1
|
Chang ML, Cheng JS, Chen WT, Shen YJ, Kuo CJ, Chien RN. Mixed cryoglobulinemia decelerates hepatocellular carcinoma development in hepatitis C patients with SVR by downregulating regulatory B cells: a 12-year prospective cohort study. Oncoimmunology 2025; 14:2470128. [PMID: 40008547 PMCID: PMC11866965 DOI: 10.1080/2162402x.2025.2470128] [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: 12/25/2024] [Revised: 02/14/2025] [Accepted: 02/14/2025] [Indexed: 02/27/2025] Open
Abstract
How mixed cryoglobulinemia (MC) affects cancer risk in chronic hepatitis C patients with sustained virologic response (SVR) remains unclear. In a 12-year prospective study, post-SVR MC was assessed every 3‒6 months. Among the 891 SVR patients, 265 (29.7%) had baseline (24 weeks after completing anti-HCV therapy) MC, and the 12-year cancer cumulative incidence was 19.7%. Among the 73 patients who developed cancer, 37 (50.7%) had hepatocellular carcinoma (HCC), with the following associated baseline variables: for cancer, male sex, age and alanine aminotransferase (ALT) levels; for HCC, male sex, age, and cirrhosis; and for non-HCC cancer, rheumatoid factor levels. Among patients with post-SVR HCC, the mean time to HCC was longer in those with than in those without baseline MC (1545.4 ± 276.5 vs. 856.9 ± 115.2 days, p = 0.014). Patients with baseline MC had decreased circulating interleukin-10 (IL-10)-positive B cell (CD19+IL-10+cells/CD19+cells) (31.24 ± 16.14 vs. 40.08 ± 15.42%, p = 0.031), regulatory B cell (Breg) (CD19+CD24hi CD27+cells/CD19+cells) (10.45 ± 7.10 vs. 15.76 ± 9.14%, p = 0.035), IL-10-positive Breg (CD19+CD24hiCD27+IL-10+cells/CD19+cells) (5.06 ± 4.68 vs. 8.83 ± 5.46%, p = 0.015) and HCC-infiltrating Breg (18.6 ± 10 vs. 33.51 ± 6.8%, p = 0.022) ratios but comparable circulating and HCC-infiltrating regulatory T cell ratios relative to patients without baseline MC. In conclusion, old male SVR patients with elevated ALT levels or cirrhosis require intensive monitoring for cancer development, especially HCC. Tailored HCC follow-up is needed for SVR patients according to their baseline MC, which might downregulate Bregs to decelerate HCC development for almost 2 years.
Collapse
MESH Headings
- Humans
- Male
- Carcinoma, Hepatocellular/immunology
- Carcinoma, Hepatocellular/virology
- Carcinoma, Hepatocellular/etiology
- Carcinoma, Hepatocellular/pathology
- Carcinoma, Hepatocellular/epidemiology
- Cryoglobulinemia/immunology
- Cryoglobulinemia/complications
- Female
- Liver Neoplasms/immunology
- Liver Neoplasms/virology
- Liver Neoplasms/etiology
- Liver Neoplasms/pathology
- Liver Neoplasms/epidemiology
- Middle Aged
- Prospective Studies
- B-Lymphocytes, Regulatory/immunology
- Hepatitis C, Chronic/drug therapy
- Hepatitis C, Chronic/complications
- Hepatitis C, Chronic/virology
- Hepatitis C, Chronic/immunology
- Sustained Virologic Response
- Aged
- Interleukin-10
- Antiviral Agents/therapeutic use
- Adult
- Hepacivirus
Collapse
Affiliation(s)
- Ming-Ling Chang
- Department of Gastroenterology and Hepatology, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
- Department of Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Jur-Shan Cheng
- Department of Gastroenterology and Hepatology, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
- Clinical Informatics and Medical Statistics Research Center, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Wei-Ting Chen
- Department of Gastroenterology and Hepatology, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - Yi-Jyun Shen
- Department of Gastroenterology and Hepatology, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - Chia-Jung Kuo
- Department of Gastroenterology and Hepatology, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - Rong-Nan Chien
- Department of Gastroenterology and Hepatology, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
- Department of Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| |
Collapse
|
2
|
Tan L, Mo Z, Gan W, Gao Z, Zhu J, Wu Z. The remodeling of B-cell subsets was correlated with the clearance of hepatitis B antigen during pegylated IFN α-2a therapy in CHB patients. Ann Med 2025; 57:2463569. [PMID: 39957563 PMCID: PMC11834791 DOI: 10.1080/07853890.2025.2463569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 04/19/2024] [Accepted: 06/01/2024] [Indexed: 02/18/2025] Open
Abstract
BACKGROUND B-cell may participate in the cellular immune process of hepatitis B antigen clearance. However, the function and specific mechanism of B-cell during interferon-pegylated interferon α-2a (Peg-IFN-α) treatment in chronic hepatitis B (CHB) patients have not yet been described. METHODS A total of 150 CHB patients enrolled in this study, who received 48 weeks of Peg-IFN α treatment. The differentiation clusters CD19, CD24, CD27, CD38, CD40, and CD80 of B cell surface markers in CHB patients were detected by flow cytometry. Spearman correlation and Logistic regression analysis were performed for the analysis. RESULTS The clearance rate of HBsAg increased significantly with the duration of Peg-IFN-α treatment, reaching 32.2% by 48 weeks. During the Peg-IFN-α therapy, the frequency of B-cell and its subsets increased significantly. However, we did not observe any significant difference in the frequency of the B-cell and its subsets in patients with or without HBsAg clearance after 48 weeks Peg-IFN-α treatment. The change in HBsAg value was negatively related to the change in plasmablasts (CD19+CD38+) level before and after 48 weeks treatment (r = -0.326, p = 0.006). Moreover, the results showed that HBsAg <288.70 IU/mL at baseline and HBsAg <58.05 IU/mL at 12 weeks were strong predictors of HBsAg clearance in patients with 48 weeks Peg-IFN-α treatment. CONCLUSION The remodeling of B cell subsets, especially plasmablasts (CD19+CD38+), during Peg-IFN-α treatment was closed associated with the clearance of hepatitis B antigen.
Collapse
Affiliation(s)
- Lei Tan
- Department of Medical Ultrasonic, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Zhishuo Mo
- Department of Infectious Diseases, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
- Key Laboratory of Topical Disease Control, Sun Yat-sen University, Ministry of Education, Guangzhou, Guangdong, China
| | - Weiqiang Gan
- Department of Infectious Diseases, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
- Key Laboratory of Topical Disease Control, Sun Yat-sen University, Ministry of Education, Guangzhou, Guangdong, China
| | - Zhiliang Gao
- Department of Infectious Diseases, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
- Key Laboratory of Topical Disease Control, Sun Yat-sen University, Ministry of Education, Guangzhou, Guangdong, China
| | - Jianyun Zhu
- Department of Infectious Diseases, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
- Key Laboratory of Topical Disease Control, Sun Yat-sen University, Ministry of Education, Guangzhou, Guangdong, China
| | - Zeqian Wu
- Department of Infectious Diseases, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
- Key Laboratory of Topical Disease Control, Sun Yat-sen University, Ministry of Education, Guangzhou, Guangdong, China
| |
Collapse
|
3
|
Sun F, Gao X, Li T, Zhao X, Zhu Y. Tumor immune microenvironment remodeling after neoadjuvant therapy in gastric cancer: Update and new challenges. Biochim Biophys Acta Rev Cancer 2025; 1880:189350. [PMID: 40355011 DOI: 10.1016/j.bbcan.2025.189350] [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: 11/23/2024] [Revised: 05/05/2025] [Accepted: 05/08/2025] [Indexed: 05/14/2025]
Abstract
Gastric cancer (GC) is a malignant tumor with one of the highest morbidity and death rates in the world. Neoadjuvant therapy, including neoadjuvant chemotherapy (NAC) and NAC combined with immunotherapy, can improve the resection and long-term survival rates. However, not all patients respond well to neoadjuvant therapy. It has been confirmed that immune cells in the tumor immune microenvironment, including T cells, B cells, and natural killer cells, can affect the efficacy of neoadjuvant therapy. This paper summarizes current preclinical and clinical evidence to more fully describe the effects of neoadjuvant therapy on the immune microenvironment of GC, to provide the impetus to identify biomarkers to predict the potency of neoadjuvant therapy, and to identify the mechanisms of drug resistance, which should promote the development of individualized and accurate treatments for GC patients.
Collapse
Affiliation(s)
- Fujing Sun
- Department of Pathology, Affiliated Cancer Hospital of Dalian University of Technology (Liaoning Cancer Hospital and Institute, Cancer Hospital of China Medical University), Shenyang, China
| | - Xiaozhuo Gao
- Department of Pathology, Affiliated Cancer Hospital of Dalian University of Technology (Liaoning Cancer Hospital and Institute, Cancer Hospital of China Medical University), Shenyang, China
| | - Tianming Li
- Department of Pathology, Affiliated Cancer Hospital of Dalian University of Technology (Liaoning Cancer Hospital and Institute, Cancer Hospital of China Medical University), Shenyang, China
| | - Xiaoyan Zhao
- Graduate School, Dalian Medical University, Dalian, China
| | - Yanmei Zhu
- Department of Pathology, Affiliated Cancer Hospital of Dalian University of Technology (Liaoning Cancer Hospital and Institute, Cancer Hospital of China Medical University), Shenyang, China.
| |
Collapse
|
4
|
McNee A, Kannan A, Jull P, Shankar S. Expanding Human Breg for Cellular Therapy in Transplantation: Time for Translation. Transplantation 2025; 109:926-937. [PMID: 39439021 PMCID: PMC12091222 DOI: 10.1097/tp.0000000000005243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 07/24/2024] [Accepted: 08/13/2024] [Indexed: 10/25/2024]
Abstract
Regulatory B cells (Breg) are instrumental in protecting allografts in transplantation. Breg signatures are identified in operationally tolerant human kidney transplant recipients and can predict organ survival and acute rejection. Animal models of transplantation and autoimmunity support the use of Breg as an adoptive cellular therapy. Detailed mechanistic studies have identified multiple signaling pathways utilized by Breg in their induction, expansion, and downstream function. These preclinical studies provide the guiding principles, which will inform protocols by which to expand this crucial immunoregulatory population before clinical use. There is an urgent need for novel therapies to improve long-term transplant outcomes and to minimize immunosuppression-related morbidity including life-threatening infection and cancer. Systematic evaluation of the signals, which drive Breg expansion, will be key to transforming the as of yet unharnessed potential of this potent immunoregulatory cell. In this review, we explore the potential avenues of translating Breg subsets from cell culture at the laboratory bench to cell therapy at the patient's bedside. We will discuss the standardization of Breg phenotypes to aid in precursor population selection and quality control of a Breg-cell therapy product. We will evaluate avenues by which to optimize protocols to drive human Breg expansion to levels sufficient for cellular therapy. Finally, we will examine the steps required in process development including scalable culture systems and quality control measures to deliver a viable Breg-cell therapy product for administration to a transplant recipient.
Collapse
Affiliation(s)
- Adam McNee
- Nuffield Department of Surgical Sciences, University of Oxford, John Radcliffe Hospital, Headington, Oxford, United Kingdom
| | - Ananya Kannan
- Oxford University Medical School, John Radcliffe Hospital, Headington, Oxford, United Kingdom
| | - Patrick Jull
- Nuffield Department of Surgical Sciences, University of Oxford, John Radcliffe Hospital, Headington, Oxford, United Kingdom
| | - Sushma Shankar
- Nuffield Department of Surgical Sciences, University of Oxford, John Radcliffe Hospital, Headington, Oxford, United Kingdom
| |
Collapse
|
5
|
Hu H, Zhang G, Chen T, Liu Y, Meng L, Holmdahl R, Dai L, Zhao Y. Immunosenescence in autoimmune diseases. Autoimmun Rev 2025; 24:103805. [PMID: 40132774 DOI: 10.1016/j.autrev.2025.103805] [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: 02/04/2025] [Revised: 03/21/2025] [Accepted: 03/21/2025] [Indexed: 03/27/2025]
Abstract
Autoimmune diseases (AIDs) are a group of disorders in which the immune system mistakenly attacks the body's own tissues, characterized by the loss of tolerance to self-antigens and destruction of tissues. Aging is a natural process of physiological decline that also alters the immune system, a condition known as immunosenescence. During immunosenescence, the immune system undergoes various changes, including modifications and antigenicity of self-antigens, abnormalities in the quantity, phenotype, and function of lymphocytes and antibodies, as well as a narrowing of the B and T cell receptor repertoire, changes that may increase susceptibility to AIDs. Additionally, senescent immune cells and the senescence-associated secretory phenotype (SASP) contribute to target organ involvement in AIDs, exacerbating chronic inflammation and tissue damage. Mitochondrial dysfunction and metabolic imbalances in AIDs lead to the accumulation of senescent cells, which act as upstream drivers of immunosenescence. In this review, we summarize the bidirectional relationship between AIDs and immunosenescence, as well as its potential mechanisms. Therapeutic approaches targeting immunosenescence in AIDs remain at an early stage. Strategies aimed at resetting or reversing the aging immune system are expected to become a novel direction in the future.
Collapse
Affiliation(s)
- Huifang Hu
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, 610041 Chengdu, Sichuan, China; Clinical Institute of Inflammation and Immunology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, 610041 Chengdu, Sichuan, China
| | - Guangyue Zhang
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, 610041 Chengdu, Sichuan, China; Clinical Institute of Inflammation and Immunology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, 610041 Chengdu, Sichuan, China
| | - Tao Chen
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, 610041 Chengdu, Sichuan, China; Clinical Institute of Inflammation and Immunology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, 610041 Chengdu, Sichuan, China
| | - Yi Liu
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, 610041 Chengdu, Sichuan, China; Clinical Institute of Inflammation and Immunology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, 610041 Chengdu, Sichuan, China
| | - Liesu Meng
- Department of Rheumatology, and National Joint Engineering Research Center of Biodiagnostics and Biotherapy, Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, PR China
| | - Rikard Holmdahl
- Medical Inflammation Research, Division of Immunology, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
| | - Lunzhi Dai
- National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, 610041 Chengdu, Sichuan, China.
| | - Yi Zhao
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, 610041 Chengdu, Sichuan, China; Clinical Institute of Inflammation and Immunology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, 610041 Chengdu, Sichuan, China.
| |
Collapse
|
6
|
Lu X, Han YD, Zu XR, Huang JC, Li L, Wang M, Wang YT, Guo LL, Zhou L, Han Y. Rapamycin-modified CD169low/-tolDC promotes skin graft survival in mice via IL-10 +Breg. Transpl Immunol 2025; 91:102244. [PMID: 40414462 DOI: 10.1016/j.trim.2025.102244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2025] [Revised: 04/29/2025] [Accepted: 05/18/2025] [Indexed: 05/27/2025]
Abstract
Skin allografts are prone to rejection because of their high immunogenicity. By achieving immune tolerance, the long-term survival of skin allografts can be extended without the need for immunosuppressants or with only short-term low-dose dependency. Tolerogenic dendritic cells (tolDCs) show a strong potential for graft tolerance. We explored the mechanism whereby rapamycin-modified CD169low/-tolDCs regulate interleukin-10 (IL-10) B regulatory (IL-10+ Breg) cell production and mediate the long-term survival of skin allografts in mice. CD169low/-tolDCs were obtained through flow cytometry sorting after treating the mesenchymal stem cell (MSC)-derived dendritic cells with a low dose of GM-CSF. A treatment regimen combining preoperative stimulation and postoperative adoptive infusion of CD169low/-tolDCs was used to treat an acute rejection (AR) mouse skin transplantation model-the adoptive infusion group. An equivalent dose of saline was administered to the control group. Survival and graft rejection rates were assessed. Mixed lymphocyte culture, flow cytometry, immunohistochemistry (IHC), and western blotting (WB) were used to elucidate the expression of different IL-10+ Breg subsets in mice treated with adoptive infusion therapy and the molecular mechanisms whereby CD169low/-tolDCs induce IL-10+ Breg production to mediate tolerance. Adoptive infusion of CD169low/-tol DCs markedly prolonged the rejection time after skin transplantation in mice and promoted graft survival. A significant increase was observed in local blood flow signals in the transplanted skin, along with mild local inflammation. Flow cytometric analysis revealed a positive correlation between high expression of IL-10+ Breg and the changes in Foxp3+ Tregs in vivo, primarily enriched in the CD19 + CD24 + CD27+ mBreg and CD19 + CD23 + CD27-CD24+ Breg subsets, with higher levels of IgM expression. Significant differences were observed compared with control mice. CD79b/NF-κB pathway was found to be involved in Breg production. CD24, CD23, CD79, BTK, NF-κB p50/p65, CD40, and IKKα levels in the adoptive infusion group were significantly increased compared with those in the control group. Adoptive infusion of CD169low/-MSCs/tolDCs may activate the NF-κB pathway through CD79/BTK-dependent and CD40/IKKα-independent pathways, inducing high expression of IL-10 + Breg and promoting graft survival of mouse skin transplantation.
Collapse
Affiliation(s)
- Xi Lu
- Department of Plastic and Reconstructive Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing 100853, China; Weizikeng outpatient Department, Central Medical Branch of PLA General Hospital, Beijing 100101, China
| | - Yu-di Han
- Department of Plastic and Reconstructive Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing 100853, China
| | - Xiao-Ran Zu
- Department of Plastic and Reconstructive Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing 100853, China
| | - Jin-Can Huang
- Department of Hepatobiliary and Pancreaticosplenic Surgery, Beijing ChaoYang Hospital, Capital Medical University, Beijing 100020, China
| | - Li Li
- Department of Plastic and Reconstructive Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing 100853, China
| | - Meng Wang
- Department of Plastic and Reconstructive Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing 100853, China
| | - Yu-Ting Wang
- Department of Plastic and Reconstructive Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing 100853, China
| | - Ling-Li Guo
- Department of Plastic and Reconstructive Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing 100853, China.
| | - Lin Zhou
- Department of Hepatobiliary and Pancreaticosplenic Surgery, Beijing ChaoYang Hospital, Capital Medical University, Beijing 100020, China; Beijing Organ Transplant Center, Beijing ChaoYang Hospital, Beijing 100020, China.
| | - Yan Han
- Department of Plastic and Reconstructive Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing 100853, China.
| |
Collapse
|
7
|
Herrington RTB, Ellenberger DT, Rosenfeld CS. Maternal probiotic supplementation and effects on the fetal placenta. Biol Reprod 2025; 112:807-823. [PMID: 40057969 DOI: 10.1093/biolre/ioaf041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 02/14/2025] [Accepted: 03/03/2025] [Indexed: 05/16/2025] Open
Abstract
Increasing number of pregnant women are consumi ng probiotics to promote their own health and that of their unborn fetuses. Such supplements are presumed to be safe for pregnant mothers and their unborn offspring. For pregnant mothers, such bioactive compounds might lower the risk of constipation, diarrhea, other gastrointestinal conditions, and pre-term birth, and prevent adverse pregnancy outcomes, including gestational diabetes mellitus and depression/anxiety. More research is needed to examine potential safety of probiotic consumption during pregnancy and long-term health consequences to offspring. The conceptus can also be indirectly affected by maternal probiotic supplementation through microorganism production of bioactive compounds. The placenta is in direct communication with the underlying uterine tissue. Thus, compounds in the maternal blood can easily transfer across the placenta and impact this hormonally sensitive organ. Select studies suggest that disruptions to the maternal microbiome dramatically affect the placenta. In the current review, we will therefore consider the evidence to date of how maternal probiotic supplementation affects the placenta. Three potential mechanisms we will explore include the possibility that maternal probiotic supplementation might impact the putative placenta microbiome. The second potential mechanism we will consider is that maternal probiotic consumption alters bacterial-derived metabolites, including short-chained fatty acids, polyamines, Vitamin B9, and Vitamin B12. The third potential mechanism to be discussed is that such supplements affect maternal and placental immune responses. Before probiotics are promoted for healthy pregnant women and those with gestational disorders, more studies, including those examining the effects on the placenta, are essential.
Collapse
Affiliation(s)
- Rosalind T B Herrington
- Biomedical Sciences, University of Missouri, Columbia, Missouri 65211, USA
- Biological Sciences, University of Missouri, Columbia, Missouri 65211, USA
| | - David T Ellenberger
- Biomedical Sciences, University of Missouri, Columbia, Missouri 65211, USA
- Microbiology, University of Missouri Columbia, Missouri 65211, USA
| | - Cheryl S Rosenfeld
- Biomedical Sciences, University of Missouri, Columbia, Missouri 65211, USA
- MU Institute for Data Science and Informatics, University of Missouri, Columbia, Missouri 65211, USA
- Department of Genetics Area Program, University of Missouri, Columbia, Missouri 65211, USA
- Department of Thompson Center for Autism and Neurobehavioral Disorders, University of Missouri, Columbia, Missouri 65211, USA
| |
Collapse
|
8
|
Cembellin-Prieto A, Luo Z, Kulaga H, Baumgarth N. B cells modulate lung antiviral inflammatory responses via the neurotransmitter acetylcholine. Nat Immunol 2025; 26:775-789. [PMID: 40263611 PMCID: PMC12043518 DOI: 10.1038/s41590-025-02124-8] [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: 06/18/2024] [Accepted: 03/11/2025] [Indexed: 04/24/2025]
Abstract
The rapid onset of innate immune defenses is critical for early control of viral replication in an infected host and yet it can also lead to irreversible tissue damage, especially in the respiratory tract. Sensitive regulators must exist that modulate inflammation, while controlling the infection. In the present study, we identified acetylcholine (ACh)-producing B cells as such early regulators. B cells are the most prevalent ACh-producing leukocyte population in the respiratory tract demonstrated with choline acetyltransferase (ChAT)-green fluorescent protein (GFP) reporter mice, both before and after infection with influenza A virus. Mice lacking ChAT in B cells, disabling their ability to generate ACh (ChatBKO), but not those lacking ChAT in T cells, significantly, selectively and directly suppressed α7-nicotinic-ACh receptor-expressing interstitial, but not alveolar, macrophage activation and their ability to secrete tumor necrosis factor (TNF), while better controlling virus replication at 1 d postinfection. Conversely, TNF blockade via monoclonal antibody treatment increased viral loads at that time. By day 10 of infection, ChatBKO mice showed increased local and systemic inflammation and reduced signs of lung epithelial repair despite similar viral loads and viral clearance. Thus, B cells are key participants of an immediate early regulatory cascade that controls lung tissue damage after viral infection, shifting the balance toward reduced inflammation at the cost of enhanced early viral replication.
Collapse
Affiliation(s)
- Antonio Cembellin-Prieto
- Graduate Group in Immunology, University of California Davis, Davis, CA, USA
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Lyme and Tickborne Diseases Research and Education Institute, Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Zheng Luo
- Department of Pathology, Microbiology and Immunology, School of Veterinary Medicine, University of California Davis, Davis, CA, USA
| | - Heather Kulaga
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Lyme and Tickborne Diseases Research and Education Institute, Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Nicole Baumgarth
- Graduate Group in Immunology, University of California Davis, Davis, CA, USA.
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Lyme and Tickborne Diseases Research and Education Institute, Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA.
- Department of Pathology, Microbiology and Immunology, School of Veterinary Medicine, University of California Davis, Davis, CA, USA.
- Department of Molecular and Comparative Pathobiology, School of Medicine, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
9
|
Gagliardi F, De Domenico P, Snider S, Roncelli F, Comai S, Mortini P. Immunomodulatory mechanisms driving tumor escape in glioblastoma: The central role of IDO and tryptophan metabolism in local and systemic immunotolerance. Crit Rev Oncol Hematol 2025; 209:104657. [PMID: 39986404 DOI: 10.1016/j.critrevonc.2025.104657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Revised: 02/04/2025] [Accepted: 02/07/2025] [Indexed: 02/24/2025] Open
Abstract
BACKGROUND Glioblastoma (GBM) is the most aggressive primary brain tumor exhibiting extensive immune evasion mechanisms that hinder effective therapeutic interventions. This narrative review explores the immunomodulatory pathways contributing to tumor escape in GBM, specifically focusing on the role of Tryptophan (TRP) metabolism and its downstream mediators Tryptophan metabolism through the kynurenine pathway (KP) is initiated by indoleamine 2,3-dioxygenase (IDO) and tryptophan-2,3-dioxygenase (TDO2) enzymes and serves as a crucial mechanism for promoting an immunosuppressive microenvironments and systemic immunotolerance. Emerging evidence also indicates a non-enzymatic role for IDO1 signaling in these processes. The downstream effectors interact with immune cells, inducing local immunosuppression within the tumor microenvironment and altering peripheral immune responses. METHODS We systematically reviewed databases (MEDLINE via PubMed, Science Direct, and Embase) through October 2024 to highlight the interplay between local immune escape mechanisms and circulating immunotolerance, emphasizing the role of TRP metabolic enzymes in supporting GBM progression. RESULTS The literature review identified 99 records. TRP-related mechanisms play a central role in fostering immunotolerance in GBM. These phenomena involve intricate interactions between the infiltrating and circulating myeloid and lymphoid compartments, ultimately shaping a tolerant, pro-tumoral environment and the peripheral immunophenotype. CONCLUSIONS The biological activity of IDO1 and TRP metabolites positions these compounds as potential markers of disease activity and promising molecular targets for future therapeutic approaches.
Collapse
Affiliation(s)
- Filippo Gagliardi
- IRCCS San Raffaele Scientific Institute, Department of Neurosurgery and Gamma Knife Radiosurgery, Milan 20132, Italy
| | - Pierfrancesco De Domenico
- IRCCS San Raffaele Scientific Institute, Department of Neurosurgery and Gamma Knife Radiosurgery, Milan 20132, Italy.
| | - Silvia Snider
- IRCCS San Raffaele Scientific Institute, Department of Neurosurgery and Gamma Knife Radiosurgery, Milan 20132, Italy
| | - Francesca Roncelli
- IRCCS San Raffaele Scientific Institute, Department of Neurosurgery and Gamma Knife Radiosurgery, Milan 20132, Italy
| | - Stefano Comai
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Padua, Italy; Department of Biomedical Sciences, University of Padua, Padua, Italy; Department of Psychiatry, McGill University, Montreal, QC, Canada; IRCSS San Raffaele Scientific Institute, Division of Neuroscience, Milan, Italy
| | - Pietro Mortini
- IRCCS San Raffaele Scientific Institute, Department of Neurosurgery and Gamma Knife Radiosurgery, Milan 20132, Italy
| |
Collapse
|
10
|
Lee SK, Kwon JH, Jang JW, Bae SH, Yoon SK, Jung ES, Choi JY. The Critical Role of Regulatory T Cells in Immune Tolerance and Rejection Following Liver Transplantation: Interactions With the Gut Microbiome. Transplantation 2025; 109:784-793. [PMID: 39375899 DOI: 10.1097/tp.0000000000005220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/09/2024]
Abstract
Liver transplantation (LT) is the ultimate treatment for patients with end-stage liver disease or early hepatocellular carcinoma. In the context of LT, because of the unique immunological characteristics of human liver allograft, 5%-20% of selected LT recipients can achieve operational tolerance. Nonetheless, there remains a risk of rejection in LT patients. Maintaining immune homeostasis is thus crucial for improving clinical outcomes in these patients. In mechanism, several immune cells, including dendritic cells, Kupffer cells, myeloid-derived suppressor cells, hepatic stellate cells, regulatory B cells, and CD4 + regulatory T cells (Treg), contribute to achieving tolerance following LT. In terms of Treg, it plays a role in successfully minimizing immunosuppression or achieving tolerance post-LT while also reducing the risk of rejection. Furthermore, the gut microbiome modulates systemic immune functions along the gut-liver axis. Recent studies have explored changes in the microbiome and its metabolites under various conditions, including post-LT, acute rejection, and tolerance. Certain functional microbiomes and metabolites exhibit immunomodulatory functions, such as the augmentation of Treg, influencing immune homeostasis. Therefore, understanding the mechanisms of tolerance in LT, the role of Treg in tolerance and rejection, as well as their interactions with gut microbiome, is vital for the management of LT patients.
Collapse
Affiliation(s)
- Soon Kyu Lee
- Division of Hepatology, Department of Internal Medicine, Incheon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- The Catholic University Liver Research Center, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jung Hyun Kwon
- Division of Hepatology, Department of Internal Medicine, Incheon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- The Catholic University Liver Research Center, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jeong Won Jang
- The Catholic University Liver Research Center, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Division of Hepatology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Si Hyun Bae
- The Catholic University Liver Research Center, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Division of Hepatology, Department of Internal Medicine, Eunpyeong St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Seung Kew Yoon
- The Catholic University Liver Research Center, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Division of Hepatology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Eun Sun Jung
- Department of Pathology, Eunpyeong St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jong Young Choi
- The Catholic University Liver Research Center, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Division of Hepatology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| |
Collapse
|
11
|
De Domenico P, Gagliardi F, Roncelli F, Snider S, Mortini P. Tumor-infiltrating and circulating B cells mediate local and systemic immunomodulatory mechanisms in Glioblastoma. J Neurooncol 2025; 172:527-548. [PMID: 40080248 DOI: 10.1007/s11060-025-04989-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Accepted: 02/24/2025] [Indexed: 03/15/2025]
Abstract
BACKGROUND Glioblastoma (GBM) demonstrates extensive immunomodulatory mechanisms that challenge effective therapeutic interventions. These phenomena extend well beyond the tumor microenvironment (TME) and are reflected in the circulating immunophenotype. B lymphocytes (B cells) have received limited attention in GBM studies despite their emerging importance in mediating both local and systemic immune responses. Recent findings highlight the complex regulatory interactions between B cells and other immune cell populations, including tumor-infiltrating macrophages (TAMs), myeloid-derived suppressor cells (MDSCs), and other infiltrating lymphocytes (TILs). B cells are believed to hinder the efficacy of modern immunotherapy strategies focusing on T cells. METHODS This is a focused review of available evidence regarding B cells in GBM through January 2025. RESULTS Peripheral blood reflects a systemically dampened immune response, with sustained lymphopenia, increased plasma cells, and dysfunctional memory B cells. The tumor immune landscape is enriched in cells of B-lineage. Subsets of poorly characterized B regulatory cells (Bregs) populate the TME, developing their phenotype due to their proximity to MDSCs, TAMs, and tumoral cells. The Bregs inhibit CD8+ T activity and may have potential prognostic significance. CONCLUSION Understanding the role of B cells, how they are recruited, and their differentiation shifted towards an immunomodulatory role could inform better therapeutic strategies and unleash their full antitumoral potential in GBM.
Collapse
Affiliation(s)
- Pierfrancesco De Domenico
- Department of Neurosurgery and Gamma Knife Radiosurgery, IRCCS San Raffaele Scientific Institute and Vita-Salute San Raffaele University, Via Olgettina 60, 20132, Milan, Italy.
| | - Filippo Gagliardi
- Department of Neurosurgery and Gamma Knife Radiosurgery, IRCCS San Raffaele Scientific Institute and Vita-Salute San Raffaele University, Via Olgettina 60, 20132, Milan, Italy
| | - Francesca Roncelli
- Department of Neurosurgery and Gamma Knife Radiosurgery, IRCCS San Raffaele Scientific Institute and Vita-Salute San Raffaele University, Via Olgettina 60, 20132, Milan, Italy
| | - Silvia Snider
- Department of Neurosurgery and Gamma Knife Radiosurgery, IRCCS San Raffaele Scientific Institute and Vita-Salute San Raffaele University, Via Olgettina 60, 20132, Milan, Italy
| | - Pietro Mortini
- Department of Neurosurgery and Gamma Knife Radiosurgery, IRCCS San Raffaele Scientific Institute and Vita-Salute San Raffaele University, Via Olgettina 60, 20132, Milan, Italy
| |
Collapse
|
12
|
Li C, Liao J, Chen B, Wang Q. Heterogeneity of the tumor immune cell microenvironment revealed by single-cell sequencing in head and neck cancer. Crit Rev Oncol Hematol 2025; 209:104677. [PMID: 40023465 DOI: 10.1016/j.critrevonc.2025.104677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 02/16/2025] [Accepted: 02/26/2025] [Indexed: 03/04/2025] Open
Abstract
Head and neck cancer (HNC) is the sixth most common disease in the world. The recurrence rate of patients is relatively high, and the heterogeneity of tumor immune microenvironment (TIME) cells may be an important reason for this. Single-cell sequencing (SCS) is currently the most promising and mature application in cancer research. It can identify unique genes expressed in cells and study tumor heterogeneity. According to current research, the heterogeneity of immune cells has become an important factor affecting the occurrence and development of HNC. SCSs can provide effective therapeutic targets and prognostic factors for HNC patients through analyses of gene expression levels and cell heterogeneity. Therefore, this study analyzes the basic theory of HNC and the development of SCS technology, elaborating on the application of SCS technology in HNC and its potential value in identifying HNC therapeutic targets and biomarkers.
Collapse
Affiliation(s)
- Chunhong Li
- Department of Oncology, Suining Central Hospital, Suining, Sichuan 629000, China
| | - Jia Liao
- Department of Oncology, Suining Central Hospital, Suining, Sichuan 629000, China
| | - Bo Chen
- Department of Oncology, Suining Central Hospital, Suining, Sichuan 629000, China
| | - Qiang Wang
- Gastrointestinal Surgical Unit, Suining Central Hospital, Suining, Sichuan 629000, China.
| |
Collapse
|
13
|
Zhang Q, Liao J, Liu Z, Song S, Tian L, Wang Y. The immune tolerance role of Bregs in inhibiting human inflammatory diseases, with a focus on diabetes mellitus. Front Immunol 2025; 16:1565158. [PMID: 40370441 PMCID: PMC12074967 DOI: 10.3389/fimmu.2025.1565158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Accepted: 04/10/2025] [Indexed: 05/16/2025] Open
Abstract
Regulatory B cells (Bregs) are pivotal modulators of immune tolerance, suppressing inflammation through cytokine secretion and cellular interactions. Their role is particularly significant in inflammatory diseases such as type 1 and type 2 diabetes mellitus (T1DM and T2DM), where immune dysregulation contributes to disease progression. In T1DM, Bregs mitigate β-cell autoimmunity via IL-10 production and FOXP3-mediated pathways, but genetic mutations and dysfunctions in these mechanisms exacerbate autoimmunity. In T2DM, chronic inflammation and metabolic stress impair Breg numbers and function, further fueling insulin resistance. While Bregs play a central role in T1DM by directly preventing β-cell destruction, their role in T2DM is more supportive, modulating inflammation in metabolically stressed tissues. Emerging therapeutic strategies aim to enhance Breg function through IL-10 induction, ex vivo expansion, or targeting Breg-specific pathways using gene-editing and small molecules. Future research should explore Breg heterogeneity, novel markers, and personalized therapies to unlock their full potential. Understanding and leveraging the immune tolerance role of Bregs may offer transformative strategies to inhibit inflammatory diseases like diabetes mellitus.
Collapse
Affiliation(s)
- Qi Zhang
- Department of Laboratory Medicine, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, Sichuan, China
| | - Jinfeng Liao
- Department of Dermatology, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Zheng Liu
- Pathology Department, University of Texas, MD Anderson Cancer Center, Texas, Houston, TX, United States
| | - Siyuan Song
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, United States
| | - Limin Tian
- Center for Geriatrics and Endocrinology, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Yi Wang
- Center for Geriatrics and Endocrinology, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
- Center for Critical Care Medicine, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
- Clinical Immunology Translational Medicine Key Laboratory of Sichuan Province, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| |
Collapse
|
14
|
He L, Li X, Jiang S, Ou Y, Wang S, Shi N, Yang Z, Yuan JL, Silverman G, Niu H. The influence of the gut microbiota on B cells in autoimmune diseases. Mol Med 2025; 31:149. [PMID: 40264032 PMCID: PMC12016346 DOI: 10.1186/s10020-025-01195-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 04/01/2025] [Indexed: 04/24/2025] Open
Abstract
Mounting evidence shows that gut microbiota communities and the human immune system coexist and influence each other, and there are a number of reports of a correlation between specific changes in gut microbiota and the occurrence of autoimmune diseases. B lymphocytes play a central role in the regulation of both gut microbiota communities and in autoimmune diseases. Here, we summarize evidence of the influence of gut microbiota-B cell pathways on autoimmune diseases and how B cells regulate microorganisms, which provides mechanistic insights with relevance for identification of potential therapeutic targets and related fields.
Collapse
Affiliation(s)
- Lun He
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education; Guangzhou Key Laboratory for Germ-free Animals and Microbiota Application, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Xin Li
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education; Guangzhou Key Laboratory for Germ-free Animals and Microbiota Application, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Shan Jiang
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education; Guangzhou Key Laboratory for Germ-free Animals and Microbiota Application, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Yanhua Ou
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education; Guangzhou Key Laboratory for Germ-free Animals and Microbiota Application, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Shanshan Wang
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education; Guangzhou Key Laboratory for Germ-free Animals and Microbiota Application, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Na Shi
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education; Guangzhou Key Laboratory for Germ-free Animals and Microbiota Application, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Zhongshan Yang
- Yunnan Provincial Key Laboratory of Molecular Biology for Sinomedicine, Yunnan University of Chinese Medicine, Kunming, Yunnan, 650500, China
| | - Jia-Li Yuan
- Yunnan Provincial Key Laboratory of Molecular Biology for Sinomedicine, Yunnan University of Chinese Medicine, Kunming, Yunnan, 650500, China.
| | - Gregg Silverman
- Division of Rheumatology, New York University School of Medicine, New York, NY, 10016, USA.
| | - Haitao Niu
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education; Guangzhou Key Laboratory for Germ-free Animals and Microbiota Application, School of Medicine, Jinan University, Guangzhou, 510632, China.
- Yunnan Provincial Key Laboratory of Molecular Biology for Sinomedicine, Yunnan University of Chinese Medicine, Kunming, Yunnan, 650500, China.
| |
Collapse
|
15
|
Viggiano D, Iulianiello P, Mancini A, Iacuzzo C, Apicella L, Di Pietro RA, Hamzeh S, Cacciola G, Lippiello E, Gigliotti A, Secondulfo C, Bilancio G, Gigliotti G. Immunological Avalanches in Renal Immune Diseases. Biomedicines 2025; 13:1003. [PMID: 40299571 PMCID: PMC12024534 DOI: 10.3390/biomedicines13041003] [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: 02/16/2025] [Revised: 04/04/2025] [Accepted: 04/09/2025] [Indexed: 05/01/2025] Open
Abstract
The complex nature of immune system behavior in both autoimmune diseases and transplant rejection can be understood through the lens of avalanche dynamics in critical-point systems. This paper introduces the concept of the "immunological avalanche" as a framework for understanding unpredictable patterns of immune activity in both contexts. Just as avalanches represent sudden releases of accumulated potential energy, immune responses exhibit periods of apparent stability followed by explosive flares triggered by seemingly minor stimuli. The model presented here draws parallels between immune system behavior and other complex systems such as earthquakes, forest fires, and neuronal activity, where localized events can propagate into large-scale disruptions. In autoimmune conditions like systemic lupus erythematosus (SLE), which affects multiple organ systems including the kidneys in approximately 50% of patients, these dynamics manifest as alternating periods of remission and flares. Similarly, in transplant recipients, the immune system exhibits metastable behavior under constant allograft stimulation. This critical-point dynamics framework is characterized by threshold-dependent activation, positive feedback loops, and dynamic non-linearity. In autoimmune diseases, triggers such as UV light exposure, infections, or stress can initiate cascading immune responses. In transplant patients, longitudinal analysis reveals how monitoring oscillatory patterns in blood parameters and biological age markers can predict rejection risk. In a preliminary study on kidney transplant, all measured variables showed temporal instability. Proteinuria exhibited precise log-log linearity in power law analysis, confirming near-critical-point system behavior. Two distinct dynamic patterns emerged: large oscillations in eGFR, proteinuria, or biological age predicted declining function, while small oscillations indicated stability. During avalanche events, biological age increased dramatically, with partial reversal leaving persistent elevation after acute episodes. Understanding these dynamics has important implications for therapeutic approaches in both contexts. Key findings suggest that monitoring parameter oscillations, rather than absolute values, better indicates system instability and potential avalanche events. Additionally, biological age calculations provide valuable prognostic information, while proteinuria measurements offer efficient sampling for system dynamics assessment. This conceptual model provides a unifying framework for understanding the pathogenesis of both autoimmune and transplant-related immune responses, potentially leading to new perspectives in disease management and rejection prediction.
Collapse
Affiliation(s)
- Davide Viggiano
- Department Translational Medical Sciences, University of Campania, 80131 Naples, Italy; (P.I.); (G.C.)
| | - Pietro Iulianiello
- Department Translational Medical Sciences, University of Campania, 80131 Naples, Italy; (P.I.); (G.C.)
| | - Antonio Mancini
- Department of Nephrology and Dialysis, Eboli Hospital, 84025 Eboli, Italy; (A.M.); (A.G.); (G.G.)
| | - Candida Iacuzzo
- Unit of Nephrology, Dialysis and Transplantation, Salerno University Hospital, 84131 Salerno, Italy; (C.I.); (L.A.); (R.A.D.P.)
| | - Luca Apicella
- Unit of Nephrology, Dialysis and Transplantation, Salerno University Hospital, 84131 Salerno, Italy; (C.I.); (L.A.); (R.A.D.P.)
| | - Renata Angela Di Pietro
- Unit of Nephrology, Dialysis and Transplantation, Salerno University Hospital, 84131 Salerno, Italy; (C.I.); (L.A.); (R.A.D.P.)
| | - Sarah Hamzeh
- Department of Public Health, Federico II University of Naples, 80131 Naples, Italy;
| | - Giovanna Cacciola
- Department Translational Medical Sciences, University of Campania, 80131 Naples, Italy; (P.I.); (G.C.)
| | - Eugenio Lippiello
- Department Mathematics and Physics, University of Campania, 81100 Caserta, Italy;
| | - Andrea Gigliotti
- Department of Nephrology and Dialysis, Eboli Hospital, 84025 Eboli, Italy; (A.M.); (A.G.); (G.G.)
| | - Carmine Secondulfo
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, 84081 Baronissi, Italy; (C.S.)
| | - Giancarlo Bilancio
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, 84081 Baronissi, Italy; (C.S.)
| | - Giuseppe Gigliotti
- Department of Nephrology and Dialysis, Eboli Hospital, 84025 Eboli, Italy; (A.M.); (A.G.); (G.G.)
| |
Collapse
|
16
|
Raja KD, Singh A, Akhtar S, Singh P, Seth A, Kaushal S, Sharma A. Phenotypic Diversity of Immunosuppressive B Cells Associated in Urothelial Carcinoma of the Bladder. Clin Genitourin Cancer 2025:102351. [PMID: 40413097 DOI: 10.1016/j.clgc.2025.102351] [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: 09/20/2024] [Revised: 12/19/2024] [Accepted: 04/07/2025] [Indexed: 05/27/2025]
Abstract
BACKGROUND Urothelial carcinoma of the bladder presents a complex tumor microenvironment, with tumor-infiltrating B cells (TIL-Bs) playing a significant role in disease progression. Although their presence is acknowledged, the phenotypic diversity of regulatory TIL-Bs in bladder cancer remain underexplored. MATERIALS AND METHODS In this study, we evaluated core B cell subsets and their immunosuppressive phenotypes in both peripheral blood (n=40) and bladder tumor tissues (n=40) to evaluate their relationship with disease severity. RESULTS Our findings revealed that high-grade bladder tumors are enriched with B cells and their subsets, particularly transitional B cells and plasmacytes (plasmablasts and plasma cells). However, total memory B cells were reduced in the tumor microenvironment compared to non-tumor tissues. It was further revealed that the high-grade tumors demonstrated significant infiltration of regulatory B cells (Breg), with elevated levels of IL10+ and TGFβ+ Breg cells as well as IL-10+TGF-β+ dual-cytokine-secreting Breg cells, suggesting their role in fostering an immunosuppressive microenvironment. Memory B cells demonstrated the highest frequency of Breg phenotypes among the B cell subsets. Additionally, Tertiary Lymphoid Structure formation and frequency were associated with disease severity, the differentiated B cells and IL10+ Breg cell counts, emphasizing the importance of these structures in bladder cancer progression and the potential involvement in Breg cells formation. CONCLUSION This study demonstrates the enrichment of the bladder cancer tumor microenvironment with diverse B cell subsets, including functional Breg cells, which correlates with disease severity.
Collapse
Affiliation(s)
| | - Aishwarya Singh
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Shamima Akhtar
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Prabhjot Singh
- Department of Urology, All India Institute of Medical Sciences, New Delhi, India
| | - Amlesh Seth
- Department of Urology, All India Institute of Medical Sciences, New Delhi, India
| | - Seema Kaushal
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India
| | - Alpana Sharma
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India.
| |
Collapse
|
17
|
Yao Y, Liu YY, Li JF, Chen YS, Shi L, Shen Y, Yang LL, Yang Q. Indoleamine 2,3-dioxygenase 1 alters the proportions of B cell subpopulations in the microenvironment of acute myeloid leukemia. MOLECULAR BIOMEDICINE 2025; 6:23. [PMID: 40234305 PMCID: PMC12000501 DOI: 10.1186/s43556-025-00262-x] [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: 07/29/2024] [Revised: 03/06/2025] [Accepted: 03/17/2025] [Indexed: 04/17/2025] Open
Abstract
Acute myeloid leukemia (AML), the most common leukemia in adults, exhibits immune escape characteristics like solid tumors. The expression of indoleamine 2,3-dioxygenase 1 (IDO1), a well-recognized immune checkpoint, has been detected in AML blast cells and is associated with poor clinical outcome. Although an imbalance of B cell subpopulations exists in AML patients' bone marrow microenvironment, the role of B cells and their interaction with IDO1 in AML have yet to be elucidated. Herein, with bioinformatic analysis, we found the close correlations between IDO1 expression and survival and B cell subpopulation proportions in AML patients. Further, our investigation into IDO1 expression and activity, B cell subpopulation proportions and immunosuppressive interleukin-10 (IL-10) level in AML cells and clinical samples revealed significant findings. Using a co-culture system of healthy human PBMCs and AML cell lines, we demonstrated that high IDO1 expression in AML cells could alter the proportions of total B, regulatory B and memory B cells, and increased the level of IL-10. Finally, with the IDO1 inhibitor RY103 designed by our laboratory, we found that IDO1 inhibition had good anti-leukemic effect and restored the abnormal proportions of B cell subpopulations in AML mice. Our study is the first to reveal the modulation of IDO1 on B cell subpopulations in AML, making a significant breakthrough in understanding the immune escape mechanisms of AML. Application of IDO1 inhibitor, such as RY103, targeting the imbalance of B cell subpopulations can lead to innovative treatments for AML.
Collapse
MESH Headings
- Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism
- Indoleamine-Pyrrole 2,3,-Dioxygenase/antagonists & inhibitors
- Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics
- Humans
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myeloid, Acute/immunology
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/enzymology
- Leukemia, Myeloid, Acute/genetics
- Tumor Microenvironment/immunology
- Animals
- Interleukin-10/metabolism
- Mice
- Cell Line, Tumor
- Male
- Female
- B-Lymphocyte Subsets/immunology
- B-Lymphocyte Subsets/metabolism
- Middle Aged
- Adult
Collapse
Affiliation(s)
- Yu Yao
- State Key Laboratory of Genetics and Development of Complex Phenotypes, School of Life Sciences, Fudan University, Songhu Road 2005, Shanghai, 200438, China
| | - Yu-Ying Liu
- State Key Laboratory of Genetics and Development of Complex Phenotypes, School of Life Sciences, Fudan University, Songhu Road 2005, Shanghai, 200438, China
| | - Jian-Feng Li
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yun-Shuo Chen
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Lei Shi
- State Key Laboratory of Genetics and Development of Complex Phenotypes, School of Life Sciences, Fudan University, Songhu Road 2005, Shanghai, 200438, China
| | - Yang Shen
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Li-Li Yang
- State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China.
| | - Qing Yang
- State Key Laboratory of Genetics and Development of Complex Phenotypes, School of Life Sciences, Fudan University, Songhu Road 2005, Shanghai, 200438, China.
| |
Collapse
|
18
|
Liao J, Yang Y, Li J, Liu Z, Song S, Zeng Y, Wang Y. Regulatory B cells, the key regulator to induce immune tolerance in organ transplantation. Front Immunol 2025; 16:1561171. [PMID: 40264774 PMCID: PMC12011811 DOI: 10.3389/fimmu.2025.1561171] [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: 01/15/2025] [Accepted: 03/24/2025] [Indexed: 04/24/2025] Open
Abstract
In solid organ transplantation, especially renal transplantation, for the induction of immune tolerance, accumulating evidence has revealed that Regulatory B cells (Breg) play a crucial role in stimulating immune tolerance, alleviating immune responses, and improving graft survival. We describe the heterogeneous nature of Bregs, focusing on their defining surface markers and regulatory functions. Meanwhile, the major cytokine secretion function and the correlation between Breg and Treg or other immune checkpoints to balance the immune responses are addressed. Furthermore, we summarized the intrinsic and extrinsic pathways or costimulatory stimuli for the differentiation from naïve B cells. More importantly, we summarized the progression of the immune tolerance induction role of Breg in solid organ (kidney, liver, heart, lung, and islet) transplantation. This is an up-to-date review from the origin of Breg to the function of Breg in solid organ transplantation and how it induces immune tolerance in both murine models and human solid organ transplantation.
Collapse
Affiliation(s)
- Jinfeng Liao
- Department of Dermatology, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Yixin Yang
- Department of Clinical Medicine, The First Clinical Medical College of Norman Bethune University of Medical Sciences, Jilin, China
| | - Jisong Li
- Department of Gastrointestinal Surgery, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Zheng Liu
- Department of Pathology, MD Anderson Cancer Center, Houston, TX, United States
| | - Siyuan Song
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, United States
| | - Yu Zeng
- Department of Hyperbaric Oxygen, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Yi Wang
- Department of Critical Care Medicine, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
- Translational Clinical Immunology Key Laboratory of Sichuan Province, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, Chengdu, China
| |
Collapse
|
19
|
Xiao ZF, Chai WH, Shu XL, Yuan HR, Guo F. Immune cell traits and causal relationships with cholecystitis: a mendelian randomization analysis. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:3817-3827. [PMID: 39358644 DOI: 10.1007/s00210-024-03493-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 09/25/2024] [Indexed: 10/04/2024]
Abstract
Cholecystitis, characterized by inflammation of the gallbladder, is intricately linked to immune cells and the cytokines they produce. Despite this association, the specific contributions of immune cells to the onset and progression of cholecystitis remain to be fully understood. To delineate this relationship, we utilized the Mendelian randomization (MR) method to scrutinize the causal connections between 731 immune cell phenotypes and cholecystitis. By conducting MR analysis on 731 immune cell markers from public datasets, this study seeks to understand their potential impact on the risk of cholecystitis. It aims to elucidate the interactions between immune phenotypes and the disease, aiming to lay the groundwork for advancing precision medicine and developing effective treatment strategies for cholecystitis. Taking immune cell phenotypes as the exposure factor and cholecystitis as the outcome event, this study used single nucleotide polymorphisms (SNPs) closely associated with both immune cell phenotypes and cholecystitis as genetic instrumental variables. We conducted a two-sample MR analysis on genome-wide association studies (GWAS) data. Our research thoroughly examined 731 immune cell markers, to determine potential causal relationships with susceptibility to cholecystitis. Sensitivity analyses were performed to ensure the robustness of our findings, excluding the potential impacts of heterogeneity and pleiotropy. To avoid reverse causality, we conducted reverse MR analyses with cholecystitis as the exposure factor and immune cell phenotypes as the outcome event. Among the 731 immune phenotypes, our study identified 21 phenotypes with a causal relationship to cholecystitis (P < 0.05). Of these, eight immune phenotypes exhibited a protective effect against cholecystitis (odds ratio (OR) < 1), while the other 13 immune phenotypes were associated with an increased risk of developing cholecystitis (OR > 1). Additionally, employing the false discovery rate (FDR) method at a significance level of 0.2, no significant causal relationship was found between cholecystitis and immune phenotypes. Our research has uncovered a significant causal relationship between immune cell phenotypes and cholecystitis. This discovery not only enhances our understanding of the role of immune cells in the onset and progression of cholecystitis but also establishes a foundation for developing more precise biomarkers and targeted therapeutic strategies. It provides a scientific basis for more effective and personalized treatments in the future. These findings are expected to substantially improve the quality of life for patients with cholecystitis and mitigate the impact of the disease on patients and their families.
Collapse
Affiliation(s)
- Ze-Fa Xiao
- The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Wei-Hao Chai
- Department of Graduate School, Xinjiang Medical University, Urumqi, China
| | - Xiao-Long Shu
- The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Hong-Rui Yuan
- The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Fei Guo
- Department of Emergency Trauma Surgery, The First Affiliated Hospital of Xinjiang Medical University, Xinjiang Uygur Autonomous Region, Urumqi, 830054, China.
| |
Collapse
|
20
|
Fu XJ, Meng C, Guo L, Lin LE. Therapeutic efficacy of rituximab combined with cyclosporin A on B-cell dysregulation in chronic graft-versus-host disease. Clin Transl Oncol 2025; 27:1789-1797. [PMID: 39231914 DOI: 10.1007/s12094-024-03684-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 08/20/2024] [Indexed: 09/06/2024]
Abstract
OBJECTIVE Chronic graft-versus-host disease (cGVHD) is a significant complication following allogenic hematopoietic stem cell transplantation, often necessitating therapeutic interventions such as rituximab (RTX) and cyclosporin A (CsA). This study aims to elucidate the mechanisms by which RTX and CsA jointly address B-cell dysregulation in cGVHD, providing a theoretical foundation and scientific rationale for the treatment and prognostic evaluation of this condition. METHODS A total of 30 cGVHD mouse models were established by subjecting recipient mice to total body irradiation followed by injection of a mixed suspension of bone marrow cells and splenocytes from donor mice. From Day 2 to Day 29 post-model establishment, the mice received subcutaneous administration of RTX and CsA. Throughout the study, body weight, clinical cGVHD scores, and survival rates were monitored. Blood samples were collected via the orbital venous plexus. Serum levels of B-cell activating factor (BAFF) and pro-inflammatory factors were measured using enzyme-linked immunosorbent assay (ELISA), and the ratio of regulatory B cells (Bregs) in the blood sample was assessed via flow cytometry. RESULTS Mice with cGVHD exhibited a 14.5% decrease in body weight, elevated clinical scores, and more severe symptoms compared to the control group. Notably, all mice in both the cGVHD and control groups survived until the conclusion of the study. Induction of cGVHD resulted in B-cell dysregulation, evidenced by elevated serum BAFF levels and a decreased proportion of Bregs. However, treatment with RTX combined with CsA ameliorated B-cell dysregulation and significantly reduced serum levels of pro-inflammatory factors in cGVHD mice, with decreases of 39.78% in TNF-α and 37.89% in IL-6. CONCLUSION The combination of RTX and CsA effectively mitigates B-cell dysregulation in cGVHD, thereby reducing the severity and progression of the disease.
Collapse
Affiliation(s)
- Xiang-Jun Fu
- Department of Hematology, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, NO.19 Xiuhua Road, Xiuying District, Haikou, 570311, Hainan Province, China
| | - Can Meng
- Department of Hematology, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, NO.19 Xiuhua Road, Xiuying District, Haikou, 570311, Hainan Province, China
| | - Li Guo
- Department of Hematology, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, NO.19 Xiuhua Road, Xiuying District, Haikou, 570311, Hainan Province, China
| | - Li-E Lin
- Department of Hematology, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, NO.19 Xiuhua Road, Xiuying District, Haikou, 570311, Hainan Province, China.
| |
Collapse
|
21
|
Ma H, Wang Z, Yu M, Zhai Y, Yan J. Aberrations in peripheral B lymphocytes and B lymphocyte subsets levels in Parkinson disease: a systematic review. Front Immunol 2025; 16:1526095. [PMID: 40230858 PMCID: PMC11994702 DOI: 10.3389/fimmu.2025.1526095] [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: 11/11/2024] [Accepted: 03/10/2025] [Indexed: 04/16/2025] Open
Abstract
Objective The association of B lymphocytes and B lymphocyte subsets and Parkinson's disease (PD) is increasingly acknowledged. However, there is inconsistence in the alterations of B lymphocytes or B lymphocyte subsets in peripheral blood of PD patients. To comprehensively understand its changes in PD patients,it is necessary to conduct a systematic review on this subject. Methods PubMed, Cochrane Library, and MEDLINE databases were searched until 3rd February 2024. Results We included 20 studies (n=2658) to conduct this systematic review. We conducted a qualitative analysis to assess the alterations of B lymphocytes and B lymphocyte subsets in the peripheral blood of individuals with PD. And studies reviewed demonstrated a significant decrease in the number of B cells, as well as immune dysregulation in the B lymphocyte subsets of these patients' peripheral blood. Conclusion Studies reviewed demonstrated that PD is linked to abnormalities in B lymphocytes and/or B lymphocytes subsets in peripheral blood. This study provides a novel perspective into the pathogenesis of PD, and future investigations into the B lymphocytes and/or B lymphocyte subsets as biomarkers and therapeutic targets for PD is warranted.
Collapse
Affiliation(s)
- Hongxia Ma
- Department of Neurology, The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| | - Ziyuan Wang
- Department of Neurology, The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| | - Miao Yu
- Department of Neurology, The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| | - Yibo Zhai
- Department of Neurology, The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| | - Junqiang Yan
- Department of Neurology, The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
- Key laboratory of Neuromolecular Biology, The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| |
Collapse
|
22
|
Gossez M, Vigneron C, Vandermoeten A, Lepage M, Courcol L, Coudereau R, Paidassai H, Jallades L, Lopez J, Kandara K, Ortillon M, Mommert M, Fabri A, Peronnet E, Grosjean C, Buisson M, Lukaszewicz AC, Rimmelé T, Argaud L, Cour M, Py BF, Thaunat O, Defrance T, Monneret G, Venet F. PD-L1 + plasma cells suppress T lymphocyte responses in patients with sepsis and mouse sepsis models. Nat Commun 2025; 16:3030. [PMID: 40155394 PMCID: PMC11953283 DOI: 10.1038/s41467-025-57706-9] [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: 03/28/2023] [Accepted: 02/28/2025] [Indexed: 04/01/2025] Open
Abstract
Sepsis, a leading cause of death in intensive care units, is associated with immune alterations that increase the patients' risk of secondary infections and mortality, so better understandings of the pathophysiology of sepsis-induced immunosuppression is essential for the development of therapeutic strategies. In a murine model of sepsis that recapitulates immune alterations observed in patients, here we demonstrate that PD-L1+CD44+B220LowCD138+IgM+ regulatory plasma cells are induced in spleen and regulate ex vivo proliferation and IFNɣ secretion induced by stimulation of T splenocytes. This effect is mediated both by cell-cell contact through increased PD-L1 expression on plasma cells and by production of a soluble factor. These observations are recapitulated in three cohorts of critically ill patients with bacterial and viral sepsis in association with increased mortality. Our findings thus reveal the function of regulatory plasma cells in the pathophysiology of sepsis-induced immune alterations, and present a potential therapeutic target for improving immune cell function impaired by sepsis.
Collapse
Affiliation(s)
- Morgane Gossez
- Hospices Civils de Lyon, Immunology Laboratory, Lyon-Sud & Edouard Herriot University Hospitals, Lyon, France
- CIRI, Centre International de Recherche en Infectiologie, Univ Lyon, Inserm U1111, Université Claude Bernard-Lyon 1, CNRS, UMR5308, ENS de Lyon, Lyon, France
| | - Clara Vigneron
- CIRI, Centre International de Recherche en Infectiologie, Univ Lyon, Inserm U1111, Université Claude Bernard-Lyon 1, CNRS, UMR5308, ENS de Lyon, Lyon, France
| | - Alexandra Vandermoeten
- Service Commun des Animaleries de Rockefeller (SCAR) - Université Claude Bernard lyon1, Structure Fédérative de Recherche (SFR) Santé Lyon Est, Lyon, France
| | - Margot Lepage
- Hospices Civils de Lyon, Immunology Laboratory, Lyon-Sud & Edouard Herriot University Hospitals, Lyon, France
- CIRI, Centre International de Recherche en Infectiologie, Univ Lyon, Inserm U1111, Université Claude Bernard-Lyon 1, CNRS, UMR5308, ENS de Lyon, Lyon, France
| | - Louise Courcol
- CIRI, Centre International de Recherche en Infectiologie, Univ Lyon, Inserm U1111, Université Claude Bernard-Lyon 1, CNRS, UMR5308, ENS de Lyon, Lyon, France
| | - Remy Coudereau
- Hospices Civils de Lyon, Immunology Laboratory, Lyon-Sud & Edouard Herriot University Hospitals, Lyon, France
- EA 7426 "Pathophysiology of Injury-Induced Immunosuppression" (Université Claude Bernard Lyon 1, Hospices Civils de Lyon, bioMérieux), Lyon, France
| | - Helena Paidassai
- CIRI, Centre International de Recherche en Infectiologie, Univ Lyon, Inserm U1111, Université Claude Bernard-Lyon 1, CNRS, UMR5308, ENS de Lyon, Lyon, France
| | - Laurent Jallades
- CIRI, Centre International de Recherche en Infectiologie, Univ Lyon, Inserm U1111, Université Claude Bernard-Lyon 1, CNRS, UMR5308, ENS de Lyon, Lyon, France
- Hospices Civils de Lyon, Lyon Sud University Hospital, Hematology Laboratory, Pierre-Bénite, France
| | - Jonathan Lopez
- Hospices Civils de Lyon, Biochemistry and Molecular Biology department, Lyon Est Faculty of Medicine, Université Claude Bernard Lyon 1, Université de Lyon, Lyon Sud University Hospital, Pierre-Bénite, France
| | - Khalil Kandara
- Hospices Civils de Lyon, Immunology Laboratory, Lyon-Sud & Edouard Herriot University Hospitals, Lyon, France
| | - Marine Ortillon
- Hospices Civils de Lyon, Immunology Laboratory, Lyon-Sud & Edouard Herriot University Hospitals, Lyon, France
| | - Marine Mommert
- EA 7426 "Pathophysiology of Injury-Induced Immunosuppression" (Université Claude Bernard Lyon 1, Hospices Civils de Lyon, bioMérieux), Lyon, France
| | - Astrid Fabri
- Hospices Civils de Lyon, Immunology Laboratory, Lyon-Sud & Edouard Herriot University Hospitals, Lyon, France
| | - Estelle Peronnet
- EA 7426 "Pathophysiology of Injury-Induced Immunosuppression" (Université Claude Bernard Lyon 1, Hospices Civils de Lyon, bioMérieux), Lyon, France
| | - Clémence Grosjean
- EA 7426 "Pathophysiology of Injury-Induced Immunosuppression" (Université Claude Bernard Lyon 1, Hospices Civils de Lyon, bioMérieux), Lyon, France
| | - Marielle Buisson
- Centre d'Investigation Clinique de Lyon (CIC 1407 Inserm), Hospices Civils de Lyon, Lyon, France
| | - Anne-Claire Lukaszewicz
- EA 7426 "Pathophysiology of Injury-Induced Immunosuppression" (Université Claude Bernard Lyon 1, Hospices Civils de Lyon, bioMérieux), Lyon, France
- Hospices Civils de Lyon, Anesthesia and Critical Care Medicine Department, Edouard Herriot Hospital, Lyon, France
| | - Thomas Rimmelé
- EA 7426 "Pathophysiology of Injury-Induced Immunosuppression" (Université Claude Bernard Lyon 1, Hospices Civils de Lyon, bioMérieux), Lyon, France
- Hospices Civils de Lyon, Anesthesia and Critical Care Medicine Department, Edouard Herriot Hospital, Lyon, France
| | - Laurent Argaud
- Hospices Civils de Lyon, Medical Intensive Care Department, Edouard Herriot Hospital, Lyon, France
| | - Martin Cour
- Hospices Civils de Lyon, Medical Intensive Care Department, Edouard Herriot Hospital, Lyon, France
| | - Bénédicte F Py
- CIRI, Centre International de Recherche en Infectiologie, Univ Lyon, Inserm U1111, Université Claude Bernard-Lyon 1, CNRS, UMR5308, ENS de Lyon, Lyon, France
| | - Olivier Thaunat
- CIRI, Centre International de Recherche en Infectiologie, Univ Lyon, Inserm U1111, Université Claude Bernard-Lyon 1, CNRS, UMR5308, ENS de Lyon, Lyon, France
| | - Thierry Defrance
- CIRI, Centre International de Recherche en Infectiologie, Univ Lyon, Inserm U1111, Université Claude Bernard-Lyon 1, CNRS, UMR5308, ENS de Lyon, Lyon, France
| | - Guillaume Monneret
- Hospices Civils de Lyon, Immunology Laboratory, Lyon-Sud & Edouard Herriot University Hospitals, Lyon, France
- EA 7426 "Pathophysiology of Injury-Induced Immunosuppression" (Université Claude Bernard Lyon 1, Hospices Civils de Lyon, bioMérieux), Lyon, France
| | - Fabienne Venet
- Hospices Civils de Lyon, Immunology Laboratory, Lyon-Sud & Edouard Herriot University Hospitals, Lyon, France.
- CIRI, Centre International de Recherche en Infectiologie, Univ Lyon, Inserm U1111, Université Claude Bernard-Lyon 1, CNRS, UMR5308, ENS de Lyon, Lyon, France.
| |
Collapse
|
23
|
Yue M, Luan R, Ding D, Wang Y, Xue Q, Yang J. Identification and validation of biomarkers related to ferroptosis in idiopathic pulmonary fibrosis. Sci Rep 2025; 15:8622. [PMID: 40075162 PMCID: PMC11904244 DOI: 10.1038/s41598-025-93217-9] [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: 09/03/2024] [Accepted: 03/05/2025] [Indexed: 03/14/2025] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a kind of interstitial lung disease (ILD). It has a high incidence rate and mortality. Its pathogenesis remains unclear. So far, no effective methods have been found for the early diagnosis of IPF. Ferroptosis has been reported to be critical in the initiation and progression of IPF. Therefore, our aim was to identify the hub gene related to ferroptosis co-expressed in the peripheral blood and pulmonary tissue of patients with IPF. Sequencing data were obtained from the Gene Expression Omnibus database. A comprehensive analysis was conducted on the differentially expressed genes (DEGs) to extract ferroptosis-related differentially expressed genes (FRDEGs). The results showed that ferroptosis-related signal paths were highly enriched in IPF, and 10 FRDEGs were identified.The hub gene was predicted through protein-protein interactions (PPI) and Cytoscape. The diagnostic utility of the hub gene was proven by enzyme-linked immunosorbent assay (ELISA) in serum and by immunohistochemistry (IHC) in pulmonary tissues. The results of ELISA indicated that the levels of ATM in the serum of patients with IPF were significantly lower than the normal levels. In contrast, the results of IHC showed that the expression of ATM in the pulmonary tissues of IPF patients exhibited a notably elevated trend. The immune status was assessed by the CIBERSORT method and so was the relevance between ATM and immune cells. These findings unveiled significant differences in various immune cell types in peripheral blood and pulmonary tissue between the IPF group and the control group. Furthermore, ATM was associated with various immune cells. This study suggests that as a ferroptosis-related gene, ATM assumes a pivotal role in the diagnosis and treatment of IPF. This discovery presents a novel approach for the clinical diagnosis and therapy of IPF.
Collapse
Affiliation(s)
- Ming Yue
- Department of Respiratory Medicine, The Second Hospital of Jilin University, Changchun, China
| | - Rumei Luan
- Department of Respiratory Medicine, Shandong First Medical University Affiliated Provincial Hospital, Jinan, China
| | - Dongyan Ding
- Department of Respiratory Medicine, The 958 Hospital of Chinese PLA/Jiangbei Campus, The First Affiliated Hospital of Army Medical University, Chongqing, China
| | - Yuhong Wang
- Department of Respiratory Medicine, Jilin Central General Hospital, Jilin, China
| | - Qianfei Xue
- Hospital of Jilin University, Changchun, China.
| | - Junling Yang
- Department of Respiratory Medicine, The Second Hospital of Jilin University, Changchun, China.
| |
Collapse
|
24
|
Zhang Y, Zhang C, Yang B, Peng C, Zhou J, Ren S, Hu Z. The effect of TIM1 + Breg cells in liver ischemia-reperfusion injury. Cell Death Dis 2025; 16:171. [PMID: 40075055 PMCID: PMC11903774 DOI: 10.1038/s41419-025-07446-x] [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: 09/05/2024] [Revised: 01/24/2025] [Accepted: 02/11/2025] [Indexed: 03/14/2025]
Abstract
Liver transplantation is the only effective method for end-stage liver disease; however, liver ischemia reperfusion injury (IRI) seriously affects donor liver function after liver transplantation. IRI is a pathophysiological process in which organ damage is aggravated after the blood flow and oxygen supply of ischemic organ tissues are restored. It combines the two stages of hypoxic cell stress triggered by ischemia and inflammation-mediated reperfusion injury. Herein, we studied the protective effect and mechanism of the anti-T cell Ig and mucin domain (TIM1) monoclonal antibody, RMT1-10, on hepatic cell injury induced by IRI. First, a liver IRI model was established in vivo. HE, TEM, and Tunel were used to detect liver tissue injury, changes in the liver ultrastructure and liver cell apoptosis, respectively. ELISA were performed to determine the levels of ALT, AST, MDA, GSH, and related inflammatory factors. We found that RMT1-10 could significantly reduce liver injury. Flow cytometry results showed that the number of TIM1+ regulatory B cells (Bregs) in the IRI liver increased briefly, while pretreatment with RMT1-10 could increase the number of TIM1+ Bregs and interleukin-10 (IL-10) secretion in liver IRI model mice, thus playing a protective role in liver reperfusion. When Anti-CD20 was used to remove B cells, RMT1-10 had a reduced effect on liver IRI. Previous data showed that the number of T helper 1 cells (Th1:CD4+; CD8+) increased significantly after IRI. RMT1-10 inhibited Th1 cells; however, it significantly activated regulatory T cells. Sequencing analysis showed that RMT1-10 could significantly downregulate the expression of nuclear factor-kappa B (NF-κB) pathway-related genes induced by IRI. These results suggested that RMT1-10 could promote the maturation of B cells through an atypical NF-κB pathway, thereby increasing the number of TIM1+ Bregs and associated IL-10 secretion to regulate the inflammatory response, thereby protecting against liver IRI.
Collapse
Affiliation(s)
- Yu Zhang
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Cheng Zhang
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Beng Yang
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Chuanhui Peng
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Jie Zhou
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Shenli Ren
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, Zhejiang University School of Medicine Fourth Affiliated Hospital, Yiwu, Zhejiang, China
| | - Zhenhua Hu
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, Zhejiang University School of Medicine Fourth Affiliated Hospital, Yiwu, Zhejiang, China.
| |
Collapse
|
25
|
Duan X, Lv X, Wang X, Zhang Y, Hu Y, Li H, Zhou Y, Jing Y. Impact of immune cell metabolism on membranous nephropathy and prospective therapy. Commun Biol 2025; 8:405. [PMID: 40065158 PMCID: PMC11893770 DOI: 10.1038/s42003-025-07816-3] [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: 08/26/2024] [Accepted: 02/25/2025] [Indexed: 03/14/2025] Open
Abstract
Membranous nephropathy (MN) is a primary glomerular disease commonly causing adult nephrotic syndrome. Characterized by thickened glomerular capillary walls due to immune complex deposition, MN is a complex autoimmune disorder. Its pathogenesis involves immune deposit formation, complement activation, and a heightened risk of renal failure. Central to MN is immune system dysfunction, particularly the dysregulation of B and T cell responses. B cells contribute to renal injury through the production of autoantibodies, particularly IgG targeting the phospholipase A2 receptor (PLA2R) on podocytes, while T cells modulate immune responses that influence disease progression. Metabolic reprogramming alters lymphocyte survival, differentiation, proliferation, and function, potentially triggering autoimmune processes. Although the link between immune cell metabolism and MN remains underexplored, this review highlights recent advances in understanding immune metabolism and its role in MN. These insights may provide novel biomarkers and therapeutic strategies for MN treatment.
Collapse
Affiliation(s)
- Xuemei Duan
- Department of Clinical Laboratory, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Xin Lv
- Department of Nephrology, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, China
| | - Xiaocui Wang
- Department of Clinical Laboratory, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Yunfei Zhang
- Department of Clinical Laboratory, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Ying Hu
- Department of Clinical Laboratory, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Haonan Li
- Department of Clinical Laboratory, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Yongnian Zhou
- Department of Clinical Laboratory, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China.
| | - Yukai Jing
- Department of Clinical Laboratory, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China.
| |
Collapse
|
26
|
Turlej E, Domaradzka A, Radzka J, Drulis-Fajdasz D, Kulbacka J, Gizak A. Cross-Talk Between Cancer and Its Cellular Environment-A Role in Cancer Progression. Cells 2025; 14:403. [PMID: 40136652 PMCID: PMC11940884 DOI: 10.3390/cells14060403] [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: 01/30/2025] [Revised: 02/27/2025] [Accepted: 03/06/2025] [Indexed: 03/27/2025] Open
Abstract
The tumor microenvironment is a dynamic and complex three-dimensional network comprising the extracellular matrix and diverse non-cancerous cells, including fibroblasts, adipocytes, endothelial cells and various immune cells (lymphocytes T and B, NK cells, dendritic cells, monocytes/macrophages, myeloid-derived suppressor cells, and innate lymphoid cells). A constantly and rapidly growing number of studies highlight the critical role of these cells in shaping cancer survival, metastatic potential and therapy resistance. This review provides a synthesis of current knowledge on the modulating role of the cellular microenvironment in cancer progression and response to treatment.
Collapse
Affiliation(s)
- Eliza Turlej
- Departament of Molecular Physiology and Neurobiology, University of Wrocław, ul. Sienkiewicza 21, 50-335 Wrocław, Poland; (E.T.); (A.D.); (J.R.)
| | - Aleksandra Domaradzka
- Departament of Molecular Physiology and Neurobiology, University of Wrocław, ul. Sienkiewicza 21, 50-335 Wrocław, Poland; (E.T.); (A.D.); (J.R.)
| | - Justyna Radzka
- Departament of Molecular Physiology and Neurobiology, University of Wrocław, ul. Sienkiewicza 21, 50-335 Wrocław, Poland; (E.T.); (A.D.); (J.R.)
| | - Dominika Drulis-Fajdasz
- Departament of Molecular Physiology and Neurobiology, University of Wrocław, ul. Sienkiewicza 21, 50-335 Wrocław, Poland; (E.T.); (A.D.); (J.R.)
| | - Julita Kulbacka
- Departament of Molecular and Cellular Biology, Faculty of Pharmacy, Wrocław Medical University, Borowska 211A, 50-556 Wrocław, Poland;
- Department of Immunology and Bioelectrochemistry, State Research Institute Centre for Innovative Medicine, LT-08406 Vilnius, Lithuania
| | - Agnieszka Gizak
- Departament of Molecular Physiology and Neurobiology, University of Wrocław, ul. Sienkiewicza 21, 50-335 Wrocław, Poland; (E.T.); (A.D.); (J.R.)
| |
Collapse
|
27
|
Yu B, Wang X, zheng Y, Wang W, Cheng X, Cao Y, Wei M, Fu Y, Chu Y, Wang L. M2 macrophages promote IL-10 +B-cell production and alleviate asthma in mice. IMMUNOTHERAPY ADVANCES 2025; 5:ltaf007. [PMID: 40342727 PMCID: PMC12059559 DOI: 10.1093/immadv/ltaf007] [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: 11/21/2024] [Accepted: 03/06/2025] [Indexed: 05/11/2025] Open
Abstract
Introduction B cells have a central regulatory role in various diseases. While macrophages are found in the disease microenvironment and interact with tissue and diverse immune cells, their relationship with B cells remains poorly explored. Methods This study used an asthma animal model and macrophage depletion and demonstrated a significant exacerbation of asthma symptoms upon macrophage removal, coupled with a marked reduction in IL-10+ B-cell expression. Results Further analysis revealed that the macrophages interacting with IL-10+ B cells in the asthma microenvironment were of the M2 subtype. Furthermore, our sequencing data indicated a potential mechanism wherein M2 macrophages promote IL-10+ B-cell activity through the TGF-β pathway and oxidative phosphorylation pathways. Conclusion These findings suggest that M2 macrophages modulate IL-10+ B cells, ultimately mitigating asthma symptoms in mouse models.
Collapse
Affiliation(s)
- Baichao Yu
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Xueqi Wang
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Yongkun zheng
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Wenjun Wang
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Xiaoqin Cheng
- Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yue Cao
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Mingxing Wei
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Ying Fu
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Yiwei Chu
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Luman Wang
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China
- Shanghai Fifth People’s Hospital, Fudan University, Shanghai, China
| |
Collapse
|
28
|
Yadav MK, Singh SP, Egwuagu CE. IL-6/IL-12 superfamily of cytokines and regulatory lymphocytes play critical roles in the etiology and suppression of CNS autoimmune diseases. Front Immunol 2025; 16:1514080. [PMID: 40114923 PMCID: PMC11922825 DOI: 10.3389/fimmu.2025.1514080] [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: 10/19/2024] [Accepted: 02/06/2025] [Indexed: 03/22/2025] Open
Abstract
Cytokines influence cell-fate decisions of naïve lymphocytes and determine outcome of immune responses by transducing signals that regulate the initiation, intensity and duration of immune responses. However, aberrant regulation of physiological levels of cytokines contribute to the development of autoimmune and other inflammatory diseases. The Interleukin 6 (IL-6)/IL-12 superfamily of cytokines have a profound influence on all aspects of host immunity and our focus in this review is on the signaling pathways that mediate their functions, with emphasis on how this enigmatic family of cytokines promote or suppress inflammation depending on the physiological context. We also describe regulatory lymphocyte populations that suppress neuroinflammatory diseases by producing cytokines, such as IL-27 (i27-Breg) or IL-35 (i35-Breg and iTR35). We conclude with emerging immunotherapies like STAT-specific Nanobodies, Exosomes and Breg therapy that ameliorate CNS autoimmune diseases in preclinical studies.
Collapse
Affiliation(s)
| | | | - Charles E. Egwuagu
- Molecular Immunology Section, Laboratory of Immunology, National Eye Institute (NEI), National Institutes of Health (NIH), Bethesda, MD, United States
| |
Collapse
|
29
|
Leiding JW, Mathews CE, Arnold DE, Chen J. The Role of NADPH Oxidase 2 in Leukocytes. Antioxidants (Basel) 2025; 14:309. [PMID: 40227295 PMCID: PMC11939230 DOI: 10.3390/antiox14030309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Revised: 02/18/2025] [Accepted: 02/26/2025] [Indexed: 04/15/2025] Open
Abstract
NADPH oxidase (NOX) family members are major resources of intracellular reactive oxygen species (ROS). In the immune system, ROS derived from phagocytic NOX (NOX2) participate in both pathogen clearance and signaling transduction. The role of NOX2 in neutrophils and macrophages has been well studied as mutations in NOX2 subunits cause chronic granulomas disease (CGD). NOX2 is expressed across a wide range of immune cells and recent reports have demonstrated that NOX2-derived ROS play important roles in other immune cells during an immune response. In this review, we summarize current knowledge of functions of NADPH oxidase 2 in each subset of leukocytes, as well as associations of NOX2 deficiency with diseases associated specifically with autoimmunity and immune deficiency. We also discuss important knowledge gaps as well as potential future directions for NOX2 research.
Collapse
Affiliation(s)
- Jennifer W. Leiding
- Division of Allergy and Immunology, John Hopkins University, Baltimore, MD 21218, USA;
- Cancer and Blood Disorders Institute, Johns Hopkins All Children’s Hospital, St. Petersburg, FL 33701, USA
| | - Clayton E. Mathews
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL 32610, USA;
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Danielle E. Arnold
- Immune Deficiency Cellular Therapy Program, National Cancer Institutes, National Institutes of Health, Bethesda, MD 20892, USA;
| | - Jing Chen
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL 32610, USA;
| |
Collapse
|
30
|
Sapra L, Saini C, Mishra PK, Garg B, Gupta M, Srivastava RK. Compromised anti-osteoclastogenic and immunomodulatory functions of regulatory B cells (Bregs) aggravate inflammatory bone loss in post-menopausal osteoporosis. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167675. [PMID: 39826852 DOI: 10.1016/j.bbadis.2025.167675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 01/09/2025] [Accepted: 01/12/2025] [Indexed: 01/22/2025]
Abstract
Regulatory-B-cells (Bregs) modulate immune-homeostasis. Variations in the number and function of Bregs have been associated with various immune-related ailments, highlighting the importance of Bregs under inflammatory-conditions. Previously, we discovered the anti-osteoclastogenic-potential of Bregs. However, the crucial role of Bregs in the onset and progression of post-menopausal osteoporosis (PMO) has never been explored. Interestingly, our temporal-kinetic study demonstrated that Bregs have a compromised dynamics and functionality which further contributes to inception and the progression of the inflammatory bone loss condition in the PMO. Our ex-vivo findings further elucidate a significant reduction in the immunomodulatory and anti-osteoclastogenic functions of Bregs under the estrogen deficient post-menopausal osteoporotic conditions. The current study for the first time reports the crucial role of dysregulated-Bregs (both functionally and numerically), in the development of PMO. Our findings thereby provide a novel concept for immuno-therapeutically targeting "Bregs", for effective management and treatment of post-menopausal osteoporosis in the long run.
Collapse
Affiliation(s)
- Leena Sapra
- Translational Immunology, Osteoimmunology & Immunoporosis Lab (TIOIL), ICMR-Collaborative Centre for Excellence in Bone Health, Department of Biotechnology, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| | - Chaman Saini
- Translational Immunology, Osteoimmunology & Immunoporosis Lab (TIOIL), ICMR-Collaborative Centre for Excellence in Bone Health, Department of Biotechnology, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| | - Pradyumna Kumar Mishra
- Department of Molecular Biology, ICMR-National Institute for Research in Environmental Health, Bhopal, MP 462001, India
| | - Bhavuk Garg
- Department of Orthopaedics, All India Institute of Medical Sciences (AIIMS), New Delhi s, India
| | - Manish Gupta
- Department of Orthopaedics, All India Institute of Medical Sciences (AIIMS), New Delhi s, India
| | - Rupesh K Srivastava
- Translational Immunology, Osteoimmunology & Immunoporosis Lab (TIOIL), ICMR-Collaborative Centre for Excellence in Bone Health, Department of Biotechnology, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India.
| |
Collapse
|
31
|
Liu T, Wang W, Du J, Liu H, Wu J, Wang C, Tang M, Liu Y, Ju Y, Qu W, Zheng J, Zhao Y, Zhang Y. Aluminum promotes B1 cells to produce IL-10 and impairs adaptive immune system. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2025; 368:125791. [PMID: 39914566 DOI: 10.1016/j.envpol.2025.125791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Revised: 01/31/2025] [Accepted: 02/02/2025] [Indexed: 02/09/2025]
Abstract
Aluminum (Al) is a metal existing in the human body, yet the immunotoxicity of Al remains elusive. To investigate the immunotoxicity of Al, C57BL/6 mice were treated with 200 or 800 ppm Al via drinking water for 3 months, and thereafter the adaptive immune system was evaluated. In addition, mouse splenocytes and human peripheral blood mononuclear cells (PBMC) were treated with Al in vitro to assess the impact of Al in vitro. Treatment with Al reduced the production of IgM and IgG in the serum, and the activation of B cells, CD4 T cells and CD8 T cells in the spleen of mice; treatment with Al in vitro suppressed the production of IgM and IgG, and the activation of B cells, CD4 T cells and CD8 T cells in mouse splenocytes and human PBMC. In vitro co-culture assays suggested that the suppressed adaptive immunity was due to B cells modified by Al. In terms of mechanism, a direct action of Al on B1 cells induced the B1 cells to be IL-10-producing cells and thereby suppressed the adaptive immune system, which was critically dependent on the Jak1/3-STAT signaling. This study reveals that Al suppresses the adaptive immunity via induction of IL-10-producing B1 cells.
Collapse
Affiliation(s)
- Ting Liu
- School of Public Health, School of Public Health and Key Laboratory of Public Health Safety, MOE, Fudan University, Shanghai, 200032, China
| | - Wei Wang
- School of Public Health, School of Public Health and Key Laboratory of Public Health Safety, MOE, Fudan University, Shanghai, 200032, China
| | - Jun Du
- Amway (Shanghai) Innovation & Science Co., Ltd, Shanghai, 201203, China
| | - Hongyue Liu
- Amway (Shanghai) Innovation & Science Co., Ltd, Shanghai, 201203, China
| | - Jiaojiao Wu
- School of Public Health, School of Public Health and Key Laboratory of Public Health Safety, MOE, Fudan University, Shanghai, 200032, China
| | - Chuanxuan Wang
- School of Public Health, School of Public Health and Key Laboratory of Public Health Safety, MOE, Fudan University, Shanghai, 200032, China
| | - MengKe Tang
- School of Public Health, School of Public Health and Key Laboratory of Public Health Safety, MOE, Fudan University, Shanghai, 200032, China
| | - Yalin Liu
- School of Public Health, School of Public Health and Key Laboratory of Public Health Safety, MOE, Fudan University, Shanghai, 200032, China
| | - Yingzi Ju
- School of Public Health, School of Public Health and Key Laboratory of Public Health Safety, MOE, Fudan University, Shanghai, 200032, China
| | - Weidong Qu
- School of Public Health, School of Public Health and Key Laboratory of Public Health Safety, MOE, Fudan University, Shanghai, 200032, China
| | - Jianheng Zheng
- Amway (Shanghai) Innovation & Science Co., Ltd, Shanghai, 201203, China.
| | - Yifan Zhao
- School of Public Health, School of Public Health and Key Laboratory of Public Health Safety, MOE, Fudan University, Shanghai, 200032, China.
| | - Yubin Zhang
- School of Public Health, School of Public Health and Key Laboratory of Public Health Safety, MOE, Fudan University, Shanghai, 200032, China
| |
Collapse
|
32
|
Dai Q, Peng Y, He P, Wu X. Interactions and communications in the prostate tumour microenvironment: evolving towards effective cancer therapy. J Drug Target 2025; 33:295-315. [PMID: 39445641 DOI: 10.1080/1061186x.2024.2418344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 10/02/2024] [Accepted: 10/14/2024] [Indexed: 10/25/2024]
Abstract
Prostate cancer is one of the most common malignancies in men. The tumour microenvironment (TME) has a critical role in the initiation, progression, and metastasis of prostate cancer. TME contains various cell types, including cancer-associated fibroblasts (CAFs), endothelial cells, immune cells such as macrophages, lymphocytes B and T, natural killer (NK) cells, and other proteins such as extracellular matrix (ECM) components. The interactions and communications between these cells within the TME are crucial for the growth and response of various solid tumours, such as prostate cancer to different anticancer modalities. In this review article, we exemplify the various mechanisms by which the TME influences prostate cancer progression. The roles of different cells, cytokines, chemokines, and growth factors in modulating the immune response and prostate tumour growth will be discussed. The impact of these cells and factors and other ECM components on tumour cell invasion and metastasis will also be discussed. We explain how these interactions in TME can affect the response of prostate cancer to therapy. We also highlight the importance of understanding these interactions to develop novel therapeutic approaches for prostate cancer.
Collapse
Affiliation(s)
- Qiang Dai
- Department of Urology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Yanling Peng
- Department of Urology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Peng He
- Department of Urology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Xiaojun Wu
- Department of Urology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| |
Collapse
|
33
|
Martinis E, Tonon S, Colamatteo A, La Cava A, Matarese G, Pucillo CEM. B cell immunometabolism in health and disease. Nat Immunol 2025; 26:366-377. [PMID: 39984733 DOI: 10.1038/s41590-025-02102-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 01/15/2025] [Indexed: 02/23/2025]
Abstract
B cells have crucial roles in the initiation and progression of many pathological conditions, and several therapeutic strategies have targeted the function of these cells. The advent of immunometabolism has provided compelling evidence that the metabolic reprogramming of immune cells can dramatically alter physiopathological immune activities. A better knowledge of the metabolic profiles of B cells can provide valuable means for developing therapies tuning defined cell pathways. Here we review the cellular and molecular mechanisms by which immunometabolism controls the physiology and pathophysiology of B cells and discuss the experimental evidence linking B cell metabolism to health, autoimmunity, and cancer. Considering that several metabolic pathways in B cells are involved differently, or even in opposite ways, in health and disease, we discuss how targeted modulation of B cell immunometabolism could be exploited mechanistically to rebalance abnormal B cell functions that have become altered in disease states.
Collapse
Affiliation(s)
| | - Silvia Tonon
- Department of Medicine, University of Udine, Udine, Italy
| | - Alessandra Colamatteo
- Treg Cell Lab, Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli 'Federico II', Napoli, Italy
| | - Antonio La Cava
- Treg Cell Lab, Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli 'Federico II', Napoli, Italy
- Department of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Giuseppe Matarese
- Treg Cell Lab, Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli 'Federico II', Napoli, Italy.
- Laboratorio di Immunologia, Istituto per l'Endocrinologia e l'Oncologia Sperimentale 'G. Salvatore' - Consiglio Nazionale delle Ricerche (IEOS-CNR), Napoli, Italy.
| | | |
Collapse
|
34
|
Ye R, Li S, Li Y, Shi K, Li L. Revealing the role of regulatory b cells in cancer: development, function and treatment significance. Cancer Immunol Immunother 2025; 74:125. [PMID: 39998678 PMCID: PMC11861783 DOI: 10.1007/s00262-025-03973-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Accepted: 02/07/2025] [Indexed: 02/27/2025]
Abstract
B cells are essential components of the immune response, primarily recognized for their ability to produce antibodies. However, emerging research reveals their important roles in regulating immune responses and influencing tumor development, independent of antibodies. The connection between tumor progression and alterations in the tumor microenvironment is well-established, as immune infiltrating cells can enhance the survival of tumor cells by modifying their surroundings. Despite this, the majority of studies have focused on T cells and macrophages, creating a gap in our understanding of B cells. Regulatory B cells (Bregs) represent a crucial subpopulation that plays a significant role in maintaining immune balance. They may have a substantial impact on tumor immunity by negatively regulating tumor-infiltrating immune cells. This paper reviews the existing literature on Bregs, examining their development, phenotypes, functions, and the mechanisms through which they exert their regulatory effects. Furthermore, we highlight their potential interventional roles and prognostic significance in cancer therapy. By addressing the current gaps in knowledge regarding Bregs within tumors, we hope to inspire further research that could lead to innovative cancer treatments and improved outcomes for patients.
Collapse
Affiliation(s)
- Ruyu Ye
- Department of Hematology, The Second Hospital of Dalian Medical University, Dalian, People's Republic of China
| | - Sijia Li
- Department of Hematology, The Second Hospital of Dalian Medical University, Dalian, People's Republic of China
| | - Yuxiao Li
- Department of Hematology, The Second Hospital of Dalian Medical University, Dalian, People's Republic of China
| | - Kaixin Shi
- Department of Hematology, The Second Hospital of Dalian Medical University, Dalian, People's Republic of China
| | - Li Li
- Department of Hematology, The Second Hospital of Dalian Medical University, Dalian, People's Republic of China.
| |
Collapse
|
35
|
Muraoka S, Baba T, Akazawa T, Katayama KI, Kusumoto H, Yamashita S, Kohjimoto Y, Iwabuchi S, Hashimoto S, Hara I, Inoue N. Tumor-derived lactic acid promotes acetylation of histone H3K27 and differentiation of IL-10-producing regulatory B cells through direct and indirect signaling pathways. Int J Cancer 2025; 156:840-852. [PMID: 39482832 DOI: 10.1002/ijc.35229] [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] [Revised: 10/02/2024] [Accepted: 10/08/2024] [Indexed: 11/03/2024]
Abstract
Tumor cells are known to enhance glycolysis, even under normoxic conditions, via the Warburg effect, producing excess lactic acid in the tumor microenvironment. Lactic acid enhances the IL-23/IL-17 pathway and induces chronic inflammation. The acidic microenvironment formed by lactic acid suppresses immune cell proliferation and activation. In the present study, we clarified that lactic acid had two novel activities for immune cells. First, lactic acid specifically enhanced acetylation at lysine 27 of histone H3 (H3K27ac) in splenic B cells and monocytes/macrophages, and this epigenetically up-regulates the expression of genes. Acetylation and methylation of other residues of histone H3 were rarely induced. Second, lactic acid induced a particularly-marked enhancement of Il10 gene expression in B cells, leading to an increase in IL-10-producing regulatory B (Breg) cells. Furthermore, two pathways should be involved in both the enhancement of H3K27ac and the induction of Breg cells by lactic acid: a direct pathway that enhances the CD40 signal in B cells, and an indirect pathway that affects B cells by activating the exchange protein directly activated by cAMP (EPAC) 1/2 in non-B cells. In tumor-bearing mice, the levels of H3K27ac of tumor-infiltrating B cells were significantly higher than splenic B cells and were suppressed by intraperitoneal injection of the EPAC1/2 inhibitor. In conclusion, tumor-derived lactic acid increases H3K27ac and IL-10-producing Breg cells, causing the suppression of anti-tumor immunity.
Collapse
Affiliation(s)
- Satoshi Muraoka
- Department of Urology, Wakayama Medical University, Wakayama, Japan
| | - Takashi Baba
- Department of Molecular Genetics, Wakayama Medical University, Wakayama, Japan
| | - Takashi Akazawa
- Department of Cancer Drug Discovery and Development, Research Center, Osaka International Cancer Institute, Chuo-ku, Osaka, Japan
| | - Kei-Ichi Katayama
- Department of Molecular Genetics, Wakayama Medical University, Wakayama, Japan
| | - Hiroki Kusumoto
- Department of Urology, Wakayama Medical University, Wakayama, Japan
| | | | - Yasuo Kohjimoto
- Department of Urology, Wakayama Medical University, Wakayama, Japan
| | - Sadahiro Iwabuchi
- Department of Molecular Pathophysiology, Wakayama Medical University, Wakayama, Japan
| | - Shinichi Hashimoto
- Department of Molecular Pathophysiology, Wakayama Medical University, Wakayama, Japan
| | - Isao Hara
- Department of Urology, Wakayama Medical University, Wakayama, Japan
| | - Norimitsu Inoue
- Department of Molecular Genetics, Wakayama Medical University, Wakayama, Japan
| |
Collapse
|
36
|
Aslanian-Kalkhoran L, Nouri N, Soltani-Zangbar MS, Mardi A, Aghebati-Maleki L. Immunoglobulin therapy for infertility and the role of immune cells in pregnancy success: An extensive investigation and update. J Reprod Immunol 2025; 169:104458. [PMID: 40015106 DOI: 10.1016/j.jri.2025.104458] [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: 12/31/2024] [Revised: 02/02/2025] [Accepted: 02/13/2025] [Indexed: 03/01/2025]
Abstract
In the United States, roughly one out of every eight couples, or 7.5 million women, experience challenges related to conceiving or maintaining a pregnancy. The body's immune response is vital during pregnancy. T cells, natural killer (NK) cells, B cells, and macrophages (MQ) are immune cells in the female reproductive tract. They are in charge of maintaining tissue homeostasis and regulating the immune system's response to invasive pathogens. Failure to regulate these immune cells might result in inflammation, which reduces fertility. The immune system modulation of pregnancy loss has been studied with intralipid, intravenous immunoglobulin (IVIG), and paternal leukocyte vaccination. A concentrated antibody called intravenous immunoglobulin (IVIG) is utilized as a biological agent to treat autoimmune, viral, and inflammatory diseases and some immunodeficiencies. The main objective of this treatment is to restore a damaged immune system. IgGs, through binding to specific antigens, promote the innate immunity's cellular and humoral immune response by activating complements and binding to Fc receptors of several immune cells. Contrariwise, IVIG regulates pathogenic autoimmunity in animal models, including skin-blister diseases, nephrotoxic nephritis, and K/BxN arthritis. IVIG has, therefore, been of great interest as an immune modulator in several immune disorders. This review aims to investigate the immunological reasons of reproductive failure, focusing on the immunomodulatory effects of IVIG in its treatment.
Collapse
Affiliation(s)
- Lida Aslanian-Kalkhoran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Narjes Nouri
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Amirhossein Mardi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran; Student's Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Leili Aghebati-Maleki
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
37
|
Yang J, Tang S, Saba NF, Shay C, Teng Y. Tumor secretome shapes the immune landscape during cancer progression. J Exp Clin Cancer Res 2025; 44:47. [PMID: 39930476 PMCID: PMC11809007 DOI: 10.1186/s13046-025-03302-0] [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: 12/22/2024] [Accepted: 01/24/2025] [Indexed: 02/14/2025] Open
Abstract
The focus of cancer immunotherapy has traditionally been on immune cells and tumor cells themselves, often overlooking the tumor secretome. This review provides a comprehensive overview of the intricate relationship between tumor cells and the immune response in cancer progression. It highlights the pivotal role of the tumor secretome - a diverse set of molecules secreted by tumor cells - in significantly influencing immune modulation, promoting immunosuppression, and facilitating tumor survival. In addition to elucidating these complex interactions, this review discusses current clinical trials targeting the tumor secretome and highlights their potential to advance personalized medicine strategies. These trials aim to overcome the challenges of the tumor microenvironment by designing therapies tailored to the secretome profiles of individual cancer patients. In addition, advances in proteomic techniques are highlighted as essential tools for unraveling the complexity of the tumor secretome, paving the way for improved cancer treatment outcomes.
Collapse
Affiliation(s)
- Jianqiang Yang
- Department of Hematology and Medical Oncology, Emory University, 201 Dowman Dr, Atlanta, GA, 30322, USA
- Winship Cancer Institute of Emory University, Atlanta, GA, 30322, USA
| | - Sijia Tang
- Department of Hematology and Medical Oncology, Emory University, 201 Dowman Dr, Atlanta, GA, 30322, USA
- Winship Cancer Institute of Emory University, Atlanta, GA, 30322, USA
| | - Nabil F Saba
- Department of Hematology and Medical Oncology, Emory University, 201 Dowman Dr, Atlanta, GA, 30322, USA
- Winship Cancer Institute of Emory University, Atlanta, GA, 30322, USA
| | - Chloe Shay
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, 30322, USA
| | - Yong Teng
- Department of Hematology and Medical Oncology, Emory University, 201 Dowman Dr, Atlanta, GA, 30322, USA.
- Winship Cancer Institute of Emory University, Atlanta, GA, 30322, USA.
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, 30322, USA.
| |
Collapse
|
38
|
Su X, Zhang M, Zhu H, Cai J, Wang Z, Xu Y, Wang L, Shen C, Cai M. Mechanisms of T-cell Depletion in Tumors and Advances in Clinical Research. Biol Proced Online 2025; 27:5. [PMID: 39905296 PMCID: PMC11792740 DOI: 10.1186/s12575-025-00265-6] [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: 11/10/2024] [Accepted: 01/20/2025] [Indexed: 02/06/2025] Open
Abstract
T lymphocytes (T cells) are essential components of the adaptive immune system that play a vital role in identifying and eliminating infected and tumor cells. In tumor immunotherapy, T cells have emerged as a promising therapeutic strategy due to their high specificity, potent cytotoxic capability, long-lasting immune memory, and adaptability within immunotherapeutic approaches. However, tumors can evade the immune system by depleting T cells through various mechanisms, such as inhibitory receptor signaling, metabolic exhaustion, and physical barriers within the tumor microenvironment. This review provided an overview of the mechanisms underlying T-cell depletion in tumors and discussed recent advances in clinical research related to T-cell immunotherapy for tumors. It highlighted the need for in-depth studies on key issues such as indications, dosage, and sequencing of combined therapeutic strategies tailored to different patients and tumor types, providing practical guidance for individualized treatment. Future research on T-cell depletion would be necessary to uncover the fundamental mechanisms and laws of T-cell depletion, offering both theoretical insights and practical guidance for the selection and optimization of tumor immunotherapy. Furthermore, interdisciplinary, cross-disciplinary, and international collaborative innovations are necessary for developing more effective and safer treatments for tumor patients.
Collapse
Affiliation(s)
- Xiangfei Su
- China Association of Chinese Medicine, Beijing, China
| | - Mi Zhang
- Department of Pharmacy, The Second Affiliated Hospital of Anhui University of Chinese Medicine, No. 300, Shouchun Road, Hefei, Anhui, 230061, China
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Hong Zhu
- Tongling People's Hospital, Tongling, Anhui, China
| | - Jingwen Cai
- Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui, China
| | - Zhen Wang
- Anhui Provincial Children's Hospital, Hefei, Anhui, China
| | - Yuewei Xu
- Department of Pharmacy, The Second Affiliated Hospital of Anhui University of Chinese Medicine, No. 300, Shouchun Road, Hefei, Anhui, 230061, China
| | - Li Wang
- Department of Pharmacy, The Second Affiliated Hospital of Anhui University of Chinese Medicine, No. 300, Shouchun Road, Hefei, Anhui, 230061, China
| | - Chen Shen
- Key Laboratory of Data Science and Innovation and Development of Traditional Chinese Medicine and Social Sciences of Anhui Province, Anhui University of Chinese Medicine, No. 350, Longzihu Road, Hefei, Anhui, 230012, China.
| | - Ming Cai
- Department of Pharmacy, The Second Affiliated Hospital of Anhui University of Chinese Medicine, No. 300, Shouchun Road, Hefei, Anhui, 230061, China.
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui, China.
| |
Collapse
|
39
|
Zhao D, Sui Y, Sun Y. Causal Relationship Between Immune Cells and Hypopituitarism: Bidirectional Mendelian Randomization Study. World Neurosurg 2025; 194:123411. [PMID: 39522807 DOI: 10.1016/j.wneu.2024.10.140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND Hypopituitarism, one or more pituitary hormones inefficiently produced by the anterior pituitary or released from the posterior pituitary to adapt to the needs of the organism. Existing epidemiological data show that immune-mediated diffuse infiltration of the anterior pituitary is important in the development of hypopituitarism. However, the precise connection between immune cells and hypopituitarism remains unclear. This study aimed to elucidate the potential causal links between the 731 immune cell types and hypopituitarism risk. METHODS Based on data from a genome-wide association study, a bidirectional two-sample Mendelian randomization analysis was performed using 5 methods to explore the potential influence of immune cell phenotypes on hypopituitarism. Sensitivity analyses were conducted to examine the robustness of these findings. RESULTS Our findings support that B cells, T cells, Tregs, dendritic cells, monocytes, and myeloid cells each have a bidirectional influence on hypopituitarism. One B-cell phenotype was associated with increased hypopituitarism risk, while 2 B-cell phenotypes play a protective role in hypopituitarism. Moreover, 7 T-cell phenotypes demonstrate significant protective properties on hypopituitarism. Seven Tregs were associated with increased hypopituitarism risk. Furthermore, one monocyte was identified to be significantly associated with hypopituitarism risk. In addition, reverse Mendelian randomization analysis revealed that hypopituitarism was positively associated with 30 additional immune cell phenotypes and negatively associated with 17 immune cells. CONCLUSIONS Our investigation shed light on the intricate potential relationship between immune cells and hypopituitarism via genetic methods, underscoring the immune-mediated pathway in hypopituitarism pathogenesis, thereby offering valuable insights for future clinical investigations.
Collapse
Affiliation(s)
- Dadi Zhao
- Department of Neurosurgery, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
| | - Yuan Sui
- Department of Neurosurgery, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
| | - Yesheng Sun
- Department of Cellular Neurobiology, Zoological Institute, Technische Universität Braunschweig, Braunschweig, Germany.
| |
Collapse
|
40
|
Wiley KS, Martínez LE, Kwon D, Knorr DA, Epeldegui M, Fox MM. Regulatory B-Cells Are Associated Negatively With Regulatory T-Cells and Positively With Cytokines in Peripheral Blood of Pregnant Women. Am J Reprod Immunol 2025; 93:e70027. [PMID: 39854121 PMCID: PMC12001748 DOI: 10.1111/aji.70027] [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: 02/22/2024] [Revised: 09/27/2024] [Accepted: 11/27/2024] [Indexed: 01/26/2025] Open
Abstract
PROBLEM Regulatory B-cells (Bregs, CD19+CD24hiCD38hi) are a specialized B-cell subset that suppresses immune responses and potentially contribute to the maintenance of an immune-privileged environment for fetal development during pregnancy. However, little is known about the surrounding immunological environment of Bregs in gestational physiology. The relationship of regulatory T-cells (Tregs, CD4+CD25hiCD127loFoxP3+) to Bregs in coordinating immunoregulation during pregnancy is unknown. We aimed to determine whether peripheral concentrations of Bregs and/or PD-L1-expressing Bregs correlated with Tregs and cytokines during pregnancy. METHOD Peripheral blood samples were obtained from 29 pregnant women at mean 12 weeks' gestation. Participants were age ≥ 18, self-identified as Latina/Hispanic, and N = 12 primigravid. Peripheral blood mononuclear cells were isolated, stained, and analyzed by flow cytometry to determine percentages of Tregs from CD4+ T-cells and five Treg subsets defined by immune checkpoint markers, and Bregs and PD-L1+ Bregs from total B-cells. Levels of 13 cytokines were measured on a Meso Scale Discovery multiplex platform. RESULTS Bregs positively correlated with pro-inflammatory cytokine interleukin (IL)-6. PD-L1+ Bregs positively correlated with T-cell suppressive cytokine IL-10. PD-L1+ Bregs negatively correlated with Tregs and Helios+, CTLA-4+, PD-1+, TIGIT+, and TIM3+ Tregs. For primigravida, PD-L1+ Bregs correlated positively with IL-10 and negatively with Helios+ and TIGIT+ Tregs. For multigravida, PD-L1+ Bregs correlated positively with IL-8 and negatively with Helios+, CTLA-4+, PD-1+, and TIGIT+ Tregs. CONCLUSIONS This study provides insight into the immunosuppressive role of Bregs and PD-L1+ Bregs during human pregnancy. Our results suggest that PD-L1+ Bregs can employ suppressive mechanisms to limit pro-inflammatory responses in primigravida.
Collapse
Affiliation(s)
- Kyle S. Wiley
- Department of Anthropology, University of California, Los Angeles, California, USA
| | - Laura E. Martínez
- UCLA AIDS Institute and David Geffen School of Medicine, University of California, Los Angeles, California, USA
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California, Los Angeles, California, USA
| | - Dayoon Kwon
- Department of Epidemiology, UCLA Fielding School of Public Health, University of California, Los Angeles, California, USA
| | - Delaney A. Knorr
- Department of Anthropology, University of California, Los Angeles, California, USA
| | - Marta Epeldegui
- UCLA AIDS Institute and David Geffen School of Medicine, University of California, Los Angeles, California, USA
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California, Los Angeles, California, USA
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, California, USA
| | - Molly M. Fox
- Department of Anthropology, University of California, Los Angeles, California, USA
- Department of Psychiatry & Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California, Los Angeles, California, USA
| |
Collapse
|
41
|
Xing C, Cui H, Li G, Liu X, Liu K, Wen Q, Huang X, Wang R, Song L. Hspa13 Deficiency Impaired Marginal Zone B Cells Regulatory Function and Contributed to Lupus Pathogenesis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2413144. [PMID: 39737854 PMCID: PMC11848637 DOI: 10.1002/advs.202413144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 12/11/2024] [Indexed: 01/01/2025]
Abstract
Dysregulated IL-10 producing regulatory B cells (Bregs) are associated with the progression of systemic lupus erythematosus. An immunomodulatory role of heat shock proteins (HSPs) is implicated in autoimmune diseases. However, the molecular basis underlying the role of Hspa13 in regulating Bregs function and lupus pathogenesis remains unclear. In this study, Bregs display higher Hspa13 expression than IL-10- B cells. Induction of IL-10 production is weakened in B cells with Hspa13 knockdown or knockout. Hspa13 binds to the IL-10 promoter via the TATA or CAAT box and activates IL-10 transcription in the nucleus. Furthermore, Hspa13 positive cells are enriched in marginal zone (MZ) B cells to regulate IL-10 production. Stimulated B220+ B or MZ B cells from CD19creHspa13fl/fl mice for Breg induction show an impaired capacity to promote CD4+Foxp3+ regulatory T cells (Treg) differentiation. In lupus MRL/lpr mice, a decline in Treg differentiation is accompanied by decreased Hspa13 expression in both Bregs and MZ B cells. Moreover, adoptive transfusion of Bregs and MZ B cells from CD19creHspa13fl/fl mice fails to increase the frequency of Tregs, attenuate renal pathology, or decrease anti-dsDNA antibody levels. These results explain the unique role of Hspa13 in determining MZ regulatory function and affecting lupus pathogenesis.
Collapse
MESH Headings
- Animals
- Lupus Erythematosus, Systemic/immunology
- Lupus Erythematosus, Systemic/metabolism
- Lupus Erythematosus, Systemic/genetics
- Lupus Erythematosus, Systemic/pathology
- Mice
- B-Lymphocytes, Regulatory/immunology
- B-Lymphocytes, Regulatory/metabolism
- Interleukin-10/metabolism
- Interleukin-10/genetics
- Mice, Inbred MRL lpr
- HSP70 Heat-Shock Proteins/genetics
- HSP70 Heat-Shock Proteins/metabolism
- HSP70 Heat-Shock Proteins/deficiency
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
- Mice, Knockout
- Female
- Disease Models, Animal
- Mice, Inbred C57BL
Collapse
Affiliation(s)
- Chen Xing
- Beijing Institute of Basic Medical SciencesBeijing100850China
| | - Haoran Cui
- Beijing Institute of Basic Medical SciencesBeijing100850China
| | - Ge Li
- Beijing Institute of Basic Medical SciencesBeijing100850China
| | - Xiaoling Liu
- Department of DermatologyFirst Medical Centre of ChinesePLA General HospitalBeijing100853China
| | - Kun Liu
- Beijing Institute of Basic Medical SciencesBeijing100850China
| | - Qing Wen
- Beijing Institute of Basic Medical SciencesBeijing100850China
| | - Xin Huang
- Beijing Institute of Basic Medical SciencesBeijing100850China
| | - Renxi Wang
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and TechnologyCollaborative Innovation Center for Brain DisordersCapital Medical UniversityBeijing100069China
| | - Lun Song
- Beijing Institute of Basic Medical SciencesBeijing100850China
| |
Collapse
|
42
|
Pan Z, Li M, Zhang P, Li T, Liu R, Liu J, Liu S, Zhang Y. Peripheral Blood Lymphocyte Subsets and Heterogeneity of B Cell Subsets in Patients of Idiopathic Inflammatory Myositis with Different Myositis-specific Autoantibodies. Inflammation 2025; 48:118-132. [PMID: 38755405 DOI: 10.1007/s10753-024-02052-z] [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: 02/06/2024] [Revised: 05/08/2024] [Accepted: 05/10/2024] [Indexed: 05/18/2024]
Abstract
Idiopathic inflammatory myopathies (IIM) are a group of myopathies that present with muscle weakness and multiple extra-muscular manifestations, in which lymphocytes play central roles in myositis pathogenesis. This study aimed to explore the clinical characteristics of lymphocyte subsets, especially B cell subsets, in patients with IIM. Our study included 176 patients with active IIM and 210 gender/age-matched healthy controls (HCs). Compared to HCs, patients have reduced counts of T cells, B cells, and natural killer cells. In addition, B cell subsets from 153 patients with IIM and 92 HCs were characterized. Patients had a lower percentage of memory B cells and translational memory B cells, while those patients were with an elevated percentage of CD19+ B cells, plasmablast and naïve B cells compared with HCs. Moreover, to further explore the heterogeneity of B cells in IIM, patients were categorized into three clusters based on clustering analysis. Cluster 1 was dominated by CD19+ B cells, Bregs and naïve B cells, cluster 3 was dominated by memory B cells and plasmablast, and cluster 2 had the highest proportion of translational memory B cells. Notably, patients in cluster 1 presented with higher CK levels, indicating muscle damage, whereas patients in cluster 3 showed a higher incidence of chest tightness. Our study indicated that lymphopenia is a common manifestation in patients with IIM. B cell subsets are abnormally expressed and showed high heterogeneity in patients with IIM. The patients with IIM were divided into three different clusters with different percentages of chest tightness and distinct CK levels.
Collapse
Affiliation(s)
- Zhou Pan
- Department of Rheumatology and Clinical Immunology, The first affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Mengdi Li
- Department of Rheumatology and Clinical Immunology, The first affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Rheumatology and Clinical Immunology, Zhongshan Hospital of Fudan University, Shanghai, China
| | - Panpan Zhang
- Department of Rheumatology and Clinical Immunology, The first affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Tianqi Li
- Department of Rheumatology and Clinical Immunology, The first affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Rui Liu
- Department of Rheumatology and Clinical Immunology, The first affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jia Liu
- Department of Rheumatology and Clinical Immunology, The first affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Shengyun Liu
- Department of Rheumatology and Clinical Immunology, The first affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Yusheng Zhang
- Department of Rheumatology and Clinical Immunology, The first affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
43
|
McDaniel Mims B, Furr KL, Enriquez J, Grisham MB. Improving bench-to-bedside translation for acute graft-versus-host disease models. Dis Model Mech 2025; 18:DMM052084. [PMID: 40019007 PMCID: PMC11892683 DOI: 10.1242/dmm.052084] [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] [Indexed: 03/01/2025] Open
Abstract
The transplantation of allogeneic hematopoietic stem cells is a potentially curative treatment for hematological malignancies, inherited blood disorders and immune deficiencies. Unfortunately, 30-50% of patients receiving allogeneic hematopoietic stem cells will develop a potentially life-threatening inflammatory disease called acute graft-versus-host disease (aGVHD). In patients with aGVHD, graft-associated T cells, which typically target the skin, intestinal tract and liver, can also damage the lungs and lymphoid tissue. Damage to lymphoid tissue creates prolonged immunodeficiency that markedly increases the risk of infections and bleeding, resulting in considerable morbidity and mortality. Although mouse models of aGVHD have been instrumental to our understanding of this condition's pathogenesis, translation of preclinical data into new and more effective treatments for human disease has been limited for reasons that remain to be fully understood. However, evidence suggests that factors associated with mouse models of aGVHD likely contribute to these unsatisfactory results. In this Review, we identify and discuss the specific factors inherent to mouse models of aGVHD that may limit the translation of preclinical data to patient treatment, and suggest how to improve the translatability of these models.
Collapse
Affiliation(s)
- Brianyell McDaniel Mims
- Department of Oral Health Sciences, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Kathryn L. Furr
- Department of Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, Lubbock, TX 79423, USA
| | - Josue Enriquez
- Department of Microbiology and Immunology, University of Gothenburg, Gothenburg 405 30, Sweden
| | - Matthew B. Grisham
- Department of Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, Lubbock, TX 79423, USA
| |
Collapse
|
44
|
Song S, Wang C, Chen Y, Zhou X, Han Y, Zhang H. Dynamic roles of tumor-infiltrating B lymphocytes in cancer immunotherapy. Cancer Immunol Immunother 2025; 74:92. [PMID: 39891668 PMCID: PMC11787113 DOI: 10.1007/s00262-024-03936-7] [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: 12/04/2024] [Accepted: 12/27/2024] [Indexed: 02/03/2025]
Abstract
The amazing diversity of B cells within the tumor microenvironment is the basis for the diverse development of B cell-based immunotherapies. Here, we focus on elucidating the mechanisms of tumor intervention mediated by four tumor-infiltrating B lymphocytes. Naive B cells present the initial antigen, germinal center B cell subsets enhance antibody affinity, and immunoglobulin subtypes exert multiple immune effects, while regulatory B cells establish immune tolerance. Together they reflect the complexity of the changing dynamics of cancer immunity. Additionally, we have investigated the dynamic effects of tumor-infiltrating B lymphocytes in immunotherapy and their relationship to prognosis, providing new insights into potential treatment strategies for patients.
Collapse
Affiliation(s)
- Shishengnan Song
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, China
| | - Chong Wang
- Department of Thoracic Surgery, Beijing Chest Hospital Affiliated to Capital Medical University (Beijing Tuberculosis and Thoracic Tumor Research Institute), 9 Beiguan Street, Tongzhou, 101149, Beijing, China
| | - Yangchao Chen
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, 999077, NT, China
| | - Xiaorong Zhou
- Department of Immunology, Medical School of Nantong University, 19 Qixiu Road, Nantong, 226001, Jiangsu, China.
| | - Yi Han
- Department of Thoracic Surgery, Beijing Chest Hospital Affiliated to Capital Medical University (Beijing Tuberculosis and Thoracic Tumor Research Institute), 9 Beiguan Street, Tongzhou, 101149, Beijing, China.
| | - Haijian Zhang
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, China.
| |
Collapse
|
45
|
Giakomidi D, Ishola A, Nus M. Targeting gut microbiota to regulate the adaptive immune response in atherosclerosis. Front Cardiovasc Med 2025; 12:1502124. [PMID: 39957996 PMCID: PMC11825770 DOI: 10.3389/fcvm.2025.1502124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 01/20/2025] [Indexed: 02/18/2025] Open
Abstract
Atherosclerosis, the leading cause of death worldwide, is a chronic inflammatory disease leading to the accumulation of lipid-rich plaques in the intima of large and medium-sized arteries. Accumulating evidence indicates the important regulatory role of the adaptive immune system in atherosclerosis during all stages of the disease. The gut microbiome has also become a key regulator of atherosclerosis and immunomodulation. Whilst existing research extensively explores the impact of the microbiome on the innate immune system, only a handful of studies have explored the regulatory capacity of the microbiome on the adaptive immune system to modulate atherogenesis. Building on these concepts and the pitfalls on the gut microbiota and adaptive immune response interaction, this review explores potential strategies to therapeutically target the microbiome, including the use of prebiotics and vaccinations, which could influence the adaptive immune response and consequently plaque composition and development.
Collapse
Affiliation(s)
- Despina Giakomidi
- Cardiovascular Division, Department of Medicine, Heart and Lung Research Institute (HLRI), University of Cambridge, Cambridge, United Kingdom
- British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge, United Kingdom
| | - Ayoola Ishola
- Cardiovascular Division, Department of Medicine, Heart and Lung Research Institute (HLRI), University of Cambridge, Cambridge, United Kingdom
| | - Meritxell Nus
- Cardiovascular Division, Department of Medicine, Heart and Lung Research Institute (HLRI), University of Cambridge, Cambridge, United Kingdom
- British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
46
|
Chayé MAM, van Hengel ORJ, Voskamp AL, Ozir-Fazalalikhan A, König MH, Stam KA, Manurung MD, Mouwenda YD, Aryeetey YA, Kurniawan A, Kruize YCM, Sartono E, Buisman AM, Yazdanbakhsh M, Tak T, Smits HH. Multi-dimensional analysis of B cells reveals the expansion of memory and regulatory B-cell clusters in humans living in rural tropical areas. Clin Exp Immunol 2025; 219:uxae074. [PMID: 39129562 PMCID: PMC11771192 DOI: 10.1093/cei/uxae074] [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: 10/09/2023] [Revised: 07/06/2024] [Accepted: 08/09/2024] [Indexed: 08/13/2024] Open
Abstract
B-cells play a critical role in the formation of immune responses against pathogens by acting as antigen-presenting cells, by modulating immune responses, and by generating immune memory and antibody responses. Here, we studied B-cell subset distributions between regions with higher and lower microbial exposure, i.e. by comparing peripheral blood B-cells from people living in Indonesia or Ghana to those from healthy Dutch residents using a 36-marker mass cytometry panel. By applying an unbiased multidimensional approach, we observed differences in the balance between the naïve and memory compartments, with higher CD11c+ and double negative (DN-IgDnegCD27neg) memory (M)B-cells in individuals from rural tropical areas, and conversely lower naïve B-cells compared to residents from an area with less pathogen exposure. Furthermore, characterization of total B-cell populations, CD11c+, DN, and Breg cells showed the emergence of specific memory clusters in individuals living in rural tropical areas. Some of these differences were more pronounced in children compared to adults and suggest that a higher microbial exposure accelerates memory B-cell formation, which "normalizes" with age.
Collapse
Affiliation(s)
- Mathilde A M Chayé
- Leiden University Center for Infectious Diseases (LUCID), LUMC, Leiden, The Netherlands
| | - Oscar R J van Hengel
- Leiden University Center for Infectious Diseases (LUCID), LUMC, Leiden, The Netherlands
| | - Astrid L Voskamp
- Leiden University Center for Infectious Diseases (LUCID), LUMC, Leiden, The Netherlands
| | | | - Marion H König
- Leiden University Center for Infectious Diseases (LUCID), LUMC, Leiden, The Netherlands
| | - Koen A Stam
- Leiden University Center for Infectious Diseases (LUCID), LUMC, Leiden, The Netherlands
| | - Mikhael D Manurung
- Leiden University Center for Infectious Diseases (LUCID), LUMC, Leiden, The Netherlands
| | - Yoanne D Mouwenda
- Leiden University Center for Infectious Diseases (LUCID), LUMC, Leiden, The Netherlands
- Centre de Recherches Médicales de Lambaréné (CERMEL), Lambaréné, Gabon
| | - Yvonne A Aryeetey
- Parasitology Department, Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana
| | - Agnes Kurniawan
- Department of Parasitology, Universitas Indonesia, Jakarta, Indonesia
| | - Yvonne C M Kruize
- Leiden University Center for Infectious Diseases (LUCID), LUMC, Leiden, The Netherlands
| | - Erliyani Sartono
- Leiden University Center for Infectious Diseases (LUCID), LUMC, Leiden, The Netherlands
| | - Anne-Marie Buisman
- Laboratory for Immunology of Infectious Diseases and Vaccines, Center for Infectious Diseases Control, National Institute for Public Health and The Environment, Bilthoven, The Netherlands
| | - Maria Yazdanbakhsh
- Leiden University Center for Infectious Diseases (LUCID), LUMC, Leiden, The Netherlands
| | - Tamar Tak
- Leiden University Center for Infectious Diseases (LUCID), LUMC, Leiden, The Netherlands
| | - Hermelijn H Smits
- Leiden University Center for Infectious Diseases (LUCID), LUMC, Leiden, The Netherlands
| |
Collapse
|
47
|
Asashima H, Akao S, Matsumoto I. Emerging roles of checkpoint molecules on B cells. Immunol Med 2025:1-12. [PMID: 39819449 DOI: 10.1080/25785826.2025.2454045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 01/07/2025] [Indexed: 01/19/2025] Open
Abstract
Immune checkpoint molecules, including both co-inhibitory molecules and co-stimulatory molecules, are known to play critical roles in regulating T-cell responses. During the last decades, immunotherapies targeting these molecules (such as programmed cell death 1 (PD-1), and lymphocyte activation gene 3 (LAG-3)) have provided clinical benefits in many cancers. It is becoming apparent that not only T cells, but also B cells have a capacity to express some checkpoint molecules. These were originally thought to be only the markers for regulatory B cells which produce IL-10, but recent studies suggest that these molecules (especially T-cell immunoglobulin and mucin domain 1 (TIM-1), T cell immunoreceptor with Ig and ITIM domains (TIGIT), and PD-1) can regulate intrinsic B-cell activation and functions. Here, we focus on these molecules and summarize their characteristics, ligands, and functions on B cells.
Collapse
Affiliation(s)
- Hiromitsu Asashima
- Department of Rheumatology, Institute of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Satoshi Akao
- Department of Rheumatology, Institute of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Isao Matsumoto
- Department of Rheumatology, Institute of Medicine, University of Tsukuba, Ibaraki, Japan
| |
Collapse
|
48
|
He M, Feng Y. Elusive modes of Foxp3 activity in versatile regulatory T cells. Front Immunol 2025; 15:1533823. [PMID: 39882241 PMCID: PMC11774722 DOI: 10.3389/fimmu.2024.1533823] [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: 11/25/2024] [Accepted: 12/23/2024] [Indexed: 01/31/2025] Open
Abstract
Foxp3-expressing CD4 regulatory T (Treg) cells play a crucial role in suppressing autoimmunity, tolerating food antigens and commensal microbiota, and maintaining tissue integrity. These multifaceted functions are guided by environmental cues through interconnected signaling pathways. Traditionally, Treg fate and function were believed to be statically determined by the forkhead box protein Foxp3 that directly binds to DNA. However, this model has not been rigorously tested in physiological and pathological conditions where Treg cells adapt their function in response to environmental cues, raising questions about the contribution of Foxp3-dependent gene regulation to their versatility. Recent research indicates that Foxp3 primarily functions as a transcriptional cofactor, whose chromatin interaction is influenced by other DNA-binding proteins that respond to cell activation, stimulation, or differentiation. This new perspective offers an opportunity to reevaluate Foxp3's activity modes in diverse biological contexts. By exploring this paradigm, we aim to unravel the fundamental principles of Treg cell biology.
Collapse
Affiliation(s)
| | - Yongqiang Feng
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, TN, United States
| |
Collapse
|
49
|
Cai H, Mu Q, Xiong H, Liu M, Yang F, Zhou L, Zhou B. Regulatory B cells in parasitic infections: roles and therapeutic potential. Parasitol Res 2025; 124:5. [PMID: 39809978 PMCID: PMC11732949 DOI: 10.1007/s00436-024-08450-4] [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: 05/26/2024] [Accepted: 12/28/2024] [Indexed: 01/16/2025]
Abstract
Parasitic infection is a complex process involving interactions among various immune cells. Regulatory B cells (Breg cells), a subset of B lymphocytes with immunosuppressive functions, play a role in modulating immune responses during infection to prevent excessive immune activation. This article reviews the origin, phenotype, and immunoregulatory mechanisms of Breg cells. We summarize the immunomodulatory roles of Breg cells in various parasitic infections. We also discuss the potential applications of activating Breg cells through parasitic infections and their derived molecules in the treatment of certain allergic, autoimmune, and inflammatory diseases. The aim is to provide new perspectives for the future treatment of parasitic diseases and other related conditions.
Collapse
Affiliation(s)
- Haojun Cai
- School of Basic Medicine, Zunyi Medical University, Zunyi, China
| | - Qianqian Mu
- School of Basic Medicine, Zunyi Medical University, Zunyi, China
| | - Haiting Xiong
- School of Basic Medicine, Zunyi Medical University, Zunyi, China
| | - Meichen Liu
- School of Basic Medicine, Zunyi Medical University, Zunyi, China
| | - Fengjiao Yang
- School of Basic Medicine, Zunyi Medical University, Zunyi, China
| | - Ling Zhou
- Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Biying Zhou
- School of Basic Medicine, Zunyi Medical University, Zunyi, China.
| |
Collapse
|
50
|
Bai Y, Ding J, He L, Zhu Z, Pan J, Qi C. β-Glucan induced plasma B cells differentiation to enhance antitumor immune responses by Dectin-1. BMC Immunol 2025; 26:2. [PMID: 39794756 PMCID: PMC11724571 DOI: 10.1186/s12865-025-00681-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Accepted: 01/06/2025] [Indexed: 01/13/2025] Open
Abstract
BACKGROUND B lymphocytes, essential in cellular immunity as antigen-presenting cells and in humoral immunity as major effector cells, play a crucial role in the antitumor response. Our previous work has shown β-glucan enhanced immunoglobulins (Ig) secretion. But the specific mechanisms of B-cell activation with β-glucan are poorly understood. Here, we took advantage of β-glucan to improve the antitumor immune response of B cells. RESULTS In vitro experiments demonstrate that β-glucan enhance the differentiation of B220lo CD138+ B cells, up-regulate co-stimulatory molecules, and increase the production of cytokines and Ig in response to various antigens. Using the Dectin-1 knockout mice, we revealed that β-glucan modulate B cell immune responses dependent on Dectin-1 receptor. In mouse models of Lewis lung cancer (LLC) tumors, combining β-glucan with programmed death-1(PD-1) blocking antibodies led to increase recruitment of CD19+ B cells in the tumor microenvironment (TME), higher numbers of germinal centers B cells (GC B) in the spleen and draining lymph node (DLN), elevate Ig production, and delay tumor progression. CONCLUSIONS These findings reveal that β-glucan can serve as a potent adjuvant to modulate B cell immune responses in a Dectin-1 dependent manner and improve immune checkpoint blockade (ICB) therapy in antitumor. CLINICAL TRIAL NUMBER Not applicable.
Collapse
Affiliation(s)
- Yu Bai
- Laboratory of Oncology, Medical Research Center, The Second People's Hospital of Changzhou, The Third Affiliated Hospital of Nanjing Medical University, Changzhou, China
| | - Jun Ding
- Laboratory of Oncology, Medical Research Center, The Second People's Hospital of Changzhou, The Third Affiliated Hospital of Nanjing Medical University, Changzhou, China
| | - Liuyang He
- Laboratory of Oncology, Medical Research Center, The Second People's Hospital of Changzhou, The Third Affiliated Hospital of Nanjing Medical University, Changzhou, China
| | - Zhichao Zhu
- Laboratory of Oncology, Medical Research Center, The Second People's Hospital of Changzhou, The Third Affiliated Hospital of Nanjing Medical University, Changzhou, China
| | - Jie Pan
- Laboratory of Oncology, Medical Research Center, The Second People's Hospital of Changzhou, The Third Affiliated Hospital of Nanjing Medical University, Changzhou, China
| | - Chunjian Qi
- Laboratory of Oncology, Medical Research Center, The Second People's Hospital of Changzhou, The Third Affiliated Hospital of Nanjing Medical University, Changzhou, China.
| |
Collapse
|