway is the core via which CXCR3 exerts physiological functions. It is not only the core pathway of CXCR3 after activation but also participates in the expression of CXCR3 ligands in the nervous system. In addition, despite CXCR3 being a common inflammatory chemokine receptor, there is no consensus on its precise roles in various diseases. This uncertainty may be attributable to distinct inflammatory characteristics, that inflammation simultaneously possesses the dual properties of damage induction and repair facilitation.
Collapse
Affiliation(s)
- Fangyuan Wang
- Department of Neurosurgery, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Bing Guo
- Department of Neurosurgery, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Ziyang Jia
- Department of Neurosurgery, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Zhou Jing
- Department of Neurosurgery, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Qingyi Wang
- Department of Neurosurgery, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Minghe Li
- Department of Neurosurgery, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Bingqi Lu
- Department of Neurosurgery, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Wulong Liang
- Department of Neurosurgery, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Weihua Hu
- Department of Neurosurgery, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Xudong Fu
- Department of Neurosurgery, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
20
|
Liu Y, Yao Y, Yang X, Wei M, Lu B, Dong K, Lyu D, Li Y, Guan W, Huang R, Xu G, Pan X. Lymphocyte activation gene 3 served as a potential prognostic and immunological biomarker across various cancer types: a clinical and pan-cancer analysis. Clin Transl Immunology 2024; 13:e70009. [PMID: 39372371 PMCID: PMC11450455 DOI: 10.1002/cti2.70009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Revised: 08/29/2024] [Accepted: 09/19/2024] [Indexed: 10/08/2024] Open
Abstract
Objectives Lymphocyte activation gene 3 (LAG3), an inhibitory receptor in T-cell activation, is a negative prognostic factor. However, its impact on tumours has yet to be comprehensively elucidated on a pan-cancer scale. Thus, we aim to reveal its role at the pan-cancer level. Methods We performed IHC staining on a retrospective cohort of 370 patients. Then we assessed the prognostic effect of LAG3 using Kaplan-Meier survival analysis and multivariate Cox regression analysis. In pan-cancer analysis, we constructed competing endogenous RNA and protein-protein interaction networks, conducted gene set enrichment analysis and identified correlations between LAG3 gene expression and various factors, including clinical characteristics, tumour purity, mutations, tumour immunity and drug sensitivity across 33 cancer types. Results LAG3 was expressed higher in normal kidney tissues than in tumours. A high level of LAG3 gene expression was an independent prognostic factor for OS (HR = 6.60, 95% CI = 2.43-17.90, P < 0.001) and PFS (HR = 3.44, 95% CI = 1.68-7.10, P < 0.001). In pan-cancer analysis, LAG3 exhibited robust correlations with survival and tumour stages in various cancers. Moreover, LAG3 was strongly associated with immune-related genes, proteins and signalling pathways. LAG3 gene expression was positively associated with increased infiltration of activated immune cells and decreased infiltration of several resting cells. LAG3 gene expression was associated with tumour mutation burden and microsatellite instability in multiple cancers. Conclusion High LAG3 gene expression was an independent risk factor in kidney neoplasms. It also functioned as a biomarker for prognosis, TIME and immunotherapy efficacy in the pan-cancer dimension.
Collapse
Affiliation(s)
- Yifan Liu
- Department of UrologyXinhua Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Yuntao Yao
- Department of UrologyXinhua Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Xinyue Yang
- Department of UrologyXinhua Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Maodong Wei
- Department of UrologyXinhua Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Bingnan Lu
- Department of UrologyXinhua Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Keqing Dong
- Department of UrologyXinhua Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Donghao Lyu
- Department of UrologyXinhua Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Yuanan Li
- Department of UrologyXinhua Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Wenbin Guan
- Department of PathologyXinhua Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Runzhi Huang
- Department of Burn SurgeryThe First Affiliated Hospital of Naval Medical UniversityShanghaiChina
| | - Guofeng Xu
- Department of UrologyXinhua Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Xiuwu Pan
- Department of UrologyXinhua Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
| |
Collapse
|
21
|
Niu L, Chen T, Yang A, Yan X, Jin F, Zheng A, Song X. Macrophages and tertiary lymphoid structures as indicators of prognosis and therapeutic response in cancer patients. Biochim Biophys Acta Rev Cancer 2024; 1879:189125. [PMID: 38851437 DOI: 10.1016/j.bbcan.2024.189125] [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: 02/20/2024] [Revised: 05/24/2024] [Accepted: 06/02/2024] [Indexed: 06/10/2024]
Abstract
Tertiary lymphoid structures (TLS) can reflect cancer prognosis and clinical outcomes in various tumour tissues. Tumour-associated macrophages (TAMs) are indispensable components of the tumour microenvironment and play crucial roles in tumour development and immunotherapy. TAMs are associated with TLS induction via the modulation of the T cell response, which is a major component of the TLS. Despite their important roles in cancer immunology, the subtypes of TAMs that influence TLS and their correlation with prognosis are not completely understood. Here, we provide novel insights into the role of TAMs in regulating TLS formation. Furthermore, we discuss the prognostic value of these TAM subtypes and TLS, as well as the current antitumour therapies for inducing TLS. This study highlights an entirely new field of TLS regulation that may lead to the development of an innovative perspective on immunotherapy for cancer treatment.
Collapse
Affiliation(s)
- Li Niu
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China
| | - Ting Chen
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Aodan Yang
- Department of Breast Surgery, the First Hospital of China Medical University, Shenyang, China
| | - Xiwen Yan
- Department of Breast Surgery, the First Hospital of China Medical University, Shenyang, China
| | - Feng Jin
- Department of Breast Surgery, the First Hospital of China Medical University, Shenyang, China
| | - Ang Zheng
- Department of Breast Surgery, the First Hospital of China Medical University, Shenyang, China.
| | - Xinyue Song
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China.
| |
Collapse
|
22
|
Cinti I, Vezyrgianni K, Denton AE. Unravelling the contribution of lymph node fibroblasts to vaccine responses. Adv Immunol 2024; 164:1-37. [PMID: 39523027 DOI: 10.1016/bs.ai.2024.07.001] [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] [Indexed: 11/16/2024]
Abstract
Vaccination is one of the most effective medical interventions, saving millions of lives and reducing the morbidity of infections across the lifespan, from infancy to older age. The generation of plasma cells and memory B cells that produce high affinity class switched antibodies is central to this protection, and these cells are the ultimate output of the germinal centre response. Optimal germinal centre responses require different immune cells to interact with one another in the right place and at the right time and this delicate cellular ballet is coordinated by a network of interconnected stromal cells. In this review we will discuss the various types of lymphoid stromal cells and how they coordinate immune cell homeostasis, the induction and maintenance of the germinal centre response, and how this is disorganised in older bodies.
Collapse
Affiliation(s)
- Isabella Cinti
- Department of Immunology and Inflammation, Imperial College London, London, United Kingdom
| | - Kassandra Vezyrgianni
- Department of Immunology and Inflammation, Imperial College London, London, United Kingdom
| | - Alice E Denton
- Department of Immunology and Inflammation, Imperial College London, London, United Kingdom.
| |
Collapse
|
23
|
Uno M, Bono H. Transcriptional Signatures of Domestication Revealed through Meta-Analysis of Pig, Chicken, Wild Boar, and Red Junglefowl Gene Expression Data. Animals (Basel) 2024; 14:1998. [PMID: 38998110 PMCID: PMC11240496 DOI: 10.3390/ani14131998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 06/25/2024] [Accepted: 07/04/2024] [Indexed: 07/14/2024] Open
Abstract
Domesticated animals have undergone significant changes in their behavior, morphology, and physiological functions during domestication. To identify the changes in gene expression associated with domestication, we collected the RNA-seq data of pigs, chickens, wild boars, and red junglefowl from public databases and performed a meta-analysis. Gene expression was quantified, and the expression ratio between domesticated animals and their wild ancestors (DW-ratio) was calculated. Genes were classified as "upregulated", "downregulated", or "unchanged" based on their DW-ratio, and the DW-score was calculated for each gene. Gene set enrichment analysis revealed that genes upregulated in pigs were related to defense from viral infection, whereas those upregulated in chickens were associated with aminoglycan and carbohydrate derivative catabolic processes. Genes commonly upregulated in pigs and chickens are involved in the immune response, olfactory learning, epigenetic regulation, cell division, and extracellular matrix. In contrast, genes upregulated in wild boar and red junglefowl are related to stress response, cell proliferation, cardiovascular function, neural regulation, and energy metabolism. These findings provide valuable insights into the genetic basis of the domestication process and highlight potential candidate genes for breeding applications.
Collapse
Affiliation(s)
- Motoki Uno
- Graduate School of Integrated Sciences for Life, Hiroshima University, 3-10-23 Kagamiyama, Higashi-Hiroshima 739-0046, Japan
| | - Hidemasa Bono
- Graduate School of Integrated Sciences for Life, Hiroshima University, 3-10-23 Kagamiyama, Higashi-Hiroshima 739-0046, Japan
- Genome Editing Innovation Center, Hiroshima University, 3-10-23 Kagamiyama, Higashi-Hiroshima 739-0046, Japan
| |
Collapse
|
24
|
Law C, Wacleche VS, Cao Y, Pillai A, Sowerby J, Hancock B, Horisberger A, Bracero S, Skidanova V, Li Z, Adejoorin I, Dillon E, Benque IJ, Nunez DP, Simmons DP, Keegan J, Chen L, Baker T, Brohawn PZ, Al-Mossawi H, Hao LY, Jones B, Rao N, Qu Y, Alves SE, Jonsson AH, Shaw KS, Vleugels RA, Massarotti E, Costenbader KH, Brenner MB, Lederer JA, Hultquist JF, Choi J, Rao DA. Interferon subverts an AHR-JUN axis to promote CXCL13 + T cells in lupus. Nature 2024; 631:857-866. [PMID: 38987586 PMCID: PMC11628166 DOI: 10.1038/s41586-024-07627-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 05/30/2024] [Indexed: 07/12/2024]
Abstract
Systemic lupus erythematosus (SLE) is prototypical autoimmune disease driven by pathological T cell-B cell interactions1,2. Expansion of T follicular helper (TFH) and T peripheral helper (TPH) cells, two T cell populations that provide help to B cells, is a prominent feature of SLE3,4. Human TFH and TPH cells characteristically produce high levels of the B cell chemoattractant CXCL13 (refs. 5,6), yet regulation of T cell CXCL13 production and the relationship between CXCL13+ T cells and other T cell states remains unclear. Here, we identify an imbalance in CD4+ T cell phenotypes in patients with SLE, with expansion of PD-1+/ICOS+ CXCL13+ T cells and reduction of CD96hi IL-22+ T cells. Using CRISPR screens, we identify the aryl hydrocarbon receptor (AHR) as a potent negative regulator of CXCL13 production by human CD4+ T cells. Transcriptomic, epigenetic and functional studies demonstrate that AHR coordinates with AP-1 family member JUN to prevent CXCL13+ TPH/TFH cell differentiation and promote an IL-22+ phenotype. Type I interferon, a pathogenic driver of SLE7, opposes AHR and JUN to promote T cell production of CXCL13. These results place CXCL13+ TPH/TFH cells on a polarization axis opposite from T helper 22 (TH22) cells and reveal AHR, JUN and interferon as key regulators of these divergent T cell states.
Collapse
Affiliation(s)
- Calvin Law
- Department of Biochemistry and Molecular Genetics, The Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Department of Dermatology, The Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Center of Human Immunobiology, The Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Center of Synthetic Biology, Northwestern University, Evanston, IL, USA
- Center for Genetic Medicine, Northwestern University, Chicago, IL, USA
| | - Vanessa Sue Wacleche
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Ye Cao
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Arundhati Pillai
- Department of Biochemistry and Molecular Genetics, The Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Department of Dermatology, The Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Center of Human Immunobiology, The Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Center of Synthetic Biology, Northwestern University, Evanston, IL, USA
- Center for Genetic Medicine, Northwestern University, Chicago, IL, USA
| | - John Sowerby
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Brandon Hancock
- Department of Biochemistry and Molecular Genetics, The Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Department of Dermatology, The Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Center of Human Immunobiology, The Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Center of Synthetic Biology, Northwestern University, Evanston, IL, USA
- Center for Genetic Medicine, Northwestern University, Chicago, IL, USA
| | - Alice Horisberger
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Sabrina Bracero
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Viktoriya Skidanova
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Zhihan Li
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Ifeoluwakiisi Adejoorin
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Eilish Dillon
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Isaac J Benque
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Diana Pena Nunez
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Daimon P Simmons
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Joshua Keegan
- Department of Surgery, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Lin Chen
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | | | | | | | - Ling-Yang Hao
- Discovery Immunology, Janssen Research & Development, Spring House, PA, USA
| | - Brian Jones
- Discovery Immunology, Janssen Research & Development, Spring House, PA, USA
| | - Navin Rao
- Discovery Immunology, Janssen Research & Development, Spring House, PA, USA
| | - Yujie Qu
- Merck & Co., Inc., Rahway, NJ, USA
| | | | - A Helena Jonsson
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Division of Rheumatology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Katharina S Shaw
- Department of Dermatology, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Ruth Ann Vleugels
- Department of Dermatology, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Elena Massarotti
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Karen H Costenbader
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Michael B Brenner
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - James A Lederer
- Department of Surgery, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Judd F Hultquist
- Division of Infectious Diseases, The Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Jaehyuk Choi
- Department of Biochemistry and Molecular Genetics, The Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
- Department of Dermatology, The Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
- Center of Human Immunobiology, The Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
- Center of Synthetic Biology, Northwestern University, Evanston, IL, USA.
- Center for Genetic Medicine, Northwestern University, Chicago, IL, USA.
| | - Deepak A Rao
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA.
| |
Collapse
|
25
|
Chen J, Wang Z, Zhu Q, Ren S, Xu Y, Wang G, Zhou L. Comprehensive analysis and experimental verification of the mechanism of action of T cell-mediated tumor-killing related genes in Colon adenocarcinoma. Transl Oncol 2024; 43:101918. [PMID: 38412662 PMCID: PMC10907202 DOI: 10.1016/j.tranon.2024.101918] [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: 10/15/2023] [Revised: 02/07/2024] [Accepted: 02/17/2024] [Indexed: 02/29/2024] Open
Abstract
BACKGROUND Colorectal cancer (CRC) is a prevalent malignancy of the digestive tract. A new prognostic scoring model for colon adenocarcinoma (COAD) is developed in this study based on the genes involved in tumor cell-mediated killing of T cells (GSTTKs), accurately stratifying COAD patients, thus improving the current status of personalized treatment. METHOD The GEO and TCGA databases served as the sources of the data for the COAD cohort. This study identified GSTTKs-related genes in COAD through single-factor Cox analysis. These genes were used to categorize COAD patients into several subtypes via unsupervised clustering analysis. The biological pathways and tumor microenvironments of different subgroups were compared. We performed intersection analysis between different subtypes to obtain intersection genes. Single-factor Cox regression analysis and Lasso-Cox analysis were conducted to establish clinical prognostic models. Two methods are used to assess the accuracy of model predictions: ROC and Kaplan-Meier analysis. Next, the prediction model was further validated in the validation cohort. Differential immune cell infiltration between various risk categories was identified via single sample gene set enrichment analysis (ssGSEA). The COAD model's gene expression was validated via single-cell data analysis and experiments. RESULT We established two distinct GSTTKs-related subtypes. Biological processes and immune cell tumor invasion differed significantly between various subtypes. Clinical prognostic models were created using five GSTTKs-related genes. The model's risk score independently served as a prognostic factor. COAD patients were classified as low- or high-risk depending on their risk scores. Patients in the low-risk category recorded a greater chance of surviving. The outcomes from the validation cohort match those from the training set. Risk scores and several tumor-infiltrating immune cells were strongly correlated, according to ssGSEA. Single-cell data illustrated that the model's genes were linked to several immune cells. The experimental results demonstrated a significant increase in the expression of HOXC6 in colon cancer tissue. CONCLUSION Our research findings established a new gene signature for COAD. This gene signature helps to accurately stratify the risk of COAD patients and improve the current status of individualized care.
Collapse
Affiliation(s)
- Jing Chen
- Department of Medical Laboratory, Clinical Medical College, Yangzhou University, Northern Jiangsu People's Hospital, Yangzhou, Jiangsu, 225009, China
| | - Zhengfang Wang
- Department of Medical Laboratory, Clinical Medical College, Yangzhou University, Northern Jiangsu People's Hospital, Yangzhou, Jiangsu, 225009, China
| | - Qin Zhu
- Department of Trauma Hand Surgery, Dalian Third People's Hospital, Dalian 116000, China
| | - Shiqi Ren
- Department of Hand Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China
| | - Yanhua Xu
- Department of Medical Laboratory, Clinical Medical College, Yangzhou University, Northern Jiangsu People's Hospital, Yangzhou, Jiangsu, 225009, China
| | - Guangzhou Wang
- Department of Medical Laboratory, Clinical Medical College, Yangzhou University, Northern Jiangsu People's Hospital, Yangzhou, Jiangsu, 225009, China.
| | - Lin Zhou
- Department of Medical Laboratory, Clinical Medical College, Yangzhou University, Northern Jiangsu People's Hospital, Yangzhou, Jiangsu, 225009, China.
| |
Collapse
|
26
|
Vahidian F, Lamaze FC, Bouffard C, Coulombe F, Gagné A, Blais F, Tonneau M, Orain M, Routy B, Manem VSK, Joubert P. CXCL13 Positive Cells Localization Predict Response to Anti-PD-1/PD-L1 in Pulmonary Non-Small Cell Carcinoma. Cancers (Basel) 2024; 16:708. [PMID: 38398098 PMCID: PMC10887067 DOI: 10.3390/cancers16040708] [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: 11/18/2023] [Revised: 01/25/2024] [Accepted: 01/29/2024] [Indexed: 02/25/2024] Open
Abstract
Background: Immune checkpoint inhibitors (ICIs) have revolutionized non-small cell lung cancers (NSCLCs) treatment, but only 20-30% of patients benefit from these treatments. Currently, PD-L1 expression in tumor cells is the only clinically approved predictor of ICI response in lung cancer, but concerns arise due to its low negative and positive predictive value. Recent studies suggest that CXCL13+ T cells in the tumor microenvironment (TME) may be a good predictor of response. We aimed to assess if CXCL13+ cell localization within the TME can predict ICI response in advanced NSCLC patients. Methods: This retrospective study included 65 advanced NSCLC patients treated with Nivolumab/Pembrolizumab at IUCPQ or CHUM and for whom a pretreatment surgical specimen was available. Good responders were defined as having a complete radiologic response at 1 year, and bad responders were defined as showing cancer progression at 1 year. IHC staining for CXCL13 was carried out on a representative slide from a resection specimen, and CXCL13+ cell density was evaluated in tumor (T), invasive margin (IM), non-tumor (NT), and tertiary lymphoid structure (TLS) compartments. Cox models were used to analyze progression-free survival (PFS) and overall survival (OS) probability, while the Mann-Whitney test was used to compare CXCL13+ cell density between responders and non-responders. Results: We showed that CXCL13+ cell density localization within the TME is associated with ICI efficacy. An increased density of CXCL13+ cells across all compartments was associated with a poorer prognostic (OS; HR = 1.22; 95%CI = 1.04-1.42; p = 0.01, PFS; HR = 1.16; p = 0.02), or a better prognostic when colocalized within TLSs (PFS; HR = 0.84, p = 0.03). Conclusion: Our results support the role of CXCL13+ cells in advanced NSCLC patients, with favorable prognosis when localized within TLSs and unfavorable prognosis when present elsewhere. The concomitant proximity of CXCL13+ and CD20+ cells within TLSs may favor antigen presentation to T cells, thus enhancing the effect of PD-1/PD-L1 axis inhibition. Further validation is warranted to confirm the potential relevance of this biomarker in a clinical setting.
Collapse
Affiliation(s)
- Fatemeh Vahidian
- Centre de Recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec (IUCPQ), Quebec City, QC G1V 4G5, Canada (F.C.L.); (M.O.)
- Faculty of Medicine, Laval University, Quebec City, QC G1V 4G5, Canada (F.B.)
| | - Fabien C. Lamaze
- Centre de Recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec (IUCPQ), Quebec City, QC G1V 4G5, Canada (F.C.L.); (M.O.)
| | - Cédrik Bouffard
- Centre de Recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec (IUCPQ), Quebec City, QC G1V 4G5, Canada (F.C.L.); (M.O.)
- Faculty of Medicine, Laval University, Quebec City, QC G1V 4G5, Canada (F.B.)
| | - François Coulombe
- Faculty of Medicine, Laval University, Quebec City, QC G1V 4G5, Canada (F.B.)
| | - Andréanne Gagné
- Centre de Recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec (IUCPQ), Quebec City, QC G1V 4G5, Canada (F.C.L.); (M.O.)
- Faculty of Medicine, Laval University, Quebec City, QC G1V 4G5, Canada (F.B.)
| | - Florence Blais
- Faculty of Medicine, Laval University, Quebec City, QC G1V 4G5, Canada (F.B.)
| | - Marion Tonneau
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC H2X 0A9, Canada; (M.T.)
| | - Michèle Orain
- Centre de Recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec (IUCPQ), Quebec City, QC G1V 4G5, Canada (F.C.L.); (M.O.)
| | - Bertrand Routy
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC H2X 0A9, Canada; (M.T.)
| | - Venkata S. K. Manem
- Centre de Recherche du CHU de Québec—Université Laval, Quebec City, QC G1V 4G5, Canada
- Department of Mathematics and Computer Science, Université du Québec à Trois-Rivières, Trois-Rivières, QC G8Z 4M3, Canada
| | - Philippe Joubert
- Centre de Recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec (IUCPQ), Quebec City, QC G1V 4G5, Canada (F.C.L.); (M.O.)
- Faculty of Medicine, Laval University, Quebec City, QC G1V 4G5, Canada (F.B.)
| |
Collapse
|
27
|
Zhang W, Liu L, Liu X, Han C, Li Q. The levels of immunosuppressive checkpoint protein PD-L1 and tumor-infiltrating lymphocytes were integrated to reveal the glioma tumor microenvironment. ENVIRONMENTAL TOXICOLOGY 2024; 39:815-829. [PMID: 37792606 DOI: 10.1002/tox.23979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Revised: 08/29/2023] [Accepted: 09/18/2023] [Indexed: 10/06/2023]
Abstract
In spite of significant strides in the realm of cancer biology and therapeutic interventions, the clinical prognosis for patients afflicted with glioblastoma (GBM) remains distressingly dismal. The tumor immune microenvironment (TIME), a crucial player in the progression, treatment response, and prognostic trajectory of glioma, warrants thorough exploration. Within this intricate microcosm, the immunosuppressive checkpoint protein PD-L1 and tumor-infiltrating lymphocytes (TILs) emerge as pivotal constituents, underscoring their potential role in deciphering glioma biology and informing treatment strategies. However, prognostic models based on the association between PD-L1 expression and TIL infiltration in the tumor immune microenvironment have not been established. The aim of this study was to explore TIME genes associated with PD-L1 expression and TIL invasion and to construct a risk score for predicting the overall survival (OS) of GBM patients based on these genes. The samples were separately classified according to the PD-L1 expression level and TIL score and TIME-related genes were identified using differential expression and weighted gene co-expression network analysis. The DEGs were subjected to least absolute contraction and selection operator (LASSO) -Cox regression to construct TIME associated risk score (TIMErisk). A TIMErisk was developed based on STEAP3 and CXCL13 genes. The STLEAP3 was demonstrated to be involved in glioma progression. The results showed that the patients in the high TIMErisk group had poor OS compared with subjects in the low TIMErisk group. The biological phenotypes associated with TIMErisk were analyzed in terms of functional enrichment, tumor immune profile, and tumor mutation profile. The results on tumor immune dysfunction and exclusion dysfunction (TIDE) score and immune surface score (IPS) showed that GBM patients with different TIME risks had different responses to immunotherapy. Tumor purity analysis indicated that PD-L1 and TIL scores were positively correlated with TIMErisk score and negatively correlated with tumor purity. These results show that the TIMErisk-based prognostic model had high predictive value for the prognosis and immune characteristics of GBM patients. Immunohistochemical staining images of patients in the high and low TIMErisk groups were analyzed, showing that the degree of immune cell infiltration was higher in the high TIMErisk group relative to the low TIMErisk group. The present study provides a basis for understanding glioma tumor microenvironment and a foundation for conducting comprehensive immunogenomic analysis.
Collapse
Affiliation(s)
- Weizhong Zhang
- Department of Neurosurgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Li Liu
- Department of Neurosurgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xiaoyan Liu
- Department of Neurosurgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Cheng Han
- Department of Neurosurgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Qun Li
- Department of Neurosurgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
28
|
Ding C, Xiao T, Deng Y, Yang H, Xu B, Li J, Lv Z. The Teleost CXCL13-CXCR5 Axis Induces Inflammatory Cytokine Expression through the Akt-NF-κB, p38-AP-1, and p38-NF-κB Pathways. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:317-334. [PMID: 38054894 DOI: 10.4049/jimmunol.2300237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 11/08/2023] [Indexed: 12/07/2023]
Abstract
The ancestors of chemokines originate in the most primitive of vertebrates, which has recently attracted great interest in the immune functions and the underlying mechanisms of fish chemokines. In the current study, we identified an evolutionarily conserved chemokine, CiCXCL13, from a teleost fish, grass carp. CiCXCL13 was characterized by a typical SCY (small cytokine CXC) domain and four cysteine residues (C34, C36, C61, C77), with the first two cysteines separated by a random amino acid residue, although it shared 24.2-54.8% identity with the counterparts from other vertebrates. CiCXCL13 was an inducible chemokine, whose expression was significantly upregulated in the immune tissues of grass carps after grass carp reovirus infection. CiCXCL13 could bind to the membrane of grass carp head kidney leukocytes and promote cell migration, NO release, and the expression of >15 inflammatory cytokines, including IL-1β, TNF-α, IL-10 and TGF-β1, thus regulating the inflammatory response. Mechanistically, CiCXCL13 interacted with its evolutionarily conserved receptor CiCXCR5 and activated the Akt-NF-κB and p38-AP-1 pathways, as well as a previously unrevealed p38-NF-κB pathway, to efficiently induce inflammatory cytokine expression, which was distinct from that reported in mammals. Zebrafish CXCL13 induced inflammatory cytokine expression through Akt, p38, NF-κB, and AP-1 as CiCXCL13. Meanwhile, the CiCXCL13-CiCXCR5 axis-mediated inflammatory activity was negatively shaped by grass carp atypical chemokine receptor 2 (CiACKR2). The present study is, to our knowledge, the first to comprehensively define the immune function of CXCL13 in inflammatory regulation and the underlying mechanism in teleosts, and it provides a valuable perspective on the evolution and biology of fish chemokines.
Collapse
Affiliation(s)
- Chunhua Ding
- Hunan Engineering Technology Research Center of Featured Aquatic Resources Utilization, Hunan Agricultural University, Changsha, China; and Fisheries College, Hunan Agricultural University, Changsha, China
| | - Tiaoyi Xiao
- Hunan Engineering Technology Research Center of Featured Aquatic Resources Utilization, Hunan Agricultural University, Changsha, China; and Fisheries College, Hunan Agricultural University, Changsha, China
| | - Yadong Deng
- Hunan Engineering Technology Research Center of Featured Aquatic Resources Utilization, Hunan Agricultural University, Changsha, China; and Fisheries College, Hunan Agricultural University, Changsha, China
| | - Hong Yang
- Hunan Engineering Technology Research Center of Featured Aquatic Resources Utilization, Hunan Agricultural University, Changsha, China; and Fisheries College, Hunan Agricultural University, Changsha, China
| | - Baohong Xu
- Hunan Engineering Technology Research Center of Featured Aquatic Resources Utilization, Hunan Agricultural University, Changsha, China; and Fisheries College, Hunan Agricultural University, Changsha, China
| | - Junhua Li
- Hunan Engineering Technology Research Center of Featured Aquatic Resources Utilization, Hunan Agricultural University, Changsha, China; and Fisheries College, Hunan Agricultural University, Changsha, China
| | - Zhao Lv
- Hunan Engineering Technology Research Center of Featured Aquatic Resources Utilization, Hunan Agricultural University, Changsha, China; and Fisheries College, Hunan Agricultural University, Changsha, China
| |
Collapse
|
29
|
Krenács L, Krenács D, Borbényi Z, Tóth E, Nagy A, Piukovics K, Bagdi E. Comparison of Follicular Helper T-Cell Markers with the Expression of the Follicular Homing Marker CXCR5 in Peripheral T-Cell Lymphomas-A Reappraisal of Follicular Helper T-Cell Lymphomas. Int J Mol Sci 2023; 25:428. [PMID: 38203606 PMCID: PMC10778845 DOI: 10.3390/ijms25010428] [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: 11/30/2023] [Revised: 12/20/2023] [Accepted: 12/25/2023] [Indexed: 01/12/2024] Open
Abstract
Peripheral T-cell lymphomas (PTCLs) expressing multiple follicular T helper (TFH) cell-related antigens are now classified as TFH lymphomas (TFHL), including angioimmunoblastic, follicular, and not otherwise specified (NOS) types. CXCR5 is the TFH cell-defining chemokine receptor that, together with its ligand CXCL13, plays a critical role in the development of follicles and the positioning of TFH and B cells within follicles. A comprehensive immunomorphologic study was performed to investigate the expression pattern of CXCR5 in a large cohort of nodal PTCLs, particularly those with a TFH cell phenotype, and to compare its expression with six other TFH cell-related antigens. We found that CXCR5 is widely expressed in neoplastic TFH cells, except in TFHL-NOS, and represents a specific marker of this lymphoma entity. Our results suggest that CXCR5 directs the distribution of neoplastic T cells in the affected lymph nodes and may influence the formation of the pathognomic pathological FDC network.
Collapse
Affiliation(s)
- László Krenács
- Laboratory of Tumor Pathology and Molecular Diagnostics, 6726 Szeged, Hungary (E.B.)
| | - Dóra Krenács
- Laboratory of Tumor Pathology and Molecular Diagnostics, 6726 Szeged, Hungary (E.B.)
- Division of Haematology, Department of Internal Medicine, Albert Szent-Györgyi Clinical Center, University of Szeged, 6721 Szeged, Hungary
| | - Zita Borbényi
- Division of Haematology, Department of Internal Medicine, Albert Szent-Györgyi Clinical Center, University of Szeged, 6721 Szeged, Hungary
| | - Erika Tóth
- Department of Pathology, National Institute of Oncology, 1122 Budapest, Hungary;
| | - Anna Nagy
- 1st Department of Pathology and Experimental Cancer Research, Faculty of Medicine, Semmelweis University, 1085 Budapest, Hungary
| | - Klára Piukovics
- Division of Haematology, Department of Internal Medicine, Albert Szent-Györgyi Clinical Center, University of Szeged, 6721 Szeged, Hungary
| | - Enikő Bagdi
- Laboratory of Tumor Pathology and Molecular Diagnostics, 6726 Szeged, Hungary (E.B.)
| |
Collapse
|
30
|
Harada T, Kikushige Y, Miyamoto T, Uno K, Niiro H, Kawakami A, Koga T, Akashi K, Yoshizaki K. Peripheral helper-T-cell-derived CXCL13 is a crucial pathogenic factor in idiopathic multicentric Castleman disease. Nat Commun 2023; 14:6959. [PMID: 37907518 PMCID: PMC10618253 DOI: 10.1038/s41467-023-42718-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 10/19/2023] [Indexed: 11/02/2023] Open
Abstract
Castleman disease (CD) is a rare lymphoproliferative disorder. Among subtypes of CD, idiopathic multicentric CD-not otherwise specified (iMCD-NOS) has a poor prognosis and its pathogenesis is largely unknown. Here we present a xenotransplantation model of iMCD-NOS pathogenesis. Immunodeficient mice, transplanted with lymph node (LN) cells from iMCD-NOS patients, develop iMCD-like lethal inflammation, while mice transplanted with LN cells from non-iMCD patients without inflammation serve as negative control. Grafts depleted of human CD3+ T cells fail to induce inflammation in vivo. Upon engraftment, peripheral helper T (Tph) cells expand and levels of human CXCL13 substantially increase in the sera of mice. A neutralizing antibody against human CXCL13 blocks development of inflammation and improves survival in the recipient mice. Our study thus indicates that Tph cells, producing CXCL13 play a critical role in the pathogenesis of iMCD-NOS, and establishes iMCD-NOS as an immunoregulatory disorder.
Collapse
Affiliation(s)
- Takuya Harada
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medicine, Fukuoka, Japan
| | - Yoshikane Kikushige
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medicine, Fukuoka, Japan
- Center for Cellular and Molecular Medicine, Kyushu University Hospital, Fukuoka, Japan
| | - Toshihiro Miyamoto
- Department of Hematology, Faculty of Medicine, Institute of Medical Pharmaceutical and Health Sciences, Kanazawa University, Ishikawa, Japan
| | - Kazuko Uno
- Luis Pasteur Center for Medical Research, Kyoto, Japan
| | - Hiroaki Niiro
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medicine, Fukuoka, Japan
| | - Atsushi Kawakami
- Department of Immunology and Rheumatology, Division of Advanced Preventive Medical Sciences, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Tomohiro Koga
- Department of Immunology and Rheumatology, Division of Advanced Preventive Medical Sciences, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Koichi Akashi
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medicine, Fukuoka, Japan.
- Center for Cellular and Molecular Medicine, Kyushu University Hospital, Fukuoka, Japan.
| | - Kazuyuki Yoshizaki
- The Institute of Scientific and Industrial Research, SANKEN, Osaka University, Osaka, Japan.
- Medical corporation of Tokushukai, Osaka, Japan.
| |
Collapse
|
31
|
Jung HH, Kim JY, Cho EY, Lee JE, Kim SW, Nam SJ, Park YH, Ahn JS, Im YH. A Retrospective Exploratory Analysis for Serum Extracellular Vesicles Reveals APRIL (TNFSF13), CXCL13, and VEGF-A as Prognostic Biomarkers for Neoadjuvant Chemotherapy in Triple-Negative Breast Cancer. Int J Mol Sci 2023; 24:15576. [PMID: 37958571 PMCID: PMC10647725 DOI: 10.3390/ijms242115576] [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: 09/13/2023] [Revised: 10/18/2023] [Accepted: 10/23/2023] [Indexed: 11/15/2023] Open
Abstract
Neoadjuvant chemotherapy (NAC) is widely used as a standard treatment for early-stage triple-negative breast cancer (TNBC). While patients who achieve pathologic complete response (pCR) have a highly favorable outcome, patients who do not achieve pCR have variable prognoses. It is important to identify patients who are most likely to have poor survival outcomes to identify candidates for more aggressive therapeutic approaches after NAC. Many studies have demonstrated that cytokines and growth factors packaged into extracellular vesicles (EVs) have an essential role in tumor progression and drug resistance. In this study, we examined the role of serum-derived EV-associated cytokines as prognostic biomarkers for long-term outcomes in patients who underwent anthracycline-taxane-based NAC. We isolated extracellular vesicles from the serum of 190 TNBC patients who underwent NAC between 2015 and 2018 at Samsung Medical Center. EV-associated cytokine concentrations were measured with ProcartaPlex Immune Monitoring 65-plex panels. The prognostic value of EV-associated cytokines was studied. We found that patients with high EV_APRIL, EV_CXCL13, and EV_VEGF-A levels had shorter overall survival (OS). We further evaluated the role of these selected biomarkers as prognostic factors in patients with residual disease (RD) after NAC. Even in patients with RD, high levels of EV_APRIL, EV_CXCL13, and EV_VEGF-A were correlated with poor OS. In all subgroup analyses, EV_CXCL13 overexpression was significantly associated with poor overall survival. Moreover, multivariate analysis indicated that a high level of EV_CXCL13 was an independent predictor of poor OS. Correlation analysis between biomarker levels in EVs and serum showed that EV_VEGF-A positively correlated with soluble VEGF-A but not CXCL13. An elevated level of soluble VEGF-A was also associated with poor OS. These findings suggest that EV_APRIL, EV_CXCL13, and EV_VEGF-A may be useful in identifying TNBC patients at risk of poor survival outcomes after NAC.
Collapse
Affiliation(s)
- Hae Hyun Jung
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul 06351, Republic of Korea; (H.H.J.); (J.-Y.K.); (Y.H.P.)
- Biomedical Research Institute, Samsung Medical Center, Seoul 06351, Republic of Korea
| | - Ji-Yeon Kim
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul 06351, Republic of Korea; (H.H.J.); (J.-Y.K.); (Y.H.P.)
- Biomedical Research Institute, Samsung Medical Center, Seoul 06351, Republic of Korea
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Seoul 06351, Republic of Korea;
- School of Medicine, Sungkyunkwan University, Suwon 16419, Republic of Korea; (E.Y.C.); (J.E.L.); (S.W.K.); (S.J.N.)
| | - Eun Yoon Cho
- School of Medicine, Sungkyunkwan University, Suwon 16419, Republic of Korea; (E.Y.C.); (J.E.L.); (S.W.K.); (S.J.N.)
- Department of Pathology, Samsung Medical Center, Seoul 06351, Republic of Korea
| | - Jeong Eon Lee
- School of Medicine, Sungkyunkwan University, Suwon 16419, Republic of Korea; (E.Y.C.); (J.E.L.); (S.W.K.); (S.J.N.)
- Department of Surgery, Samsung Medical Center, Seoul 06351, Republic of Korea
| | - Seok Won Kim
- School of Medicine, Sungkyunkwan University, Suwon 16419, Republic of Korea; (E.Y.C.); (J.E.L.); (S.W.K.); (S.J.N.)
- Department of Surgery, Samsung Medical Center, Seoul 06351, Republic of Korea
| | - Seok Jin Nam
- School of Medicine, Sungkyunkwan University, Suwon 16419, Republic of Korea; (E.Y.C.); (J.E.L.); (S.W.K.); (S.J.N.)
- Department of Surgery, Samsung Medical Center, Seoul 06351, Republic of Korea
| | - Yeon Hee Park
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul 06351, Republic of Korea; (H.H.J.); (J.-Y.K.); (Y.H.P.)
- Biomedical Research Institute, Samsung Medical Center, Seoul 06351, Republic of Korea
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Seoul 06351, Republic of Korea;
- School of Medicine, Sungkyunkwan University, Suwon 16419, Republic of Korea; (E.Y.C.); (J.E.L.); (S.W.K.); (S.J.N.)
| | - Jin Seok Ahn
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Seoul 06351, Republic of Korea;
- School of Medicine, Sungkyunkwan University, Suwon 16419, Republic of Korea; (E.Y.C.); (J.E.L.); (S.W.K.); (S.J.N.)
| | - Young-Hyuck Im
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul 06351, Republic of Korea; (H.H.J.); (J.-Y.K.); (Y.H.P.)
- Biomedical Research Institute, Samsung Medical Center, Seoul 06351, Republic of Korea
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Seoul 06351, Republic of Korea;
- School of Medicine, Sungkyunkwan University, Suwon 16419, Republic of Korea; (E.Y.C.); (J.E.L.); (S.W.K.); (S.J.N.)
| |
Collapse
|
32
|
Grabarczyk M, Ksiazek-Winiarek D, Glabinski A, Szpakowski P. Dietary Polyphenols Decrease Chemokine Release by Human Primary Astrocytes Responding to Pro-Inflammatory Cytokines. Pharmaceutics 2023; 15:2294. [PMID: 37765263 PMCID: PMC10537369 DOI: 10.3390/pharmaceutics15092294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 08/26/2023] [Accepted: 09/01/2023] [Indexed: 09/29/2023] Open
Abstract
Astrocytes are considered to be the dominant cell fraction of the central nervous system. They play a supportive and protective role towards neurons, and regulate inflammatory processes; they thus make suitable targets for drugs and supplements, such as polyphenolic compounds. However, due to their wide range, knowledge of their anti-inflammatory potential remains relatively incomplete. The aim of this study was therefore to determine whether myricetin and chrysin are able to decrease chemokine release in reactive astrocytes. To assess the antioxidant and anti-inflammatory potential of polyphenols, human primary astrocytes were cultured in the presence of a reactive and neurotoxic astrocyte-inducing cytokine mixture (TNF-α, IL-1a, C1q), either alone or in the presence of myricetin or chrysin. The examined polyphenols were able to modify the secretion of chemokines by human cortical astrocytes, especially CCL5 (chrysin), CCL1 (myricetin) and CCL2 (both), while cell viability was not affected. Surprisingly, the compounds did not demonstrate any antioxidant properties in the astrocyte cultures.
Collapse
|
33
|
Matz H, Dooley H. 450 million years in the making: mapping the evolutionary foundations of germinal centers. Front Immunol 2023; 14:1245704. [PMID: 37638014 PMCID: PMC10450919 DOI: 10.3389/fimmu.2023.1245704] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 07/25/2023] [Indexed: 08/29/2023] Open
Abstract
Germinal centers (GCs) are distinct microanatomical structures that form in the secondary lymphoid organs of endothermic vertebrates (i.e., mammals and some birds). Within GCs, B cells undergo a Darwinian selection process to identify clones which can respond to pathogen insult as well as affinity mature the B cell repertoire. The GC response ultimately generates memory B cells and bone marrow plasma cells which facilitate humoral immunological memory, the basis for successful vaccination programs. GCs have not been observed in the secondary lymphoid organs of ectothermic jawed vertebrates (i.e., fishes, reptiles, and amphibians). However, abundant research over the past decades has indicated these organisms can produce antigen specific B cell responses and some degree of affinity maturation. This review examines data demonstrating that the fundamentals of B cell selection may be more conserved across vertebrate phylogeny than previously anticipated. Further, research in both conventional mammalian model systems and comparative models raises the question of what evolutionary benefit GCs provide endotherms if they are seemingly unnecessary for generating the basic functional components of jawed vertebrate humoral adaptive immune responses.
Collapse
|
34
|
Sato-Fukuba M, Arakaki R, Ushio A, Otsuka K, Nagao R, Matsuzawa S, Tawara H, Tsunematsu T, Ishimaru N. CD4 + T-cell-dependent differentiation of CD23 + follicular B cells contributes to the pulmonary pathology in a primary Sjögren's syndrome mouse model. Front Immunol 2023; 14:1217492. [PMID: 37475871 PMCID: PMC10354287 DOI: 10.3389/fimmu.2023.1217492] [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: 05/05/2023] [Accepted: 06/16/2023] [Indexed: 07/22/2023] Open
Abstract
Introduction Primary Sjögren's syndrome (pSS) is a systemic autoimmune disease that affects the function of exocrine glands, such as the lacrimal and the salivary glands. Extraglandular lesions and malignant lymphoma also occur during the progressive stage of pSS. We have, herein, focused on the pulmonary lesions of pSS and have aimed clarifying their pathophysiological mechanism by comparing the glandular with the extraglandular lesions observed in a mouse model of pSS. Results The histopathological analysis of lung tissues obtained from NFS/sld mice that have undergone neonatal thymectomy was performed. Moreover, in vivo and in vitro experiments were conducted along with immunological analyses in order to characterize the unique phenotypes of the pulmonary lesions identified in these pSS model mice. Inflammatory lesions with a bronchus-associated lymphoid tissue-like structure were identified in the lungs of pSS model mice. In addition, relative to salivary gland lesions, pulmonary lesions showed increased CD23+ follicular B (FB) cells. In vitro and pulmonary B cells were more readily driven to CD23+ FB cell phenotype than salivary gland B cells in pSS model mice. Furthermore, the CD23+ FB cell differentiation was found to be enhanced in a CD4+ T-cell-dependent manner under a Th2-type condition in the lungs of herein examined pSS model mice. Discussion A Th2-type response in the pSS lung may promote the progression of autoimmune lesions through an enhanced abnormal differentiation of B cells.
Collapse
Affiliation(s)
- Mami Sato-Fukuba
- Department of Oral Molecular Pathology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
- Department of Oral Medicine, Tokushima University Hospital, Tokushima, Japan
| | - Rieko Arakaki
- Department of Oral Molecular Pathology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Aya Ushio
- Department of Oral Molecular Pathology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Kunihiro Otsuka
- Department of Oral Molecular Pathology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Ruka Nagao
- Department of Oral Molecular Pathology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Shigefumi Matsuzawa
- Department of Oral Molecular Pathology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Hiroaki Tawara
- Department of Oral Molecular Pathology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Takaaki Tsunematsu
- Department of Oral Molecular Pathology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Naozumi Ishimaru
- Department of Oral Molecular Pathology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| |
Collapse
|
35
|
Trunfio M, Mighetto L, Napoli L, Atzori C, Nigra M, Guastamacchia G, Bonora S, Di Perri G, Calcagno A. Cerebrospinal Fluid CXCL13 as Candidate Biomarker of Intrathecal Immune Activation, IgG Synthesis and Neurocognitive Impairment in People with HIV. J Neuroimmune Pharmacol 2023; 18:169-182. [PMID: 37166552 DOI: 10.1007/s11481-023-10066-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Accepted: 04/17/2023] [Indexed: 05/12/2023]
Abstract
Plasma C-X-C-motif chemokine ligand-13 (CXCL13) has been linked to disease progression and mortality in people living with HIV (PLWH) and is a candidate target for immune-based strategies for HIV cure. Its role in central nervous system (CNS) of PLWH has not been detailed. We described CSF CXCL13 levels and its potential associations with neurological outcomes. Cross-sectional study enrolling PLWH without confounding for CXCL13 production. Subjects were divided according to CSF HIV-RNA in undetectable (< 20 cp/mL) and viremics. CSF CXCL13, and biomarkers of blood-brain barrier (BBB) impairment, intrathecal synthesis, and immune activation were measured by commercial immunoturbidimetric and ELISA assays. All subjects underwent neurocognitive assessment. Sensitivity analyses were conducted in subjects with intact BBB only. 175 participants were included. Detectable CSF CXCL13 was more common in the viremic (31.4%) compared to the undetectable group (13.5%; OR 2.9 [1.4-6.3], p = 0.006), but median levels did not change (15.8 [8.2-91.0] vs 10.0 [8.1-14.2] pg/mL). In viremics (n = 86), CXCL13 associated with higher CSF HIV-RNA, proteins, neopterin, intrathecal synthesis and BBB permeability. In undetectable participants (n = 89), CXCL13 associated with higher CD4+T-cells count, CD4/CD8 ratio, CSF proteins, neopterin, and intrathecal synthesis. The presence of CXCL13 in the CSF of undetectable participants was associated with increased odds of HIV-associated neurocognitive disorders (58.3% vs 28.6%, p = 0.041). Sensitivity analyses confirmed all these findings. CXCL13 is detectable in the CSF of PLWH that show increased intrathecal IgG synthesis and immune activation. In PLWH with CSF viral suppression, CXCL13 was also associated with neurocognitive impairment.
Collapse
Affiliation(s)
- Mattia Trunfio
- Infectious Diseases Unit, Department of Medical Sciences, Amedeo di Savoia Hospital, University of Torino, Torino, 10149, Italy.
- HIV Neurobehavioral Research Center (HNRC), Department of Psychiatry, University of California San Diego, San Diego, CA, 92093, USA.
| | - Lorenzo Mighetto
- Diagnostic Laboratory Unit, Maria Vittoria Hospital, ASL Città di Torino, Torino, 10144, Italy
| | - Laura Napoli
- Infectious Diseases Unit, Department of Medical Sciences, Amedeo di Savoia Hospital, University of Torino, Torino, 10149, Italy
| | - Cristiana Atzori
- Unit of Neurology, Maria Vittoria Hospital, ASL Città di Torino, Torino, 10144, Italy
| | - Marco Nigra
- Diagnostic Laboratory Unit, Maria Vittoria Hospital, ASL Città di Torino, Torino, 10144, Italy
| | - Giulia Guastamacchia
- Unit of Neurology, Maria Vittoria Hospital, ASL Città di Torino, Torino, 10144, Italy
| | - Stefano Bonora
- Infectious Diseases Unit, Department of Medical Sciences, Amedeo di Savoia Hospital, University of Torino, Torino, 10149, Italy
| | - Giovanni Di Perri
- Infectious Diseases Unit, Department of Medical Sciences, Amedeo di Savoia Hospital, University of Torino, Torino, 10149, Italy
| | - Andrea Calcagno
- Infectious Diseases Unit, Department of Medical Sciences, Amedeo di Savoia Hospital, University of Torino, Torino, 10149, Italy
| |
Collapse
|
36
|
Marcial-Juárez E, Pérez-Toledo M, Nayar S, Pipi E, Alshayea A, Persaud R, Jossi SE, Lamerton R, Barone F, Henderson IR, Cunningham AF. Salmonella infection induces the reorganization of follicular dendritic cell networks concomitant with the failure to generate germinal centers. iScience 2023; 26:106310. [PMID: 36950118 PMCID: PMC10025972 DOI: 10.1016/j.isci.2023.106310] [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: 12/13/2022] [Revised: 02/07/2023] [Accepted: 02/26/2023] [Indexed: 03/06/2023] Open
Abstract
Germinal centers (GCs) are sites where plasma and memory B cells form to generate high-affinity, Ig class-switched antibodies. Specialized stromal cells called follicular dendritic cells (FDCs) are essential for GC formation. During systemic Salmonella Typhimurium (STm) infection GCs are absent, whereas extensive extrafollicular and switched antibody responses are maintained. The mechanisms that underpin the absence of GC formation are incompletely understood. Here, we demonstrate that STm induces a reversible disruption of niches within the splenic microenvironment, including the T and B cell compartments and the marginal zone. Alongside these effects after infection, mature FDC networks are strikingly absent, whereas immature FDC precursors, including marginal sinus pre-FDCs (MadCAM-1+) and perivascular pre-FDCs (PDGFRβ+) are enriched. As normal FDC networks re-establish, extensive GCs become detectable throughout the spleen. Therefore, the reorganization of FDC networks and the loss of GC responses are key, parallel features of systemic STm infections.
Collapse
Affiliation(s)
- Edith Marcial-Juárez
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, West Midlands, B15 2TT, United Kingdom
| | - Marisol Pérez-Toledo
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, West Midlands, B15 2TT, United Kingdom
| | - Saba Nayar
- Rheumatology Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, West Midlands, B15 2TT, United Kingdom
| | - Elena Pipi
- Rheumatology Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, West Midlands, B15 2TT, United Kingdom
| | - Areej Alshayea
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, West Midlands, B15 2TT, United Kingdom
| | - Ruby Persaud
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, West Midlands, B15 2TT, United Kingdom
| | - Sian E. Jossi
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, West Midlands, B15 2TT, United Kingdom
| | - Rachel Lamerton
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, West Midlands, B15 2TT, United Kingdom
| | - Francesca Barone
- Rheumatology Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, West Midlands, B15 2TT, United Kingdom
- National Institute for Health Research (NIHR) Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust, UK and Sandwell and West Birmingham Trust, Birmingham, West Midlands, B15 2TH, United Kingdom
| | - Ian R. Henderson
- Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD4072, Australia
| | - Adam F. Cunningham
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, West Midlands, B15 2TT, United Kingdom
| |
Collapse
|
37
|
Le Coz C, Oldridge DA, Herati RS, De Luna N, Garifallou J, Cruz Cabrera E, Belman JP, Pueschl D, Silva LV, Knox AVC, Reid W, Yoon S, Zur KB, Handler SD, Hakonarson H, Wherry EJ, Gonzalez M, Romberg N. Human T follicular helper clones seed the germinal center-resident regulatory pool. Sci Immunol 2023; 8:eade8162. [PMID: 37027481 PMCID: PMC10329285 DOI: 10.1126/sciimmunol.ade8162] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 03/16/2023] [Indexed: 04/09/2023]
Abstract
The mechanisms by which FOXP3+ T follicular regulatory (Tfr) cells simultaneously steer antibody formation toward microbe or vaccine recognition and away from self-reactivity remain incompletely understood. To explore underappreciated heterogeneity in human Tfr cell development, function, and localization, we used paired TCRVA/TCRVB sequencing to distinguish tonsillar Tfr cells that are clonally related to natural regulatory T cells (nTfr) from those likely induced from T follicular helper (Tfh) cells (iTfr). The proteins iTfr and nTfr cells differentially expressed were used to pinpoint their in situ locations via multiplex microscopy and establish their divergent functional roles. In silico analyses and in vitro tonsil organoid tracking models corroborated the existence of separate Treg-to-nTfr and Tfh-to-iTfr developmental trajectories. Our results identify human iTfr cells as a distinct CD38+, germinal center-resident, Tfh-descended subset that gains suppressive function while retaining the capacity to help B cells, whereas CD38- nTfr cells are elite suppressors primarily localized in follicular mantles. Interventions differentially targeting specific Tfr cell subsets may provide therapeutic opportunities to boost immunity or more precisely treat autoimmune diseases.
Collapse
Affiliation(s)
- Carole Le Coz
- Division of Immunology and Allergy, Children’s Hospital of Philadelphia, Philadelphia, PA
| | - Derek A. Oldridge
- Center for Computational and Genomic Medicine, The Children’s Hospital of Philadelphia, Philadelphia, PA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, Philadelphia, PA
| | - Ramin S. Herati
- Department of Medicine, NYU Grossman School of Medicine, New York, NY
| | - Nina De Luna
- Division of Immunology and Allergy, Children’s Hospital of Philadelphia, Philadelphia, PA
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - James Garifallou
- Center for Applied Genomics, Children’s Hospital of Philadelphia, Philadelphia, PA
| | - Emylette Cruz Cabrera
- Division of Immunology and Allergy, Children’s Hospital of Philadelphia, Philadelphia, PA
| | - Jonathan P Belman
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, Philadelphia, PA
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Dana Pueschl
- Division of Translational Medicine and Human Genetics, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Luisa V. Silva
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Ainsley V. C. Knox
- Division of Immunology and Allergy, Children’s Hospital of Philadelphia, Philadelphia, PA
| | - Whitney Reid
- Division of Immunology and Allergy, Children’s Hospital of Philadelphia, Philadelphia, PA
| | - Samuel Yoon
- Division of Immunology and Allergy, Children’s Hospital of Philadelphia, Philadelphia, PA
| | - Karen B. Zur
- Pediatric Otolaryngology, Children’s Hospital of Philadelphia, Philadelphia, PA
- Department of Otolaryngology: Head and Neck Surgery, University of Pennsylvania School of Medicine, Philadelphia, PA
| | - Steven D. Handler
- Pediatric Otolaryngology, Children’s Hospital of Philadelphia, Philadelphia, PA
- Department of Otolaryngology: Head and Neck Surgery, University of Pennsylvania School of Medicine, Philadelphia, PA
| | - Hakon Hakonarson
- Center for Applied Genomics, Children’s Hospital of Philadelphia, Philadelphia, PA
- Department of Pediatrics, Perelman School of Medicine, Philadelphia, PA
| | - E. John Wherry
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Michael Gonzalez
- Center for Applied Genomics, Children’s Hospital of Philadelphia, Philadelphia, PA
- Center for Cytokine Storm Treatment & Laboratory, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Neil Romberg
- Division of Immunology and Allergy, Children’s Hospital of Philadelphia, Philadelphia, PA
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
- Department of Pediatrics, Perelman School of Medicine, Philadelphia, PA
| |
Collapse
|
38
|
Sato S, Zhang XK, Matsuoka N, Sumichika Y, Saito K, Yoshida S, Matsumoto H, Temmoku J, Fujita Y, Asano T, Migita K. Transcription factor Fli-1 impacts the expression of CXCL13 and regulates immune cell infiltration into the kidney in MRL/lpr mouse. Lupus Sci Med 2023; 10:10/1/e000870. [PMID: 37094946 PMCID: PMC10152041 DOI: 10.1136/lupus-2022-000870] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 04/06/2023] [Indexed: 04/26/2023]
Abstract
OBJECTIVE Friend leukaemia virus integration 1 (Fli-1) regulates chemokine/cytokine expression and thus plays an important role in the development of lupus nephritis. Chemokine CXC ligand 13 (CXCL13) is a chemokine that promotes the formation of ectopic lymphoid structures and has been reported to be associated with the pathogenesis of lupus nephritis. The relationship between Fli-1 and CXCL13 is unknown. This study aims to elucidate whether Fli-1 impacts CXCL13 expression and contributes to the progression of lupus-like nephritis in adult MRL/lpr mouse. METHODS Serum CXCL13 levels were measured in adult wild-type (WT) MRL/lpr mice and Fli-1 heterozygote knockout (Fli-1+/-) MRL/lpr mice (4 months old or older) using ELISA. Renal mRNA expression (CXCL13 and related molecules) was measured using real-time PCR method. Kidneys were removed, stained and evaluated using a pathology scoring system. The grade of CXCL13 or CXC-chemokine receptor type 5 (CXCR5)-positive immune cell infiltration into the kidney was evaluated using immunostaining with anti-CXCL13 or anti-CXCR5 antibodies. We also used immunofluorescence staining with CXCL13- and CD11b-specific antibodies to detect the infiltration of CXCL13/CD11b double-positive immune cells. RESULTS Serum CXCL13 levels in Fli-1+/- MRL/lpr mice were significantly lower than that in WT MRL/lpr mice (545.5 and 960.5 pg/mL, p=0.02). Renal expression of CXCL13 mRNA and SRY-related HMG box4 (Sox4) (an important factor for B-cell development) levels were significantly lower in Fli-1+/- MRL/lpr mice. Renal histology scores in WT MRL/lpr mice revealed significantly increased glomerular inflammation. Despite similar interstitial immune cell infiltration into the kidney, the number of CXCL13- and CXCR5-positive cells was significantly lower in Fli-1+/- MRL/lpr mice than in WT mice. Furthermore, immunofluorescence staining revealed that Fli-1+/-MRL/lpr mice had significantly fewer CXCL13/CD11b double-positive immune cells. CONCLUSION Fli-1 regulates renal Sox4 mRNA expression and infiltration of CXCR5-positive cells as well as CXCL13/CD11b double-positive immune cells into the kidney, which affects CXCL13 expression and lupus-like nephritis.
Collapse
Affiliation(s)
- Shuzo Sato
- Department of Rheumatology, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Xian K Zhang
- Department of Rheumatology and Immunology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Naoki Matsuoka
- Department of Rheumatology, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Yuya Sumichika
- Department of Rheumatology, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Kenji Saito
- Department of Rheumatology, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Shuhei Yoshida
- Department of Rheumatology, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Haruki Matsumoto
- Department of Rheumatology, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Jumpei Temmoku
- Department of Rheumatology, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Yuya Fujita
- Department of Rheumatology, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Tomoyuki Asano
- Department of Rheumatology, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Kiyoshi Migita
- Department of Rheumatology, Fukushima Medical University School of Medicine, Fukushima, Japan
| |
Collapse
|
39
|
Cutaneous Lymphoma and Antibody-Directed Therapies. Antibodies (Basel) 2023; 12:antib12010021. [PMID: 36975368 PMCID: PMC10045448 DOI: 10.3390/antib12010021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 02/24/2023] [Accepted: 02/28/2023] [Indexed: 03/06/2023] Open
Abstract
The introduction of monoclonal antibodies such as rituximab to the treatment of cancer has greatly advanced the treatment scenario in onco-hematology. However, the response to these agents may be limited by insufficient efficacy or resistance. Antibody–drug conjugates are an attractive strategy to deliver payloads of toxicity or radiation with high selectivity toward malignant targets and limited unwanted effects. Primary cutaneous lymphomas are a heterogeneous group of disorders and a current area of unmet need in dermato-oncology due to the limited options available for advanced cases. This review briefly summarizes our current understanding of T and B cell lymphomagenesis, with a focus on recognized molecular alterations that may provide investigative therapeutic targets. The authors reviewed antibody-directed therapies investigated in the setting of lymphoma: this term includes a broad spectrum of approaches, from antibody–drug conjugates such as brentuximab vedotin, to bi-specific antibodies, antibody combinations, antibody-conjugated nanotherapeutics, radioimmunotherapy and, finally, photoimmunotherapy with specific antibody–photoadsorber conjugates, as an attractive strategy in development for the future management of cutaneous lymphoma.
Collapse
|
40
|
Sorini C, Tripathi KP, Wu S, Higdon SM, Wang J, Cheng L, Banerjee S, Reinhardt A, Kreslavsky T, Thorell A, Engstrand L, Du J, Villablanca EJ. Metagenomic and single-cell RNA-Seq survey of the Helicobacter pylori-infected stomach in asymptomatic individuals. JCI Insight 2023; 8:161042. [PMID: 36810249 PMCID: PMC9977493 DOI: 10.1172/jci.insight.161042] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 01/11/2023] [Indexed: 02/23/2023] Open
Abstract
Helicobacter pylori colonization of the gastric niche can persist for years in asymptomatic individuals. To deeply characterize the host-microbiota environment in H. pylori-infected (HPI) stomachs, we collected human gastric tissues and performed metagenomic sequencing, single-cell RNA-Seq (scRNA-Seq), flow cytometry, and fluorescent microscopy. HPI asymptomatic individuals had dramatic changes in the composition of gastric microbiome and immune cells compared with noninfected individuals. Metagenomic analysis uncovered pathway alterations related to metabolism and immune response. scRNA-Seq and flow cytometry data revealed that, in contrast to murine stomachs, ILC2s are virtually absent in the human gastric mucosa, whereas ILC3s are the dominant population. Specifically, proportion of NKp44+ ILC3s out of total ILCs were highly increased in the gastric mucosa of asymptomatic HPI individuals, and correlated with the abundance of selected microbial taxa. In addition, CD11c+ myeloid cells and activated CD4+ T cells and B cells were expanded in HPI individuals. B cells of HPI individuals acquired an activated phenotype and progressed into a highly proliferating germinal-center stage and plasmablast maturation, which correlated with the presence of tertiary lymphoid structures within the gastric lamina propria. Our study provides a comprehensive atlas of the gastric mucosa-associated microbiome and immune cell landscape when comparing asymptomatic HPI and uninfected individuals.
Collapse
Affiliation(s)
- Chiara Sorini
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet and University Hospital, Stockholm, Sweden.,Center of Molecular Medicine, Stockholm, Sweden
| | - Kumar P Tripathi
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet and University Hospital, Stockholm, Sweden.,Center of Molecular Medicine, Stockholm, Sweden
| | - Shengru Wu
- Department of Microbiology, Tumor and Cell Biology, Centre for Translational Microbiome Research, Karolinska Institutet, Stockholm, Sweden
| | - Shawn M Higdon
- Department of Microbiology, Tumor and Cell Biology, Centre for Translational Microbiome Research, Karolinska Institutet, Stockholm, Sweden
| | - Jing Wang
- Department of Microbiology, Tumor and Cell Biology, Centre for Translational Microbiome Research, Karolinska Institutet, Stockholm, Sweden
| | - Liqin Cheng
- Department of Microbiology, Tumor and Cell Biology, Centre for Translational Microbiome Research, Karolinska Institutet, Stockholm, Sweden
| | - Sanghita Banerjee
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet and University Hospital, Stockholm, Sweden.,Center of Molecular Medicine, Stockholm, Sweden
| | - Annika Reinhardt
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet and University Hospital, Stockholm, Sweden.,Center of Molecular Medicine, Stockholm, Sweden
| | - Taras Kreslavsky
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet and University Hospital, Stockholm, Sweden.,Center of Molecular Medicine, Stockholm, Sweden
| | | | - Lars Engstrand
- Department of Microbiology, Tumor and Cell Biology, Centre for Translational Microbiome Research, Karolinska Institutet, Stockholm, Sweden
| | - Juan Du
- Department of Microbiology, Tumor and Cell Biology, Centre for Translational Microbiome Research, Karolinska Institutet, Stockholm, Sweden
| | - Eduardo J Villablanca
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet and University Hospital, Stockholm, Sweden.,Center of Molecular Medicine, Stockholm, Sweden
| |
Collapse
|
41
|
Revisiting the Role of the CXCL13/CXCR5-Associated Immune Axis in Melanoma: Potential Implications for Anti-PD-1-Related Biomarker Research. Life (Basel) 2023; 13:life13020553. [PMID: 36836910 PMCID: PMC9958642 DOI: 10.3390/life13020553] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 02/13/2023] [Accepted: 02/14/2023] [Indexed: 02/18/2023] Open
Abstract
CXCL13 is a potent chemoattractant cytokine that promotes the migration of cells expressing its cognate receptor, CXCR5. Accordingly, T follicular helper cells and B cells migrate towards B cell follicles in lymph nodes, where the resulting spatial proximity promotes B cell/T cell interaction and antibody formation. Moreover, effector cells of the CXCL13/CXCR5-associated immune axis express PD-1, with corresponding circulating cells occurring in the blood. The formation of so-called ectopic or tertiary lymphoid structures, recently detected in different cancer types, represents an integral part of this axis, particularly in the context of its emerging role in anti-tumor defense. These aspects of the CXCL13/CXCR5-associated immune axis are highlighted in this review, which focuses on cutaneous malignant melanoma. Specifically, we elaborate on the role of this important immune axis as a possible ancillary target of immune checkpoint inhibition with anti-PD-1 antibodies in different therapeutic settings and as a potential source of predictive biomarkers regarding treatment efficacy.
Collapse
|
42
|
Pampusch MS, Sevcik EN, Quinn ZE, Davey BC, Berg JM, Gorrell-Brown I, Abdelaal HM, Rakasz EG, Rendahl A, Skinner PJ. Assessment of anti-CD20 antibody pre-treatment for augmentation of CAR-T cell therapy in SIV-infected rhesus macaques. Front Immunol 2023; 14:1101446. [PMID: 36825014 PMCID: PMC9941136 DOI: 10.3389/fimmu.2023.1101446] [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: 11/17/2022] [Accepted: 01/25/2023] [Indexed: 02/10/2023] Open
Abstract
During chronic HIV and SIV infections, the majority of viral replication occurs within lymphoid follicles. In a pilot study, infusion of SIV-specific CD4-MBL-CAR-T cells expressing the follicular homing receptor, CXCR5, led to follicular localization of the cells and a reduction in SIV viral loads in rhesus macaques. However, the CAR-T cells failed to persist. We hypothesized that temporary disruption of follicles would create space for CAR-T cell engraftment and lead to increased abundance and persistence of CAR-T cells. In this study we treated SIV-infected rhesus macaques with CAR-T cells and preconditioned one set with anti-CD20 antibody to disrupt the follicles. We evaluated CAR-T cell abundance and persistence in four groups of SIVmac239-infected and ART-suppressed animals: untreated, CAR-T cell treated, CD20 depleted, and CD20 depleted/CAR-T cell treated. In the depletion study, anti-CD20 was infused one week prior to CAR-T infusion and cessation of ART. Anti-CD20 antibody treatment led to temporary depletion of CD20+ cells in blood and partial depletion in lymph nodes. In this dose escalation study, there was no impact of CAR-T cell infusion on SIV viral load. However, in both the depleted and non-depleted animals, CAR-T cells accumulated in and around lymphoid follicles and were Ki67+. CAR-T cells increased in number in follicles from 2 to 6 days post-treatment, with a median 15.2-fold increase in follicular CAR-T cell numbers in depleted/CAR-T treated animals compared to an 8.1-fold increase in non-depleted CAR-T treated animals. The increase in CAR T cells in depleted animals was associated with a prolonged elevation of serum IL-6 levels and a rapid loss of detectable CAR-T cells. Taken together, these data suggest that CAR-T cells likely expanded to a greater extent in depleted/CAR-T cell treated animals. Further studies are needed to elucidate mechanisms mediating the rapid loss of CAR-T cells and to evaluate strategies to improve engraftment and persistence of HIV-specific CAR-T cells. The potential for an inflammatory cytokine response appears to be enhanced with anti-CD20 antibody treatment and future studies may require CRS control strategies. These studies provide important insights into cellular immunotherapy and suggest future studies for improved outcomes.
Collapse
Affiliation(s)
- Mary S. Pampusch
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, MN, United States
| | - Emily N. Sevcik
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, MN, United States
| | - Zoe E. Quinn
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, MN, United States
| | - Brianna C. Davey
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, MN, United States
| | - James M. Berg
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, MN, United States
| | - Ian Gorrell-Brown
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, MN, United States
| | - Hadia M. Abdelaal
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, MN, United States
| | - Eva G. Rakasz
- Wisconsin National Primate Research Center, University of Wisconsin, Madison WI, United States
| | - Aaron Rendahl
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, MN, United States
| | - Pamela J. Skinner
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, MN, United States
| |
Collapse
|
43
|
Schropp V, Chunder R, Dietel B, Tacke S, Kuerten S. The presence of cerebellar B cell aggregates is associated with a specific chemokine profile in the cerebrospinal fluid in a mouse model of multiple sclerosis. J Neuroinflammation 2023; 20:18. [PMID: 36717913 PMCID: PMC9885581 DOI: 10.1186/s12974-023-02695-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 01/11/2023] [Indexed: 01/31/2023] Open
Abstract
BACKGROUND The presence of meningeal ectopic lymphoid structures (ELS) in a subgroup of patients diagnosed with secondary progressive multiple sclerosis (SPMS) corresponds to a pronounced cortical inflammation and an aggravated disease course. In MP4-induced experimental autoimmune encephalomyelitis (EAE), a mouse model of multiple sclerosis (MS), B cell aggregates develop in the central nervous system (CNS) in the chronic stage of the disease. Therefore, the model is suitable for studying key molecules of ELS development and maintenance. Here, we investigated whether there is a specific cytokine and chemokine signature in paired cerebrospinal fluid (CSF) and serum samples associated with the presence of cerebellar B cell and T cell pathology and B cell aggregates of MP4-immunized mice. METHODS Paired CSF and serum samples were collected from the cisterna magna and periphery of MP4-immunized mice at the chronic stage of disease. A control group with mice immunized only with the adjuvant (vehicle) was included in the study. A selected panel of 34 cytokines and chemokines were measured by MAGPIX® for both cohorts. For the assessment of B cell and T cell infiltration, immunohistochemical staining was performed and analyzed using light microscopy. To detect specific chemokine receptors additional staining was conducted. RESULTS While we detected several upregulated cytokines and chemokines in the CSF of MP4-immunized mice independent of the extent of B cell and T cell pathology compared to vehicle-immunized mice, C-C motif chemokine ligand (CCL)-1 was associated with high B cell and T cell infiltration. Furthermore, the level of certain chemokines, including CCL1, CCL5, CCL7, CCL12, CCL22 and C-X-C motif chemokine ligand (CXCL)-13, was significantly increased (p < 0.05) in MP4-immunized mice showing a high number of B cell aggregates. While C-C motif chemokine receptor (CCR)5 had a ubiquitous expression independent of the extent of B cell and T cell pathology, C-X-C motif chemokine receptor (CXCR)-5 and CXCR6 expression was specifically associated with high B cell and T cell pathology. CONCLUSION Our data suggest that multiple cytokines and chemokines are involved in the pathophysiology of MP4-induced EAE. Furthermore, the presence of B cell aggregates was associated with a specific chemokine profile in the CSF, which might be useful for predicting the presence of these aggregates without the necessity to histologically screen the CNS tissue.
Collapse
Affiliation(s)
- Verena Schropp
- grid.10388.320000 0001 2240 3300Medical Faculty, Institute of Neuroanatomy, University of Bonn, 53115 Bonn, Germany ,grid.5330.50000 0001 2107 3311Institute of Anatomy and Cell Biology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
| | - Rittika Chunder
- grid.10388.320000 0001 2240 3300Medical Faculty, Institute of Neuroanatomy, University of Bonn, 53115 Bonn, Germany ,grid.5330.50000 0001 2107 3311Institute of Anatomy and Cell Biology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
| | - Barbara Dietel
- grid.5330.50000 0001 2107 3311Department of Cardiology and Angiology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen University Hospital, 91054 Erlangen, Germany
| | - Sabine Tacke
- grid.5330.50000 0001 2107 3311Institute of Anatomy and Cell Biology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
| | - Stefanie Kuerten
- grid.10388.320000 0001 2240 3300Medical Faculty, Institute of Neuroanatomy, University of Bonn, 53115 Bonn, Germany ,grid.5330.50000 0001 2107 3311Institute of Anatomy and Cell Biology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
| |
Collapse
|
44
|
Julé AM, Lam KP, Taylor M, Hoyt KJ, Wei K, Gutierrez-Arcelus M, Case SM, Chandler M, Chang MH, Cohen EM, Dedeoglu F, Halyabar O, Hausmann J, Hazen MM, Janssen E, Lo J, Lo MS, Meidan E, Roberts JE, Wobma H, Son MBF, Sundel RP, Lee PY, Sage PT, Chatila TA, Nigrovic PA, Rao DA, Henderson LA. Disordered T cell-B cell interactions in autoantibody-positive inflammatory arthritis. Front Immunol 2023; 13:1068399. [PMID: 36685593 PMCID: PMC9849554 DOI: 10.3389/fimmu.2022.1068399] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 12/12/2022] [Indexed: 01/06/2023] Open
Abstract
T peripheral helper (Tph) cells, identified in the synovium of adults with seropositive rheumatoid arthritis, drive B cell maturation and antibody production in non-lymphoid tissues. We sought to determine if similarly dysregulated T cell-B cell interactions underlie another form of inflammatory arthritis, juvenile oligoarthritis (oligo JIA). Clonally expanded Tph cells able to promote B cell antibody production preferentially accumulated in the synovial fluid (SF) of oligo JIA patients with antinuclear antibodies (ANA) compared to autoantibody-negative patients. Single-cell transcriptomics enabled further definition of the Tph gene signature in inflamed tissues and showed that Tph cells from ANA-positive patients upregulated genes associated with B cell help to a greater extent than patients without autoantibodies. T cells that co-expressed regulatory T and B cell-help factors were identified. The phenotype of these Tph-like Treg cells suggests an ability to restrain T cell-B cell interactions in tissues. Our findings support the central role of disordered T cell-help to B cells in autoantibody-positive arthritides.
Collapse
Affiliation(s)
- Amélie M. Julé
- Division of Immunology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Ki Pui Lam
- Division of Immunology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Maria Taylor
- Division of Immunology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Kacie J. Hoyt
- Division of Immunology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Kevin Wei
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Maria Gutierrez-Arcelus
- Division of Immunology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, United States
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, United States
| | - Siobhan M. Case
- Division of Immunology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, United States
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Mia Chandler
- Division of Immunology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Margaret H. Chang
- Division of Immunology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, United States
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Ezra M. Cohen
- Division of Immunology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, United States
- Division of Rheumatology, Boston Medical Center, Boston University School of Medicine, Boston, MA, United States
| | - Fatma Dedeoglu
- Division of Immunology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Olha Halyabar
- Division of Immunology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Jonathan Hausmann
- Division of Immunology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Melissa M. Hazen
- Division of Immunology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Erin Janssen
- Division of Immunology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Jeffrey Lo
- Division of Immunology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Mindy S. Lo
- Division of Immunology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Esra Meidan
- Division of Immunology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Jordan E. Roberts
- Division of Immunology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Holly Wobma
- Division of Immunology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Mary Beth F. Son
- Division of Immunology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Robert P. Sundel
- Division of Immunology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Pui Y. Lee
- Division of Immunology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, United States
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Peter T. Sage
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Talal A. Chatila
- Division of Immunology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Peter A. Nigrovic
- Division of Immunology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, United States
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Deepak A. Rao
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Lauren A. Henderson
- Division of Immunology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
45
|
CHEN J, CHEN J, WANG L. Tertiary lymphoid structures as unique constructions associated with the organization, education, and function of tumor-infiltrating immunocytes. J Zhejiang Univ Sci B 2022; 23:812-822. [PMID: 36226536 PMCID: PMC9561406 DOI: 10.1631/jzus.b2200174] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Tertiary lymphoid structures (TLSs) are formations at sites with persistent inflammatory stimulation, including tumors. These ectopic lymphoid organs mainly consist of chemo-attracting B cells, T cells, and supporting dendritic cells (DCs). Mature TLSs exhibit functional organization for the optimal development and collaboration of adaptive immune response, delivering an augmented effect on the tumor microenvironment (TME). The description of the positive correlation between TLSs and tumor prognosis is reliable only under a certain condition involving the localization and maturation of TLSs. Emerging evidence suggests that underlying mechanisms of the anti-tumor effect of TLSs pave the way for novel immunotherapies. Several approaches have been developed to take advantage of intratumoral TLSs, either by combining it with therapeutic agents or by inducing the neogenesis of TLSs.
Collapse
Affiliation(s)
- Jing CHEN
- Department of Gastrointestinal Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou310009, China,Institute of Immunology and Bone Marrow Transplantation Center, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou310003, China
| | - Jian CHEN
- Department of Gastrointestinal Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou310009, China,Jian CHEN,
| | - Lie WANG
- Institute of Immunology and Bone Marrow Transplantation Center, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou310003, China,Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou311121, China,Cancer Center, Zhejiang University, Hangzhou310058, China,Lie WANG,
| |
Collapse
|
46
|
FDCSP Is an Immune-Associated Prognostic Biomarker in HPV-Positive Head and Neck Squamous Carcinoma. Biomolecules 2022; 12:biom12101458. [PMID: 36291667 PMCID: PMC9599724 DOI: 10.3390/biom12101458] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 10/05/2022] [Accepted: 10/09/2022] [Indexed: 12/24/2022] Open
Abstract
Background: Head and neck squamous carcinoma (HNSC) poses a major threat to human life. The role of human papillomavirus (HPV) infection in the initiation and progression of HNSC is becoming more widely accepted. HPV-positive (HPV+) HNSC has shown unique responses to cancer therapies, which may be due to differences in immune cell infiltration. It is critical to determine how the immune responses to HPV in HNSC are regulated. Methods: Transcriptome data of HNSC from The Cancer Genome Atlas (TCGA) and the Gene Expression Omnibus (GEO) database were analyzed. Then, the CIBERSORT algorithm was used to calculate immune cell infiltration in HNSC. FDCSP expression level was detected by qPCR in the HNSC tissues collected from the Nanfang Hospital. Results: Follicular dendritic cell secreted protein (FDCSP) was highly expressed in HPV+ HNSC, and higher expression of FDSCP was associated with a favorable prognosis. In HPV+ HNSC samples, FDCSP significantly increased the proportion of T follicular helper cells (TFHs). FDCSP expression was also found to be associated with TP53 mutation status in HPV+ HNSC. The function of FDCSP was intimately connected to chemokine pathways, particularly with the C-X-C motif chemokine ligand 13 (CXCL13). We verified that the high expression of FDCSP in HPV+ HNSC and higher FDCSP is closely related to prognosis in HNSC samples we collected by qPCR. Conclusions: Collectively, these findings may provide fresh evidence that FDCSP is a potential chemokine-associated prognostic biomarker in HPV+ HNSC.
Collapse
|
47
|
Beider K, Voevoda-Dimenshtein V, Zoabi A, Rosenberg E, Magen H, Ostrovsky O, Shimoni A, Weiss L, Abraham M, Peled A, Nagler A. CXCL13 chemokine is a novel player in multiple myeloma osteolytic microenvironment, M2 macrophage polarization, and tumor progression. J Hematol Oncol 2022; 15:144. [PMID: 36217194 PMCID: PMC9549634 DOI: 10.1186/s13045-022-01366-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 10/02/2022] [Indexed: 12/04/2022] Open
Abstract
Background We assessed the mechanism by which multiple myeloma (MM) shapes the bone marrow (BM) microenvironment and affects MΦ polarization. Methods In vivo xenograft model of BM-disseminated human myeloma, as well as analysis of MM cell lines, stromal components, and primary samples from patients with MM, was utilized. Results Analysis of the BM from MM-bearing mice inoculated with human CXCR4-expressing RPMI8226 cells revealed a significant increase in M2 MΦ cell numbers (p < 0.01). CXCL13 was one of the most profoundly increased factors upon MM growth with increased levels in the blood of MM-bearing animals. Myeloid cells were the main source of the increased murine CXCL13 detected in MM-infiltrated BM. MM cell lines induced CXCL13 and concurrent expression of M2 markers (MERTK, CD206, CD163) in co-cultured human MΦ in vitro. Interaction with MΦ reciprocally induced CXCL13 expression in MM cell lines. Mechanistically, TGFβ signaling was involved in CXCL13 induction in MM cells, while BTK signaling was implicated in MM-stimulated increase of CXCL13 in MΦ. Recombinant CXCL13 increased RANKL expression and induced TRAP+ osteoclast (OC) formation in vitro, while CXCL13 neutralization blocked these activities. Moreover, mice inoculated with CXCL13-silenced MM cells developed significantly lower BM disease. Reduced tumor load correlated with decreased numbers of M2 MΦ in BM, decreased bone disease, and lower expression of OC-associated genes. Finally, higher levels of CXCL13 were detected in the blood and BM samples of MM patients in comparison with healthy individuals. Conclusions Altogether, our findings suggest that bidirectional interactions of MΦ with MM tumor cells result in M2 MΦ polarization, CXCL13 induction, and subsequent OC activation, enhancing their ability to support bone resorption and MM progression. CXCL13 may thus serve as a potential novel target in MM. Supplementary Information The online version contains supplementary material available at 10.1186/s13045-022-01366-5.
Collapse
Affiliation(s)
- Katia Beider
- Division of Hematology and CBB, Chaim Sheba Medical Center, Tel Aviv University, Tel-Hashomer, Israel
| | | | - Ali Zoabi
- Division of Hematology and CBB, Chaim Sheba Medical Center, Tel Aviv University, Tel-Hashomer, Israel
| | - Evgenia Rosenberg
- Division of Hematology and CBB, Chaim Sheba Medical Center, Tel Aviv University, Tel-Hashomer, Israel
| | - Hila Magen
- Division of Hematology and CBB, Chaim Sheba Medical Center, Tel Aviv University, Tel-Hashomer, Israel
| | - Olga Ostrovsky
- Division of Hematology and CBB, Chaim Sheba Medical Center, Tel Aviv University, Tel-Hashomer, Israel
| | - Avichai Shimoni
- Division of Hematology and CBB, Chaim Sheba Medical Center, Tel Aviv University, Tel-Hashomer, Israel
| | - Lola Weiss
- Goldyne Savad Institute of Gene Therapy, Hebrew University Hospital, Jerusalem, Israel
| | - Michal Abraham
- Goldyne Savad Institute of Gene Therapy, Hebrew University Hospital, Jerusalem, Israel
| | - Amnon Peled
- Goldyne Savad Institute of Gene Therapy, Hebrew University Hospital, Jerusalem, Israel
| | - Arnon Nagler
- Division of Hematology and CBB, Chaim Sheba Medical Center, Tel Aviv University, Tel-Hashomer, Israel.
| |
Collapse
|
48
|
Ullah A, Pervaz S, Adu-Gyamfi EA, Czika A, Guo M, Wang MJ, Wang YX. CXCL13 and CXCR5 are upregulated in PCOS mice ovaries but downregulated following metformin administration. Mol Cell Endocrinol 2022; 556:111730. [PMID: 35921919 DOI: 10.1016/j.mce.2022.111730] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Revised: 07/19/2022] [Accepted: 07/22/2022] [Indexed: 10/16/2022]
Abstract
Polycystic ovary syndrome (PCOS) is becoming a common pathology among women, yet its pathogenesis remains enigmatic. The chemokine C-X-C motif ligand 13 (CXCL13) and its receptor type 5 (CXCR5) regulate inflammatory responses but their roles in PCOS remain unknown. Metformin is commonly administered to PCOS patients but its mechanism of action remains unclear. Thus, we aimed to determine the expression of CXCL13 and CXCR5 in the ovaries of PCOS mice and to evaluate the therapeutic effect of metformin on them. The study comprised four groups of mice: control, PCOS, PCOS plus metformin, and PCOS plus vehicle. CXCL13 and CXCR5 were found to be elevated in the ovarian tissues of the PCOS mice. Metformin reduced ovarian CXCL13 and CXCR5 expressions in the PCOS mice. Hence, CXCL13 and CXCR5 are potentially involved in PCOS pathogenesis; and metformin may help alleviate the symptoms of PCOS by inhibiting CXCL13 expression and actions.
Collapse
Affiliation(s)
- Amin Ullah
- Department of Reproductive Sciences, School of Public Health, Chongqing Medical University, Chongqing, People's Republic of China; Joint International Research Laboratory of Reproduction and Development, School of Public Health, Chongqing Medical University, Chongqing, People's Republic of China
| | - Sadaf Pervaz
- Department of Reproductive Sciences, School of Public Health, Chongqing Medical University, Chongqing, People's Republic of China; Joint International Research Laboratory of Reproduction and Development, School of Public Health, Chongqing Medical University, Chongqing, People's Republic of China
| | - Enoch Appiah Adu-Gyamfi
- Department of Reproductive Sciences, School of Public Health, Chongqing Medical University, Chongqing, People's Republic of China; Joint International Research Laboratory of Reproduction and Development, School of Public Health, Chongqing Medical University, Chongqing, People's Republic of China
| | - Armin Czika
- Department of Reproductive Sciences, School of Public Health, Chongqing Medical University, Chongqing, People's Republic of China; Joint International Research Laboratory of Reproduction and Development, School of Public Health, Chongqing Medical University, Chongqing, People's Republic of China; Faculty of Medicine, Transilvania University of Brasov, Brasov, Romania
| | - Man Guo
- Department of Physiology of School of Basic Medicine, Chongqing Medical University, Chongqing, People's Republic of China
| | - Mei-Jiao Wang
- Joint International Research Laboratory of Reproduction and Development, School of Public Health, Chongqing Medical University, Chongqing, People's Republic of China; Department of Physiology of School of Basic Medicine, Chongqing Medical University, Chongqing, People's Republic of China.
| | - Ying-Xiong Wang
- Department of Reproductive Sciences, School of Public Health, Chongqing Medical University, Chongqing, People's Republic of China; Joint International Research Laboratory of Reproduction and Development, School of Public Health, Chongqing Medical University, Chongqing, People's Republic of China.
| |
Collapse
|
49
|
Charmetant X, Chen CC, Hamada S, Goncalves D, Saison C, Rabeyrin M, Rabant M, Duong van Huyen JP, Koenig A, Mathias V, Barba T, Lacaille F, le Pavec J, Brugière O, Taupin JL, Chalabreysse L, Mornex JF, Couzi L, Graff-Dubois S, Jeger-Madiot R, Tran-Dinh A, Mordant P, Paidassi H, Defrance T, Morelon E, Badet L, Nicoletti A, Dubois V, Thaunat O. Inverted direct allorecognition triggers early donor-specific antibody responses after transplantation. Sci Transl Med 2022; 14:eabg1046. [PMID: 36130013 DOI: 10.1126/scitranslmed.abg1046] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The generation of antibodies against donor-specific major histocompatibility complex (MHC) antigens, a type of donor-specific antibodies (DSAs), after transplantation requires that recipient's allospecific B cells receive help from T cells. The current dogma holds that this help is exclusively provided by the recipient's CD4+ T cells that recognize complexes of recipient's MHC II molecules and peptides derived from donor-specific MHC alloantigens, a process called indirect allorecognition. Here, we demonstrated that, after allogeneic heart transplantation, CD3ε knockout recipient mice lacking T cells generate a rapid, transient wave of switched alloantibodies, predominantly directed against MHC I molecules. This is due to the presence of donor CD4+ T cells within the graft that recognize intact recipient's MHC II molecules expressed by B cell receptor-activated allospecific B cells. Indirect evidence suggests that this inverted direct pathway is also operant in patients after transplantation. Resident memory donor CD4+ T cells were observed in perfusion liquids of human renal and lung grafts and acquired B cell helper functions upon in vitro stimulation. Furthermore, T follicular helper cells, specialized in helping B cells, were abundant in mucosa-associated lymphoid tissue of lung and intestinal grafts. In the latter, more graft-derived passenger T cells correlated with the detection of donor T cells in recipient's circulation; this, in turn, was associated with an early transient anti-MHC I DSA response and worse transplantation outcomes. We conclude that this inverted direct allorecognition is a possible explanation for the early transient anti-MHC DSA responses frequently observed after lung or intestinal transplantations.
Collapse
Affiliation(s)
- Xavier Charmetant
- CIRI, Centre International de Recherche en Infectiologie, Université de Lyon, INSERM U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, 69007 Lyon, France
| | - Chien-Chia Chen
- Department of Surgery, National Taiwan University Hospital, Taipei 100, Taiwan
| | - Sarah Hamada
- French National Blood Service (EFS), HLA Laboratory, 69150 Décines, France
| | - David Goncalves
- CIRI, Centre International de Recherche en Infectiologie, Université de Lyon, INSERM U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, 69007 Lyon, France
| | - Carole Saison
- French National Blood Service (EFS), HLA Laboratory, 69150 Décines, France
| | - Maud Rabeyrin
- Department of Pathology, Hospices Civils de Lyon, Groupement Hospitalier Est, 69500 Bron, France
| | - Marion Rabant
- Pathology Department, Assistance Publique-Hôpitaux de Paris, Hôpital Necker, 75015 Paris, France
| | | | - Alice Koenig
- CIRI, Centre International de Recherche en Infectiologie, Université de Lyon, INSERM U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, 69007 Lyon, France
- Lyon-Est Medical Faculty, Claude Bernard University (Lyon 1), 69008 Lyon, France
- Department of Transplantation, Nephrology and Clinical Immunology, Hospices Civils de Lyon, Edouard Herriot Hospital, 69003 Lyon, France
| | - Virginie Mathias
- French National Blood Service (EFS), HLA Laboratory, 69150 Décines, France
| | - Thomas Barba
- CIRI, Centre International de Recherche en Infectiologie, Université de Lyon, INSERM U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, 69007 Lyon, France
| | - Florence Lacaille
- Pediatric Gastroenterology-Hepatology-Nutrition Unit, Hôpital Universitaire Necker-Enfants malades, 75015 Paris, France
| | - Jérôme le Pavec
- Department of Pulmonology and Lung Transplantation, Marie Lannelongue Hospital, 92350 Le Plessis Robinson, France
| | - Olivier Brugière
- Pulmonology Department, Adult Cystic Fibrosis Centre and Lung Transplantation Department, Foch Hospital, 92150 Suresnes, France
| | - Jean-Luc Taupin
- Laboratory of Immunology and Histocompatibility, Hôpital Saint-Louis APHP, 75010 Paris, France
- INSERM U976 Institut de Recherche Saint-Louis, Université Paris Diderot, 75010 Paris, France
| | - Lara Chalabreysse
- Department of Pathology, Hospices Civils de Lyon, Groupement Hospitalier Est, 69500 Bron, France
| | - Jean-François Mornex
- Université de Lyon, Université Lyon 1, INRAE, IVPC, UMR754, 69000 Lyon, France
- Department of Pneumology, GHE, Hospices Civils de Lyon, 69000 Lyon, France
| | - Lionel Couzi
- Department of Nephrology, Transplantation, Dialysis, Apheresis, Pellegrin Hospital, 33000 Bordeaux, France
| | - Stéphanie Graff-Dubois
- Sorbonne Université, INSERM, Immunology-Immunopathology-Immunotherapy (i3), 75013 Paris, France
| | - Raphaël Jeger-Madiot
- Sorbonne Université, INSERM, Immunology-Immunopathology-Immunotherapy (i3), 75013 Paris, France
| | - Alexy Tran-Dinh
- Université de Paris, LVTS, INSERM U1148, 75018 Paris, France
| | - Pierre Mordant
- Department of Vascular and Thoracic Surgery, Assistance Publique-Hôpitaux de Paris, Bichat-Claude Bernard Hospital, 75018 Paris, France
| | - Helena Paidassi
- CIRI, Centre International de Recherche en Infectiologie, Université de Lyon, INSERM U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, 69007 Lyon, France
| | - Thierry Defrance
- CIRI, Centre International de Recherche en Infectiologie, Université de Lyon, INSERM U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, 69007 Lyon, France
| | - Emmanuel Morelon
- CIRI, Centre International de Recherche en Infectiologie, Université de Lyon, INSERM U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, 69007 Lyon, France
- Lyon-Est Medical Faculty, Claude Bernard University (Lyon 1), 69008 Lyon, France
- Department of Transplantation, Nephrology and Clinical Immunology, Hospices Civils de Lyon, Edouard Herriot Hospital, 69003 Lyon, France
| | - Lionel Badet
- Lyon-Est Medical Faculty, Claude Bernard University (Lyon 1), 69008 Lyon, France
- Department of Urology and Transplantation Surgery, Hospices Civils de Lyon, Edouard Herriot Hospital, 69003 Lyon, France
| | | | - Valérie Dubois
- French National Blood Service (EFS), HLA Laboratory, 69150 Décines, France
| | - Olivier Thaunat
- CIRI, Centre International de Recherche en Infectiologie, Université de Lyon, INSERM U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, 69007 Lyon, France
- Lyon-Est Medical Faculty, Claude Bernard University (Lyon 1), 69008 Lyon, France
- Department of Transplantation, Nephrology and Clinical Immunology, Hospices Civils de Lyon, Edouard Herriot Hospital, 69003 Lyon, France
| |
Collapse
|
50
|
Wang B, Wang M, Ao D, Wei X. CXCL13-CXCR5 axis: Regulation in inflammatory diseases and cancer. Biochim Biophys Acta Rev Cancer 2022; 1877:188799. [PMID: 36103908 DOI: 10.1016/j.bbcan.2022.188799] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Revised: 09/06/2022] [Accepted: 09/06/2022] [Indexed: 01/10/2023]
Abstract
Chemokine C-X-C motif ligand 13 (CXCL13), originally identified as a B-cell chemokine, plays an important role in the immune system. The interaction between CXCL13 and its receptor, the G-protein coupled receptor (GPCR) CXCR5, builds a signaling network that regulates not only normal organisms but also the development of many diseases. However, the precise action mechanism remains unclear. In this review, we discussed the functional mechanisms of the CXCL13-CXCR5 axis under normal conditions, with special focus on its association with diseases. For certain refractory diseases, we emphasize the diagnostic and therapeutic role of CXCL13-CXCR5 axis.
Collapse
Affiliation(s)
- Binhan Wang
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Manni Wang
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Danyi Ao
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Xiawei Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|