51
|
Feng R, Li X, Li B, Luan T, He J, Liu G, Yue J. Integrating transcriptomics and scPagwas analysis predicts naïve CD4 T cell-related gene DRAM2 as a potential biomarker and therapeutic target for colorectal cancer. BMC Cancer 2025; 25:317. [PMID: 39984869 PMCID: PMC11843817 DOI: 10.1186/s12885-025-13731-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: 10/09/2024] [Accepted: 02/13/2025] [Indexed: 02/23/2025] Open
Abstract
OBJECTIVE The interaction between T cells, particularly naïve CD4 T cells (CD4Tn), and colorectal cancer (CRC) is highly complex. CD4Tn play a crucial role in modulating immune responses within the tumor microenvironment, yet the precise mechanisms by which they influence tumor progression remain elusive. This study aims to explore the relationship between CRC and CD4Tn, identify biomarkers and therapeutic targets, and focus on the role of CD4Tn in shaping the immune environment of CRC. METHODS Single-cell transcriptomics, alongside the scPagwas algorithm, were employed to identify pivotal T cell subsets involved in CRC progression. Bulk transcriptomic data were further analyzed using deconvolution algorithms to elucidate the roles of these key T cell subsets. The abundance of naïve CD4 T cells (CD4Tn) was specifically assessed to gauge patient responses to immunotherapy, alterations in the immune microenvironment, and correlations with genetic mutations. Key genes linked to CD4Tn were identified using weighted gene co-expression network analysis and Pearson correlation scores. The SMR algorithm was subsequently used for validation, with experimental verification following. RESULTS Through single-cell transcriptomics and the scPagwas algorithm, CD4Tn was confirmed as a critical cell type in CRC progression. High infiltration of CD4Tn cells in CRC patients was correlated with poorer prognosis and suboptimal responses to immunotherapy. SMR analysis suggested a potential causal link between DRAM2 gene expression and CRC progression. Experimental knockdown of DRAM2 in colorectal cancer cells significantly inhibited tumor growth. CONCLUSION The DRAM2 gene, associated with CD4Tn cells, appears to play a pivotal role in the advancement of CRC and may represent a promising therapeutic target for treatment.
Collapse
Affiliation(s)
- Rui Feng
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaofang Li
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Benhua Li
- The Second People's Hospital of Liangshan Yi Autonomous Prefecture, Xichang, China
| | - Tiankuo Luan
- Department of Human Anatomy, Basic Medical School, Chongqing Medical University, Chongqing, China
| | - Jiaming He
- Department of Human Anatomy, Basic Medical School, Chongqing Medical University, Chongqing, China
| | - Guojing Liu
- Department of Neurosurgery, The University-Town Hospital of Chongqing Medical University, NO.55 of university-town middle Road, Shapingba District, Chongqing, 400000, China.
| | - Jian Yue
- Department of Breast Surgery, Gaozhou People's Hospital, No.89 Xiguan Road, Gaozhou, Guangdong, 525200, China.
| |
Collapse
|
52
|
Venegoni C, Raineri D, Mazzucca CB, Ghazanfar A, Cappellano G, Baricich A, Patrucco F, Zeppegno P, Gramaglia C, Balbo PE, Cantaluppi V, Patti G, Giordano M, Manfredi M, Rolla R, Sainaghi PP, Pirisi M, Bellan M, Chiocchetti A. Post-COVID-19 sequelae are associated with sustained SARS-CoV-2-specific CD4 + immune responses. Int Immunopharmacol 2025; 148:114103. [PMID: 39874845 DOI: 10.1016/j.intimp.2025.114103] [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: 10/29/2024] [Revised: 01/03/2025] [Accepted: 01/14/2025] [Indexed: 01/30/2025]
Abstract
BACKGROUND Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has led to widespread post-acute sequelae of COVID-19 (PASC), affecting multiple body systems. Despite its prevalence, PASC's pathogenesis remains unclear, with hypotheses suggesting viral persistence, immune activation, and autoimmune responses among the pathogenetic mechanism. This study aimed to evaluate T cell memory response in PASC patients, one year post-hospital discharge and correlate it with clinical parameters to identify a potential PASC-associated fingerprint. METHODS Peripheral blood mononuclear cells (PBMCs) from PASC patients and healthy controls (HC) were stimulated with a pool of spike peptides. CD4+ and CD8+ T cell responses were evaluated by flow cytometry using the activation-induced markers assay (AIM). RESULTS Findings showed significant activation of the CD4+ T cell compartment, with a higher proportion of responders among PASC patients. Central memory (CM) T cells expressing pro-inflammatory cytokines were more prevalent in responders. Clinical correlations revealed higher SARS-CoV-2-specific T cell responses in patients with reduced diffuse lung capacity for carbon monoxide (DLCO) and residual symptoms. CONCLUSION These immune changes, especially in CM T cells, could play a pivotal role in PASC's development and persistence, impacting patients' daily lives.
Collapse
Affiliation(s)
- Chiara Venegoni
- Department of Health Sciences, Interdisciplinary Research Center of Autoimmune Diseases-IRCAD, University of Eastern Piedmont 28100 Novara, Italy; Center for Translational Research on Autoimmune and Allergic Diseases, University of Eastern Piedmont 28100 Novara, Italy.
| | - Davide Raineri
- Department of Health Sciences, Interdisciplinary Research Center of Autoimmune Diseases-IRCAD, University of Eastern Piedmont 28100 Novara, Italy; Center for Translational Research on Autoimmune and Allergic Diseases, University of Eastern Piedmont 28100 Novara, Italy.
| | - Camilla Barbero Mazzucca
- Department of Health Sciences, Interdisciplinary Research Center of Autoimmune Diseases-IRCAD, University of Eastern Piedmont 28100 Novara, Italy; Center for Translational Research on Autoimmune and Allergic Diseases, University of Eastern Piedmont 28100 Novara, Italy.
| | - Ali Ghazanfar
- Department of Health Sciences, Interdisciplinary Research Center of Autoimmune Diseases-IRCAD, University of Eastern Piedmont 28100 Novara, Italy; Center for Translational Research on Autoimmune and Allergic Diseases, University of Eastern Piedmont 28100 Novara, Italy.
| | - Giuseppe Cappellano
- Department of Health Sciences, Interdisciplinary Research Center of Autoimmune Diseases-IRCAD, University of Eastern Piedmont 28100 Novara, Italy; Center for Translational Research on Autoimmune and Allergic Diseases, University of Eastern Piedmont 28100 Novara, Italy.
| | - Alessio Baricich
- Translational Medicine Department, University of Eastern Piedmont, Novara, Italy.
| | - Filippo Patrucco
- Translational Medicine Department, University of Eastern Piedmont, Novara, Italy; AOU University Hospital "Maggiore della Carità" 28100 Novara, Italy.
| | - Patrizia Zeppegno
- Translational Medicine Department, University of Eastern Piedmont, Novara, Italy; AOU University Hospital "Maggiore della Carità" 28100 Novara, Italy.
| | - Carla Gramaglia
- Translational Medicine Department, University of Eastern Piedmont, Novara, Italy; AOU University Hospital "Maggiore della Carità" 28100 Novara, Italy.
| | | | - Vincenzo Cantaluppi
- Translational Medicine Department, University of Eastern Piedmont, Novara, Italy; AOU University Hospital "Maggiore della Carità" 28100 Novara, Italy.
| | - Giuseppe Patti
- Translational Medicine Department, University of Eastern Piedmont, Novara, Italy; AOU University Hospital "Maggiore della Carità" 28100 Novara, Italy.
| | - Mara Giordano
- Department of Health Sciences, Interdisciplinary Research Center of Autoimmune Diseases-IRCAD, University of Eastern Piedmont 28100 Novara, Italy; AOU University Hospital "Maggiore della Carità" 28100 Novara, Italy.
| | - Marcello Manfredi
- Center for Translational Research on Autoimmune and Allergic Diseases, University of Eastern Piedmont 28100 Novara, Italy; Translational Medicine Department, University of Eastern Piedmont, Novara, Italy.
| | - Roberta Rolla
- Department of Health Sciences, Interdisciplinary Research Center of Autoimmune Diseases-IRCAD, University of Eastern Piedmont 28100 Novara, Italy; Translational Medicine Department, University of Eastern Piedmont, Novara, Italy.
| | - Pier Paolo Sainaghi
- Center for Translational Research on Autoimmune and Allergic Diseases, University of Eastern Piedmont 28100 Novara, Italy; Translational Medicine Department, University of Eastern Piedmont, Novara, Italy; AOU University Hospital "Maggiore della Carità" 28100 Novara, Italy.
| | - Mario Pirisi
- Center for Translational Research on Autoimmune and Allergic Diseases, University of Eastern Piedmont 28100 Novara, Italy; Translational Medicine Department, University of Eastern Piedmont, Novara, Italy.
| | - Mattia Bellan
- Center for Translational Research on Autoimmune and Allergic Diseases, University of Eastern Piedmont 28100 Novara, Italy; Translational Medicine Department, University of Eastern Piedmont, Novara, Italy; AOU University Hospital "Maggiore della Carità" 28100 Novara, Italy.
| | - Annalisa Chiocchetti
- Department of Health Sciences, Interdisciplinary Research Center of Autoimmune Diseases-IRCAD, University of Eastern Piedmont 28100 Novara, Italy; Center for Translational Research on Autoimmune and Allergic Diseases, University of Eastern Piedmont 28100 Novara, Italy.
| |
Collapse
|
53
|
Zemelka-Wiacek M. The Interaction Among Effector, Regulatory, and Tγδ Cells Determines the Development of Allergy or Tolerance to Chromium. J Clin Med 2025; 14:1370. [PMID: 40004900 PMCID: PMC11856200 DOI: 10.3390/jcm14041370] [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: 01/01/2025] [Revised: 02/05/2025] [Accepted: 02/11/2025] [Indexed: 02/27/2025] Open
Abstract
Background/Objectives: Chromium, a common environmental and occupational sensitizer, frequently induces allergic contact dermatitis (ACD). This study investigates the role of CD4+ (T helper), CD8+ (T cytotoxic), regulatory (Tregs: CD4+CD25+ and CD8+CD25+), and gamma delta (Tγδ) T cells in chromium tolerance versus hypersensitivity. Methods: Six chromium-allergic patients and six healthy controls were recruited, confirmed via patch testing. Peripheral blood mononuclear cells (PBMCs) were isolated and cultured, with chromium exposure and proliferation assays conducted. Specific T cell subtypes were isolated and analyzed for chromium-specific proliferative responses, cytokine production, and metabolic activity. Results: Chromium-allergic individuals exhibited broad proliferation across PBMC and T cell subsets, contrasting with restricted responses in controls. Treg cells in healthy subjects effectively suppressed T cell proliferation in response to chromium, while allergic individuals showed unmodulated T cell activity, indicative of impaired regulatory function. Cytokine analysis revealed elevated IL-2 and TNF-α but absent IL-10 in allergic patients. Metabolic assessments showed higher glycolytic activity in Tregs of healthy controls, suggesting enhanced regulatory potential. Conclusions: These findings highlight the importance of balanced effector and regulatory T cell interactions for chromium tolerance. Dysregulated Treg and Tγδ cell functions in allergic individuals may contribute to hypersensitivity, with implications for targeted therapeutic strategies to restore immune balance and reduce allergic responses in chromium-sensitive patients.
Collapse
Affiliation(s)
- Magdalena Zemelka-Wiacek
- Department of Clinical Immunology, Faculty of Medicine, Wroclaw Medical University, 50-367 Wrocław, Poland
| |
Collapse
|
54
|
Shu A, Tian X, Yue J, Jiang Y, Liu Y. Unveiling the role of lncRNA ERDR1 in immune cell regulation. Heliyon 2025; 11:e42085. [PMID: 39991241 PMCID: PMC11847233 DOI: 10.1016/j.heliyon.2025.e42085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Revised: 01/11/2025] [Accepted: 01/16/2025] [Indexed: 02/25/2025] Open
Abstract
Long non-coding RNAs (lncRNAs) are a class of RNA molecules that exceed 200 nucleotides in length and lack the capacity to encode proteins. In recent years, there has been a surge of interest in lncRNA research, leading to the discovery of their diverse structures and functions. This review focused on elucidating the regulatory roles of lncRNA erythroid differentiation regulatory 1 (Erdr1) within immune cells and its involvement in related disorders. By synthesizing findings from recent studies sourced from PubMed, this paper examined the biological functions and underlying mechanisms by which lncRNA Erdr1 influences immune cells and contributes to various diseases. Emerging research highlights that lncRNA Erdr1 exerts significant effects on the functionality of immune cells, particularly T lymphocytes (T cells), natural killer (NK) cells, and macrophages. Furthermore, Erdr1 has been implicated in the mitigation of several diseases, including acne, wound healing, osteoarthritis, melanoma, gastric cancer, obesity, and autism. Given its complex biological functions and mechanisms, Erdr1 presents itself as a promising biomarker and a potential therapeutic target for a range of immune cell-related disorders.
Collapse
Affiliation(s)
- Aihua Shu
- Department of Anesthesiology, The First College of Clinical Medical Science, China Three Gorges University, Yichang, Hubei Province, 443000, China
- Yichang Central People's Hospital, Yichang, Hubei Province, 443000, China
- The Institute of Geriatric Anesthesia, China Three Gorges University, Yichang, Hubei Province, 443000, China
| | - Xu Tian
- Department of Anesthesiology, The First College of Clinical Medical Science, China Three Gorges University, Yichang, Hubei Province, 443000, China
- Yichang Central People's Hospital, Yichang, Hubei Province, 443000, China
- The Institute of Geriatric Anesthesia, China Three Gorges University, Yichang, Hubei Province, 443000, China
| | - Jie Yue
- Department of Anesthesiology, The First College of Clinical Medical Science, China Three Gorges University, Yichang, Hubei Province, 443000, China
- Yichang Central People's Hospital, Yichang, Hubei Province, 443000, China
- The Institute of Geriatric Anesthesia, China Three Gorges University, Yichang, Hubei Province, 443000, China
| | - Yuxia Jiang
- Department of Anesthesiology, The First College of Clinical Medical Science, China Three Gorges University, Yichang, Hubei Province, 443000, China
- Yichang Central People's Hospital, Yichang, Hubei Province, 443000, China
- The Institute of Geriatric Anesthesia, China Three Gorges University, Yichang, Hubei Province, 443000, China
| | - Yifei Liu
- Department of Anesthesiology, The First College of Clinical Medical Science, China Three Gorges University, Yichang, Hubei Province, 443000, China
- Yichang Central People's Hospital, Yichang, Hubei Province, 443000, China
- The Institute of Geriatric Anesthesia, China Three Gorges University, Yichang, Hubei Province, 443000, China
| |
Collapse
|
55
|
Wang G, Li J, Sun S, Yang Y, Han Z, Pei Z, Cheng L. An electrically activable nanochip to intensify gas-ionic-immunotherapy. Sci Bull (Beijing) 2025; 70:390-406. [PMID: 39667986 DOI: 10.1016/j.scib.2024.11.035] [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: 07/23/2024] [Revised: 10/07/2024] [Accepted: 11/15/2024] [Indexed: 12/14/2024]
Abstract
Excess intracellular H2S induces destructive mitochondrial toxicity, while overload of Zn2+ results in cell pyroptosis and potentiates the tumor immunogenicity for immunotherapy. However, the precise delivery of both therapeutics remains a great challenge. Herein, an electrically activable ZnS nanochip for the controlled release of H2S and Zn2+ was developed for enhanced gas-ionic-immunotherapy (GIIT). Under an electric field, a locality with particularly high concentrations of H2S and Zn2+ was established by the voltage-controlled degradation of the ZnS nanoparticles (NPs). Consequently, the ZnS nanochip-mediated gas-ionic therapy (GIT) resulted in mitochondrial membrane potential depolarization, energy generation inhibition, and oxidative stress imbalance in tumor cells. Interestingly, the cyclic guanosine monophosphate-adenosine monophosphate synthase-stimulator of interferon genes (cGAS-STING) signaling pathway was activated due to the mitochondrial destruction. Moreover, the released Zn2+ resulted in the increase of the intracellular Zn levels and cell pyroptosis, which enhanced the immunogenicity via the release of damage-associated molecular patterns (DAMPs). In vitro and in vivo studies revealed that the ZnS nanochip-based GIT effectively eliminated the tumors under an electric field and mobilized the cytotoxic T lymphocytes for immunotherapy. The combination with αCTLA-4 further promoted the adaptive immune response and inhibited tumor metastasis and long-term tumor recurrence. This work presented an electrically activable ZnS nanochip for combined immunotherapy, which might inspire the development of electric stimulation therapy.
Collapse
Affiliation(s)
- Gang Wang
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou 215123, China
| | - Jingrui Li
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou 215123, China
| | - Shumin Sun
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou 215123, China
| | - Yuqi Yang
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou 215123, China
| | - Zhihui Han
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou 215123, China
| | - Zifan Pei
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou 215123, China
| | - Liang Cheng
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou 215123, China.
| |
Collapse
|
56
|
Fleischer AB, Amann B, von Toerne C, Degroote RL, Schmalen A, Weißer T, Hauck SM, Deeg CA. Differential Expression of ARG1 and MRC2 in Retinal Müller Glial Cells During Autoimmune Uveitis. Biomolecules 2025; 15:288. [PMID: 40001591 PMCID: PMC11853277 DOI: 10.3390/biom15020288] [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: 12/16/2024] [Revised: 02/11/2025] [Accepted: 02/13/2025] [Indexed: 02/27/2025] Open
Abstract
Retinal Müller glial cells (RMG) play a crucial role in retinal neuroinflammation, including autoimmune uveitis. Increasing evidence supports their function as active modulators of immune responses and potential atypical antigen-presenting cells (APCs). To further investigate this hypothesis, we conducted a differential proteome analysis of primary equine RMG from healthy controls and horses with equine recurrent uveitis (ERU), a spontaneous model of autoimmune uveitis. This analysis identified 310 proteins with differential abundance. Among these, the Major Histocompatibility Complex (MHC) class II and the enzyme Arginase 1 (ARG1) were significantly enriched in RMG from uveitis-affected horses, whereas Mannose Receptor C-type 2 (MRC2) and its interactor Thrombospondin 1 (THBS1) were more abundant in healthy RMG. The detection of MHC class II in equine RMG, consistent with previous studies, validates the robustness of our approach. Furthermore, the identification of ARG1 and MRC2, together with THBS1, provides new insights into the immunomodulatory and antigen-presenting properties of RMG. Immunohistochemical analyses confirmed the proteomic findings and revealed the spatial distribution of ARG1 and MRC2. ARG1 and MRC2 are thus markers for RMG in the neuroinflammatory or physiological milieu and highlight potential differences in the immune function of RMG, particularly in antigen presentation.
Collapse
Affiliation(s)
- Amelie B. Fleischer
- Chair of Physiology, Department of Veterinary Sciences, LMU Munich, D-82152 Martinsried, Germany (T.W.)
| | - Barbara Amann
- Chair of Physiology, Department of Veterinary Sciences, LMU Munich, D-82152 Martinsried, Germany (T.W.)
| | - Christine von Toerne
- Metabolomics and Proteomics Core, Helmholtz Center Munich, German Research Center for Environmental Health, D-80939 Munich, Germany
| | - Roxane L. Degroote
- Chair of Physiology, Department of Veterinary Sciences, LMU Munich, D-82152 Martinsried, Germany (T.W.)
| | - Adrian Schmalen
- Chair of Physiology, Department of Veterinary Sciences, LMU Munich, D-82152 Martinsried, Germany (T.W.)
| | - Tanja Weißer
- Chair of Physiology, Department of Veterinary Sciences, LMU Munich, D-82152 Martinsried, Germany (T.W.)
| | - Stefanie M. Hauck
- Metabolomics and Proteomics Core, Helmholtz Center Munich, German Research Center for Environmental Health, D-80939 Munich, Germany
| | - Cornelia A. Deeg
- Chair of Physiology, Department of Veterinary Sciences, LMU Munich, D-82152 Martinsried, Germany (T.W.)
| |
Collapse
|
57
|
Ono R, Maeda K, Tanioka T, Isozaki T. Monocyte-derived Langerhans cells express Delta-like 4 induced by peptidoglycan and interleukin-4 mediated suppression. Front Immunol 2025; 16:1532620. [PMID: 40018044 PMCID: PMC11865044 DOI: 10.3389/fimmu.2025.1532620] [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: 11/22/2024] [Accepted: 01/30/2025] [Indexed: 03/01/2025] Open
Abstract
T cells contribute to immunotherapy and autoimmune pathogenesis and Langerhans cells (LCs) have a substantial ability to activate T cells. In vitro-generated monocyte-derived LCs (Mo-LCs) are useful models to study LC function in autoimmune diseases and to test future LC-based immunotherapies. Although dendritic cells (DCs) expressing high levels of Delta-like 4 (DLL4+ DCs), which is a member of the Notch ligand family, have greater ability than DLL4- DCs to activate T cells, the induction method of human DLL4+ DCs has yet to be determined. The aim of this study is to establish whether Mo-LCs express DLL4 and establish the induction method of antigen presenting cells, which most potently activate T cells, similar to our previously established induction method of human Mo-LCs. We compared the ratios of DLL4 expression and T cell activation via flow cytometry among monocyte-derived cells, which have a greater ability than the resident cells to activate T cells. Here, we discovered that Mo-LCs expressed DLL4, which most potently activated T cells among monocyte-derived cells, and that Mo-LCs and DLL4 expression were induced by DLL4, granulocyte macrophage colony-stimulating factor, and transforming growth factor-β1. Additionally, peptidoglycan was required for DLL4 expression, whereas interleukin-4 repressed it. These findings provide insights into the roles of DLL4-expressing cells such as DLL4+ Mo-LCs in human diseases, which will assist with the development of more effective therapeutic strategies in the future.
Collapse
Affiliation(s)
- Rei Ono
- Department of Pathogenesis and Translational Medicine, Showa University Graduate School of Pharmacy, Tokyo, Japan
| | - Kohei Maeda
- Department of Pathogenesis and Translational Medicine, Showa University Graduate School of Pharmacy, Tokyo, Japan
| | - Toshihiro Tanioka
- Department of Pathogenesis and Translational Medicine, Showa University Graduate School of Pharmacy, Tokyo, Japan
| | - Takeo Isozaki
- Department of Pathogenesis and Translational Medicine, Showa University Graduate School of Pharmacy, Tokyo, Japan
- Department of Rheumatology, Showa University Graduate School of Medicine, Tokyo, Japan
| |
Collapse
|
58
|
Rahman MS, Hadrick K, Chung SJ, Carley I, Yoo JY, Nahar S, Kim TH, Kim T, Jeong JW. Nanoceria as a non-steroidal anti-inflammatory drug for endometriosis theranostics. J Control Release 2025; 378:1015-1029. [PMID: 39742921 PMCID: PMC11830557 DOI: 10.1016/j.jconrel.2024.12.074] [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: 07/31/2024] [Revised: 12/02/2024] [Accepted: 12/28/2024] [Indexed: 01/04/2025]
Abstract
Endometriosis, the growth of endometrial-like tissue outside the uterus, causes chronic pain and infertility in 10 % of reproductive-aged women worldwide. Unfortunately, no permanent cure exists, and current medical and surgical treatments offer only temporary relief. Endometriosis is a chronic inflammatory disease characterized by immune system dysfunction. Our previous study showed aberrant activation of signal transducer and activator of transcription 3 (STAT3) in endometriosis. Our transcriptomic analysis of uterine tissue from uterine-specific Stat3 knock-out mice identifies that STAT3 regulates inflammatory and immune-related genes. Here, we evaluate cerium-oxide nanoparticles (nanoceria) as a non-steroidal anti-inflammatory drug for endometriosis theranostics. Our in vitro studies validate the multi-enzymatic properties of nanoceria, enabling the transition of pro-inflammatory macrophages to an anti-inflammatory state in J774 macrophage cells. In vivo, treatment of endometriosis mice with nanoceria reveals its ability to passively accumulate at ectopic lesions. The nanoceria conjugated with indocyanine green are non-invasively trackable to ectopic lesions. Therefore, immune modulation and anti-inflammatory effects of nanoceria significantly reduce development of ectopic lesions while minimizing off-target effects, such as avoiding interference with pregnancy including implantation and decidualization. Our results suggest that aberrant STAT3 activation is a major contributor to endometriosis, and nanoceria offers a novel theranostic approach for endometriosis.
Collapse
Affiliation(s)
- Md Saidur Rahman
- Department of Obstetrics, Gynecology and Women's Health, University of Missouri School of Medicine, 1030 Hitt Street, Columbia, MO 65211, USA
| | - Kay Hadrick
- Department of Biomedical Engineering, Michigan State University, 775 Woodlot Dr, East Lansing, MI 48824, USA; Institute for Quantitative Health Science and Engineering, Michigan State University, 775 Woodlot Dr, East Lansing, MI 48824, USA
| | - Seock-Jin Chung
- Department of Biomedical Engineering, Michigan State University, 775 Woodlot Dr, East Lansing, MI 48824, USA; Institute for Quantitative Health Science and Engineering, Michigan State University, 775 Woodlot Dr, East Lansing, MI 48824, USA
| | - Ian Carley
- Institute for Quantitative Health Science and Engineering, Michigan State University, 775 Woodlot Dr, East Lansing, MI 48824, USA; Department of Biochemistry and Molecular Biology, Michigan State University, 603 Wilson Rd, East Lansing, MI 48823, USA
| | - Jung-Yoon Yoo
- Department of Biomedical Laboratory Science, Yonsei University Mirae Campus, Wonju, 26493, Republic of Korea
| | - Shamsun Nahar
- Department of Obstetrics, Gynecology and Women's Health, University of Missouri School of Medicine, 1030 Hitt Street, Columbia, MO 65211, USA
| | - Tae Hoon Kim
- Department of Obstetrics, Gynecology and Women's Health, University of Missouri School of Medicine, 1030 Hitt Street, Columbia, MO 65211, USA
| | - Taeho Kim
- Department of Biomedical Engineering, Michigan State University, 775 Woodlot Dr, East Lansing, MI 48824, USA; Institute for Quantitative Health Science and Engineering, Michigan State University, 775 Woodlot Dr, East Lansing, MI 48824, USA.
| | - Jae-Wook Jeong
- Department of Obstetrics, Gynecology and Women's Health, University of Missouri School of Medicine, 1030 Hitt Street, Columbia, MO 65211, USA.
| |
Collapse
|
59
|
Lin Y, Lin P, Xu R, Chen X, Lu Y, Zheng J, Zheng Y, Zhou Z, Mai Z, Zhao X, Cui L. Nanovaccines empowering CD8 + T cells: a precision strategy to enhance cancer immunotherapy. Theranostics 2025; 15:3098-3121. [PMID: 40083941 PMCID: PMC11898294 DOI: 10.7150/thno.107856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Accepted: 01/20/2025] [Indexed: 03/16/2025] Open
Abstract
Cancer immunotherapy leveraging nanovaccines represents a cutting-edge frontier in precision medicine, specifically designed to potentiate CD8+ T cell-based immunotherapy. This review thoroughly delineates the evolving landscape of cancer nanovaccine development, emphasizing their advantageous role in modulating the immunosuppressive tumor microenvironment (TME) to enhance CD8+ T cell efficacy. We critically analyze current innovations in nanovaccine design, focusing on their capacity to deliver tumor antigens and immunostimulatory adjuvants effectively. These nanovaccines are engineered to overcome the physical and immunological barriers of the TME, facilitating the robust activation and proliferation of CD8+ T cells. Challenges such as delivery efficacy, safety, and scalable manufacturing are discussed, alongside future prospects which include the potential of developing specific biomaterial approaches to provide durable antitumor immunity. This comprehensive analysis not only underscores the transformative potential of cancer nanovaccines in enhancing CD8+ T cell responses but also highlights the critical need for advanced solutions to overcome the complex interplay of factors that limit the efficacy of current immunotherapies.
Collapse
Affiliation(s)
- Yunfan Lin
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, Guangdong, China
| | - Pei Lin
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, Guangdong, China
| | - Rongwei Xu
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, Guangdong, China
| | - Xu Chen
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, Guangdong, China
| | - Ye Lu
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, Guangdong, China
| | - Jiarong Zheng
- Department of Dentistry, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Yucheng Zheng
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, Guangdong, China
| | - Zihao Zhou
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, Guangdong, China
| | - Zizhao Mai
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, Guangdong, China
| | - Xinyuan Zhao
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, Guangdong, China
| | - Li Cui
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, Guangdong, China
- School of Dentistry, University of California, Los Angeles, Los Angeles, 90095, CA, USA
| |
Collapse
|
60
|
Gao C, Jian C, Wang L, Liu Y, Xiong Y, Wu T, Shi C. FAP-targeting biomimetic nanosystem to restore the activated cancer-associated fibroblasts to quiescent state for breast cancer radiotherapy. Int J Pharm 2025; 670:125190. [PMID: 39788396 DOI: 10.1016/j.ijpharm.2025.125190] [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] [Revised: 12/07/2024] [Accepted: 01/06/2025] [Indexed: 01/12/2025]
Abstract
Cancer associated fibroblasts (CAFs) are one of the most important stromal cells in the tumor microenvironment, playing a pivotal role in the development, recurrence, metastasis, and immunosuppression of cancer and treatment resistance. Here, we developed a core-shell biomimetic nanosystem termed as FAP-C NPs. This system was comprised of 4T1 extracellular vesicles fused with a FAP single-chain antibody fragment to form the biomimetic shell, and PLGA nanoparticles loaded with calcipotriol as the core. The FAP-modified shell endowed this nanosystem with active targeting ability to CAFs. Calcipotriol, a vitamin D analog, can activate the vitamin D receptor expressed on CAFs, promoting their transition from an activated to quiescent state. This process would help to reduce the pro-tumorigenic signals generated by CAFs, inhibit the stemness of cancer cells, and attenuate the inhibitory effect of CAFs on immune cells. The hydrated particle size of FAP-C NPs was approximately 206 nm, with a narrow distribution (polydispersity index < 0.2). The zeta potential of FAP-C NPs was -12.63 ± 0.61 mV. FAP-C NPs can restore CAFs to a quiescent state to shield the function of activated CAFs, inhibit tumor cell stemness, facilitate the maturation of dendritic cell, and relieve the inhibition of CAFs on lymphocytes. Besides, when combined with radiotherapy, this biomimetic nanosystem could inhibit the activation of CAFs, improve the sensitivity to radiation, and stimulate potent anti-tumor immune response with a 2-fold increase in the infiltration of cytotoxic T cells in tumor microenvironment, thereby effectively suppressing tumor growth with the tumor inhibitory rate as 78.3 %. Therefore, FAP-C NPs hold great potential for targeted breast cancer therapy.
Collapse
Affiliation(s)
- Chen Gao
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, Wuhan, 430022, China
| | - Chen Jian
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, Wuhan, 430022, China
| | - Lulu Wang
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, Wuhan, 430022, China
| | - Yajing Liu
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, Wuhan, 430022, China
| | - Yiquan Xiong
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Tingting Wu
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, Wuhan, 430022, China.
| | - Chen Shi
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, Wuhan, 430022, China.
| |
Collapse
|
61
|
Xiong S, Zhang S, Yue N, Cao J, Wu C. CAR-T cell therapy in the treatment of relapsed or refractory primary central nervous system lymphoma: recent advances and challenges. Leuk Lymphoma 2025:1-13. [PMID: 39898872 DOI: 10.1080/10428194.2025.2458214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Revised: 01/01/2025] [Accepted: 01/20/2025] [Indexed: 02/04/2025]
Abstract
Primary central nervous system lymphoma (PCNSL) is a rare and aggressive lymphoma that is isolated in the central nervous system (CNS) or vitreoretinal space. High-dose methotrexate (HD-MTX)-based immunochemotherapy is the frontline for its treatment, with a high early response rate. However, relapsed or refractory (R/R) patients present numerous difficulties and challenges in clinical treatment. Chimeric antigen receptor (CAR)-T cells offer a promising option for the treatment of hematologic malignancies, especially in the R/R B-cell lymphoma and multiple myeloma. Despite the exclusion of most PCNSL cases from pivotal CAR-T cell trials due to their specific tumor microenvironment (TME), available preclinical and clinical studies with small cohorts suggest an overall acceptable safety profile and remarkable anti-tumor effects. In this review, we will provide the development process of CAR-T cells and summarize the research progress, limitations, and future perspectives of CAR-T cell therapy in patients with R/R PCNSL.
Collapse
Affiliation(s)
- Shuzhen Xiong
- Department of Hematology, Lanzhou University Second Hospital, Lanzhou, China
| | | | | | | | | |
Collapse
|
62
|
Shi X, Han S, Wang G, Zhou G. Mitochondrial-associated programmed-cell-death patterns for predicting the prognosis of non-small-cell lung cancer. Front Med 2025; 19:101-120. [PMID: 39576480 DOI: 10.1007/s11684-024-1093-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 05/20/2024] [Indexed: 02/27/2025]
Abstract
Mitochondria are the convergence point of multiple pathways that trigger programmed cell death (PCD). Mitochondrial-associated PCD (mtPCD) is involved in the pathogenesis of several diseases. However, the role of mtPCD in the prognostic prediction of cancers including non-small-cell lung cancer (NSCLC) remains to be investigated. Here, 12 mtPCD patterns were analyzed in transcriptomics, genomics, and clinical data collected from 4 datasets containing 977 patients. A risk-score assessment system containing 18 genes was established. We found that NSCLC patients with a high-risk score had a poorer prognosis. A nomogram was constructed by incorporating the risk score with clinical features. The risk score was further associated with clinicopathological information, tumor-mutation frequency, and immunotherapy responses. NSCLC patients with a high risk score had more Treg cells infiltration. However, these patients had higher tumor-mutation burden scores and may be more sensitive to immunotherapy. Moreover, receptor-interacting serine/threonine protein kinase 2 (RIPK2) was selected from mtPCD gene model for validation. We found that RIPK2 exhibited oncogenic function, and its expression level was inversely associated with the overall survival of NSCLC. Taken together, our results indicated the accuracy and practicability of the mtPCD gene model and RIPK2 in predicting the prognosis of NSCLC.
Collapse
Affiliation(s)
- Xueyan Shi
- State Key Laboratory of Molecular Oncology & Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Sichong Han
- State Key Laboratory of Molecular Oncology & Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Guizhen Wang
- State Key Laboratory of Molecular Oncology & Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Guangbiao Zhou
- State Key Laboratory of Molecular Oncology & Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
63
|
Watanabe I, Suzuki N, Takara T. Supplementation with heat-sterilized Lactobacillus crispatus strain KT-11 stimulates the T cell-related immune function of healthy Japanese adults: A pilot randomized, placebo-controlled, double-blind, parallel-group study. Nutr Res 2025; 134:99-112. [PMID: 39889684 DOI: 10.1016/j.nutres.2024.12.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 12/23/2024] [Accepted: 12/23/2024] [Indexed: 02/03/2025]
Abstract
Viral infections are a global public health threat, reaffirming the importance of immune function. We previously identified Lactobacillus crispatus strain KT-11 (KT-11) and found that heat-sterilized KT-11 affect counts of T cell and dendritic cell in vitro, as well as promoted immunoglobulin A production and prevented weight loss caused by influenza virus infection in vivo. It was hypothesized that heat-sterilized KT-11 affects immune cell count/activity even in healthy individuals. We conducted a pilot study to examine the design to verify the effects of heat-sterilized KT-11 supplementation on immune cell count/activity and physical condition. This was a pilot randomized, double-blind, placebo-controlled, parallel-group study including 22 healthy adults who consumed either KT-11 or placebo for 4 weeks. Immunological status (immune cell count/activity and its score) and various immune-related indicators including SARS-CoV-2 immunoglobulin G antibody were assessed, and a physical condition questionnaire was administered. The primary outcome was immune cell count/activity (T cell subsets, B cell, natural killer cell) and overall immunological status score after 4 weeks. Two patients were excluded because of noncompliance; the final analysis included 20 participants (10 participants/group). The KT-11 group had a significantly higher CD3+ T cell count versus placebo group. The female subgroup also had a significantly higher CD8+CD28+ T cell count. Although the KT-11 group showed no changes in SARS-CoV-2 immunoglobulin G titer, it had fewer self-reported common cold-like symptoms, particularly fatigue. This pilot study showed that KT-11 affected immune cell profiles, suggesting that the feasibility of a verification study. This trial was registered at UMIN Clinical Trials Registry (UMIN000046991).
Collapse
Affiliation(s)
- Itsuki Watanabe
- KITII Corporation, 4F Marukashiwa tama bldg., 1-6-1, Nihonbashihoncho, Chuo-ku, Tokyo, 103-0023, Japan.
| | - Naoko Suzuki
- ORTHOMEDICO Inc., 2F Sumitomo Fudosan Korakuen Bldg., 1-4-1, Koishikawa, Bunkyo-ku, Tokyo, 112-0002, Japan
| | - Tsuyoshi Takara
- Medical Corporation Seishinkai, Takara Clinic. 9F Taisei Bldg., 2-3-2, Higashi-gotanda, Shinagawa-ku, Tokyo, 141-0022, Japan
| |
Collapse
|
64
|
Weiner HL. Immune mechanisms and shared immune targets in neurodegenerative diseases. Nat Rev Neurol 2025; 21:67-85. [PMID: 39681722 DOI: 10.1038/s41582-024-01046-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/15/2024] [Indexed: 12/18/2024]
Abstract
The immune system plays a major part in neurodegenerative diseases. In some, such as multiple sclerosis, it is the primary driver of the disease. In others, such as Alzheimer disease, amyotrophic lateral sclerosis and Parkinson disease, it has an amplifying role. Immunotherapeutic approaches that target the adaptive and innate immune systems are being explored for the treatment of almost all neurological diseases, and the targets and approaches are often common across diseases. Microglia are the primary immune cells in the brain that contribute to disease pathogenesis, and are consequently a common immune target for therapy. Other therapeutic approaches target components of the peripheral immune system, such as regulatory T cells and monocytes, which in turn act within the CNS. This Review considers in detail how microglia, monocytes and T cells contribute to the pathogenesis of multiple sclerosis, Alzheimer disease, amyotrophic lateral sclerosis and Parkinson disease, and their potential as shared therapeutic targets across these diseases. The microbiome is also highlighted as an emerging therapeutic target that indirectly modulates the immune system. Therapeutic approaches being developed to target immune function in neurodegenerative diseases are discussed, highlighting how immune-based approaches developed to treat one disease could be applicable to multiple other neurological diseases.
Collapse
Affiliation(s)
- Howard L Weiner
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
65
|
Gajewski PD, Bröde P, Claus M, Golka K, Hengstler JG, Watzl C, Wascher E, Getzmann S. The association between hair cortisol and burnout is moderated by age, psychosocial, and immunological markers. Brain Behav Immun Health 2025; 43:100909. [PMID: 39717874 PMCID: PMC11664079 DOI: 10.1016/j.bbih.2024.100909] [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: 11/19/2024] [Accepted: 11/21/2024] [Indexed: 12/25/2024] Open
Abstract
Background Exhaustion and depersonalization are the core symptoms of the occupational burnout. However, burnout is not an all-or-nothing phenomenon, but can occur in a milder to moderate form in otherwise healthy employees. In the last two decades hair cortisol concentrations (HCC) were increasingly related to the cumulative effect of psychosocial stress at work. We analyzed data of the Dortmund Vital Study (Clinicaltrials.gov: NCT05155397) to explore the relationship of HCC and burnout symptoms. Moreover, we asked whether the HCC - burnout association was moderated by work ability, chronic stress, neuroticism, depressive symptoms, and stress-related immunological biomarkers such as T cell concentration, CD4/CD8 cell ratio, and proinflammatory cytokines TNF- α, IL-6, and IL-18. Methods Burnout was assessed by the Oldenburg Burnout Inventory (OLBI), and the Maslach Burnout Inventory (MBI-D) in 196 working adults aged between 20 and 65 years (mean age 42.2 years). Several self-reported variables and biomarkers were collected. Results The results showed an association between HCC and the burnout measures. A series of moderator analyses revealed that the association between HCC and burnout symptoms was substantial for low work ability, high chronic stress level, high neuroticism level, and mild to moderate depressive symptoms. Immunological markers moderated the HCC - burnout association for high concentrations of T cells, low CD4/CD8 ratio and low IL-6, IL-18 and TNF-α concentrations. These interactions were moderated by age showing the largest impact in middle-aged to older individuals. Conclusions The present findings shed light on the complex interaction between burnout symptoms and work ability, chronic stress, personality, and the endocrinological and immunological responses across the working lifespan. These parameters should be considered when assessing the risk for developing burnout and validating the diagnosis of burnout. Trial registration ClinicalTrials.gov NCT05155397; https://clinicaltrials.gov/ct2/show/NCT05155397.
Collapse
Affiliation(s)
- Patrick D. Gajewski
- Leibniz Research Centre for Working Environment and Human Factors (IfADo), Dortmund, Germany
| | - Peter Bröde
- Leibniz Research Centre for Working Environment and Human Factors (IfADo), Dortmund, Germany
| | - Maren Claus
- Leibniz Research Centre for Working Environment and Human Factors (IfADo), Dortmund, Germany
| | - Klaus Golka
- Leibniz Research Centre for Working Environment and Human Factors (IfADo), Dortmund, Germany
| | - Jan G. Hengstler
- Leibniz Research Centre for Working Environment and Human Factors (IfADo), Dortmund, Germany
| | - Carsten Watzl
- Leibniz Research Centre for Working Environment and Human Factors (IfADo), Dortmund, Germany
| | - Edmund Wascher
- Leibniz Research Centre for Working Environment and Human Factors (IfADo), Dortmund, Germany
- German Center for Mental Health (DZPG), Partner Site Bochum/Marburg, Germany
| | - Stephan Getzmann
- Leibniz Research Centre for Working Environment and Human Factors (IfADo), Dortmund, Germany
| |
Collapse
|
66
|
Kurmangaliyeva SS, Madenbayeva AM, Urazayeva ST, Bazargaliyev YS, Kudabayeva KI, Kurmangaliyev KB. The Role of Memory T-Cell Mediated Immunity in Long-term COVID-19: Effects of Vaccination Status. IRANIAN JOURNAL OF MEDICAL SCIENCES 2025; 50:61-68. [PMID: 40026299 PMCID: PMC11870859 DOI: 10.30476/ijms.2024.104003.3744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 11/05/2024] [Accepted: 11/26/2024] [Indexed: 03/05/2025]
Abstract
T-cell-mediated immunity is essential for controlling severe acute respiratory syndrome coronavirus 2 (SARSCoV2) infection, preventing severe disease, and potentially reducing the risk of long-term coronavirus disease (COVID). This study investigated the impact of natural infection, vaccination, and hybrid immunity on T-cell responses, with a particular emphasis on the role of memory T-cells in long-term COVID-19. The present study reviewed current literature on T-cell responses, including memory T-cell development, in individuals with natural SARS-CoV-2 infection, those vaccinated with messenger RNA (mRNA) vaccines, and those with hybrid immunity. It examined studies that compared T-cell activity, immune regulation, and the prevalence of long-term COVID-19 across these groups. Natural infection induces variable T-cell responses, with severe cases showing stronger but sometimes dysregulated immunological activity, which may contribute to prolonged COVID-19. Vaccination, particularly with mRNA vaccines, elicits targeted and consistent T-cell responses, including memory T-cells, reducing disease severity, and the incidence of long-term COVID-19. Hybrid immunity combines natural infection and vaccination, provides the most robust protection, enhanceds memory T-cell responses, and reduces the risk of long-term COVID-19 through balanced immune regulation. Memory T-cells play a critical role in mitigating long-term COVID-19. Vaccination significantly enhances T-cell-mediated immunity, minimizing the risk of chronic symptoms compared to natural infection alone. Hybrid immunity provides the most effective defense, emphasizing the importance of vaccination, even after natural infection, to prevent long-term COVID-19.
Collapse
Affiliation(s)
- Saulesh S. Kurmangaliyeva
- Department of Microbiology, Virology, and Immunology, West Kazakhstan Marat Ospanov Medical University, Aktobe, Kazakhstan
| | - Akzhan M. Madenbayeva
- Department of Internal Diseases 1, West Kazakhstan Marat Ospanov Medical University, Aktobe, Kazakhstan
| | - Saltanat T. Urazayeva
- Department of Epidemiology, West Kazakhstan Marat Ospanov Medical University, Aktobe, Kazakhstan
| | - Yerlan Sh. Bazargaliyev
- Department of Internal Diseases 1, West Kazakhstan Marat Ospanov Medical University, Aktobe, Kazakhstan
| | - Khatimya I. Kudabayeva
- Department of Internal Diseases 1, West Kazakhstan Marat Ospanov Medical University, Aktobe, Kazakhstan
| | - Kairat B. Kurmangaliyev
- Department of Microbiology, Virology, and Immunology, West Kazakhstan Marat Ospanov Medical University, Aktobe, Kazakhstan
| |
Collapse
|
67
|
Pacher-Deutsch C, Schweighofer N, Hanemaaijer M, Marut W, Žukauskaitė K, Horvath A, Stadlbauer V. The microplastic-crisis: Role of bacteria in fighting microplastic-effects in the digestive system. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2025; 366:125437. [PMID: 39631654 DOI: 10.1016/j.envpol.2024.125437] [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: 09/17/2024] [Revised: 11/13/2024] [Accepted: 12/01/2024] [Indexed: 12/07/2024]
Abstract
Plastic particles smaller than 5 mm, referred to as Microplastics, pose health risks, like metabolic, immunological, neurological, reproductive, and carcinogenic effects, after being ingested. Smaller plastic particles are more likely to be absorbed by human cells, with nanoplastics showing higher potential for cellular damage, including DNA fragmentation and altered protein functions. Micro- and nanoplastics (MNPs) affect the gastrointestinal tract by altering the microbial composition, they could influence digestive enzymes, and possibly disrupt mucus layers. In the stomach, they potentially interfere with digestion and barrier functions, while in the intestines, they could increase permeability via inflammation and tissue disruption. MNPs can lead to microbial dysbiosis, leading to gastrointestinal symptoms. By activating inflammatory pathways, altering T cell functions and affecting dendritic cells and macrophages, immune system homeostasis could possibly be disrupted. Probiotics offer potential strategies to alleviate plastic effects, by either degrading plastic particles or directly countering health effects. We compared genetic sequences of probiotics to the genome of known plastic degraders and concluded that no probiotic bacteria could serve the role of plastic degradation. However, probiotics could directly mitigate MNP-health effects. They can restore microbial diversity, enhance the gut barrier, regulate bile acid metabolism, reduce inflammation, regulate insulin balance, and counteract metabolic disruptions. Antioxidative properties protect against lipid peroxidation and MNP-related reproductive system damage. Probiotics can also bind and degrade toxins, like heavy metals and bisphenol A. Additionally, bacteria could be used to aggregate MNPs and reduce their impact. Therefore, probiotics offer a variety of strategies to counter MNP-induced health effects.
Collapse
Affiliation(s)
- Christian Pacher-Deutsch
- Division of Gastroenterology and Hepatology, Medical University of Graz, Graz, Austria; Center for Biomarker Research in Medicine (CBmed), Graz, Austria.
| | | | | | | | - Kristina Žukauskaitė
- Division of Gastroenterology and Hepatology, Medical University of Graz, Graz, Austria; Center for Biomarker Research in Medicine (CBmed), Graz, Austria; Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Angela Horvath
- Division of Gastroenterology and Hepatology, Medical University of Graz, Graz, Austria; Center for Biomarker Research in Medicine (CBmed), Graz, Austria
| | - Vanessa Stadlbauer
- Division of Gastroenterology and Hepatology, Medical University of Graz, Graz, Austria; Center for Biomarker Research in Medicine (CBmed), Graz, Austria; BioTechMed-Graz, Graz, Austria
| |
Collapse
|
68
|
Eltayeb A, Redwan EM. T-cell immunobiology and cytokine storm of COVID-19. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2025; 213:1-30. [PMID: 40246342 DOI: 10.1016/bs.pmbts.2024.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/19/2025]
Abstract
The 2019 coronavirus illness (COVID 2019) first manifests as a newly identified pneumonia and may quickly escalate to acute respiratory distress syndrome, which has caused a global pandemic. Except for individualized supportive care, no curative therapy has been steadfastly advised for COVID-19 up until this point. T cells and virus-specific T lymphocytes are required to guard against viral infection, particularly COVID-19. Delayed immunological reconstitution (IR) and cytokine storm (CS) continue to be significant barriers to COVID-19 cure. While severe COVID-19 patients who survived the disease had considerable lymphopenia and increased neutrophils, especially in the elderly, their T cell numbers gradually recovered. Exhausted T lymphocytes and elevated levels of pro-inflammatory cytokines, including IL6, IL10, IL2, and IL17, are observed in peripheral blood and the lungs. It implies that while convalescent plasma, IL-6 blocking, mesenchymal stem cells, and corticosteroids might decrease CS, Thymosin α1 and adaptive COVID-19-specific T cells could enhance IR. There is an urgent need for more clinical research in this area throughout the world to open the door to COVID-19 treatment in the future.
Collapse
Affiliation(s)
- Ahmed Eltayeb
- Biological Science Department, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Elrashdy M Redwan
- Biological Science Department, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia.
| |
Collapse
|
69
|
Zhou J, An W, Guan L, Shi J, Qin Q, Zhong S, Huang Z, Liu R, Wu C, Ma Z, Qi X, Jiang X, Wang Y, Li S. The clinical significance of T cell infiltration and immune checkpoint expression in central nervous system germ cell tumors. Front Immunol 2025; 16:1536722. [PMID: 39958339 PMCID: PMC11825448 DOI: 10.3389/fimmu.2025.1536722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Accepted: 01/14/2025] [Indexed: 02/18/2025] Open
Abstract
Background Primary central nervous system germ cell tumors (CNS GCTs) are rare intracranial malignancies, and their tumor microenvironment plays a crucial role in tumor initiation and progression. However, the specific characteristics of the immune microenvironment and their clinical significance remain poorly understood. Methods This study included 93 paraffin-embedded tissue samples from 90 patients diagnosed with CNS GCTs. Immunohistochemistry and immunofluorescence staining were used to assess the infiltration patterns of T cell subsets (CD3+, CD4+, CD8+, Foxp3+) and the expression levels of immune checkpoints (CTLA-4, PD-1, PD-L1). Additionally, the study explored the relationship between these immune features and the patient's clinical characteristics and prognosis. Results The study revealed that germinomas exhibited significantly higher infiltration of CD4+ and Foxp3+ T cells compared to non-germinomatous GCTs (NGGCTs). Additionally, CTLA-4 expression was detected in 58.06% of cases, while PD-1 and PD-L1 were expressed in over 90%, with higher CTLA-4 levels in germinomas and elevated PD-L1 levels in NGGCTs. T cell infiltration was positively correlated with immune checkpoint expression, particularly in germinomas. The results also highlighted the strong immunosuppressive nature of the CNS GCTs' tumor microenvironment. Furthermore, T cell infiltration and immune checkpoint expression were closely associated with clinical characteristics and prognosis. Notably, PD-1 expression was identified as an independent prognostic factor for progression-free survival (PFS) and recurrence-free survival (RFS). Conclusion Our study highlighted the distinct characteristics of T cell infiltration and the significant expression of immune checkpoints in CNS GCTs, revealing the highly heterogeneous and immunosuppressive nature of the tumor microenvironment. PD-1 expression was identified as an independent prognostic predictor, offering a foundation for enhancing risk stratification in CNS GCT patients. These findings also support the potential for future clinical applications of immune checkpoint inhibitors, such as PD-1 monoclonal antibodies.
Collapse
Affiliation(s)
- Jiajun Zhou
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing, China
- Department of Neuroimmunology, Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Wenhao An
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing, China
| | - Lei Guan
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing, China
- Department of Neuroimmunology, Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Jinyu Shi
- Department of Neuroimmunology, Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Qiaozhen Qin
- Department of Neuroimmunology, Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Shuai Zhong
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing, China
| | - Zheng Huang
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing, China
| | - Rui Liu
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing, China
| | - Chenxing Wu
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing, China
| | - Zhong Ma
- Department of Pathology, Sanbo Brain Hospital, Capital Medical University, Beijing, China
| | - Xueling Qi
- Department of Pathology, Sanbo Brain Hospital, Capital Medical University, Beijing, China
| | - Xiaoxia Jiang
- Department of Neuroimmunology, Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Yan Wang
- Department of Neuroimmunology, Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Shouwei Li
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
70
|
Leshem R, Sefton KN, Wong CW, Lin IH, Isaac DT, Niepel M, Hurlstone A. Combined PARP14 inhibition and PD-1 blockade promotes cytotoxic T cell quiescence and modulates macrophage polarization in relapsed melanoma. J Immunother Cancer 2025; 13:e010683. [PMID: 39870492 PMCID: PMC11772928 DOI: 10.1136/jitc-2024-010683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 01/09/2025] [Indexed: 01/29/2025] Open
Abstract
BACKGROUND Programmed cell death 1 (PD-1) signaling blockade by immune checkpoint inhibitors (ICI) effectively restores immune surveillance to treat melanoma. However, chronic interferon-gamma (IFNγ)-induced immune homeostatic responses in melanoma cells contribute to immune evasion and acquired resistance to ICI. Poly ADP ribosyl polymerase 14 (PARP14), an IFNγ-responsive gene product, partially mediates IFNγ-driven resistance. PARP14 inhibition prolongs PD-1 blockade responses in preclinical models, but fails to achieve full tumor clearance, suggesting the involvement of additional resistance mechanisms. METHODS We identified a robust PARP14 catalytic inhibitor gene signature and evaluated its association with patient survival. Using preclinical models and single-cell RNA sequencing, we investigated immune and tumor cell adaptations to PARP14 inhibition combined with PD-1 blockade. RESULTS Combining PARP14 inhibition and PD-1 blockade suppressed tumor-associated macrophages while increasing proinflammatory memory macrophages. Moreover, this combination mitigated the terminal exhaustion of cytotoxic T cells by inducing a quiescent state, thereby preserving functionality. Despite the enhanced immune responses, tumor cells developed adaptive resistance by engaging alternative immune evasion pathways. CONCLUSIONS Although adaptive resistance mechanisms re-emerge, PARP14 inhibition combined with PD-1 blockade offers a promising strategy to enhance treatment outcomes and overcome ICI resistance in melanoma, as immune cells are primed for further therapeutic interventions that leverage the quiescent state.
Collapse
Affiliation(s)
- Rotem Leshem
- Division of Infection, Immunity and Respiratory Medicine, The University of Manchester, Manchester, UK
- Lydia Becker Institute of Immunology and Inflammation, The University of Manchester, Manchester, UK
- School of Biological Sciences, The University of Manchester, Manchester, UK
| | - Kieran Neil Sefton
- Division of Infection, Immunity and Respiratory Medicine, The University of Manchester, Manchester, UK
- Lydia Becker Institute of Immunology and Inflammation, The University of Manchester, Manchester, UK
- School of Biological Sciences, The University of Manchester, Manchester, UK
| | - Chun Wai Wong
- Division of Infection, Immunity and Respiratory Medicine, The University of Manchester, Manchester, UK
- Lydia Becker Institute of Immunology and Inflammation, The University of Manchester, Manchester, UK
- School of Biological Sciences, The University of Manchester, Manchester, UK
- Massachusetts General Hospital Cancer Center, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - I-Hsuan Lin
- Bioinformatics Core Facility, The University of Manchester, Manchester, UK
| | | | - Mario Niepel
- Ribon Therapeutics, Cambridge, Massachusetts, USA
| | - Adam Hurlstone
- Division of Infection, Immunity and Respiratory Medicine, The University of Manchester, Manchester, UK
- Lydia Becker Institute of Immunology and Inflammation, The University of Manchester, Manchester, UK
- School of Biological Sciences, The University of Manchester, Manchester, UK
| |
Collapse
|
71
|
Ghazanfari T, Rezaei A, Rezaei R, Kariminia A, Naghizadeh MM, Soroush M, Shams J, Faghihzadeh S, Hassan ZM. The immune cells profiles of individuals with sulfur mustard-induced serious long-term respiratory complications. Int Immunopharmacol 2025; 146:113851. [PMID: 39708483 DOI: 10.1016/j.intimp.2024.113851] [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: 09/19/2024] [Revised: 12/08/2024] [Accepted: 12/10/2024] [Indexed: 12/23/2024]
Abstract
Sulfur mustard (SM) induced pulmonary disorder is a heterogeneous disease characterized by uncontrolled inflammatory immune responses. In this cross-sectional study carried out in Isfahan-Iran, our objective was to thoroughly evaluate the clinical health and peripheral blood leukocyte profiles of adult veterans exposed to SM 25-30 years. In total, 361 people were studied in two groups, 287 chemical veterans with pulmonary complications and 64 healthy individuals as a control group. The participants underwent a comprehensive lung evaluation, including physical examination, Pulmonary Assessment, and Spirometry Assessment. Blood samples were collected in EDTA-treated tubes and flow cytometry analysis was employed to study different population of leukocytes including lymphocytes, monocytes, and natural killer cells. In our results, SM-exposed patients showed a significant increase in mean WBC and lymphocyte absolute count. However, the frequency of CD14+ monocytes and CD3+ CD4+ CD25+Hi as regulatory T cell subsets significantly decreased in SM-exposed patients. In addition, there was a negative correlation between CD45+ CD14+ cells and residual volume (RV). The population of NK cells showed a negative correlation with forced expiratory volume in the first one second to the forced vital capacity (FEV1/FVC). On the other hand, the percentage of CD19+ B cells positively correlated with Mid-maximum expiratory flow (MMEF) rate, ppm Reading, Carboxyhemoglobin (CoHb), and FEV1, and it was negatively correlated with airway resistance (RAW). Evaluation of CD3+ CD8+ cytotoxic T cells frequency negatively correlated with CoHb, ppm Reading, total lung capacity (TLC), and RV. Furthermore, the count of CD3+ CD4+ T cells demonstrated a negative correlation with TLC. The percentage of CD3+ CD4+ CD25+ cells was positively correlated with ppm reading and CoHb. Overall, our findings revealed modifications in total lymphocyte dynamics and a decrease in the percentage and absolute number of regulatory T cells, compromising the regulatory arm of the immune system to modulate SM-induced inflammatory damages.
Collapse
Affiliation(s)
- Tooba Ghazanfari
- Immunoregulation Research Center, Shahed University, Tehran 3319118651, Iran; Department of Immunology, Shahed University, Tehran, Iran.
| | - Abbas Rezaei
- Immunology Department, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Ramazan Rezaei
- Department of Immunology, Medical Faculty, Shahed University, Tehran, Iran
| | - Amina Kariminia
- School of Medicine, University of Washington, Seattle, WA, USA
| | | | | | - Jalaledin Shams
- Hematology-Oncology Unit, Internal Medicine Department, Shahed University, Tehran 3319118651, Iran; Department of Oncology and Hematology, Shahed University, Tehran 3319118651, Iran
| | - Soghrat Faghihzadeh
- Department of Biostatistics and Social Medicine, Zanjan University of Medical Sciences, Zanjan, Iran; Faculty of Medicine, Zanjan University of Medical Science, Zanjan 4515613191, Iran
| | | |
Collapse
|
72
|
Kim YH, Park CH, Kim JM, Yoon YC. Chitooligosaccharides suppress airway inflammation, fibrosis, and mucus hypersecretion in a house dust mite-induced allergy model. FRONTIERS IN ALLERGY 2025; 6:1533928. [PMID: 39927112 PMCID: PMC11799285 DOI: 10.3389/falgy.2025.1533928] [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: 11/25/2024] [Accepted: 01/09/2025] [Indexed: 02/11/2025] Open
Abstract
Background Respiratory allergy is a serious respiratory disorder characterized by inflammation, mucus hypersecretion, and airway tissue sclerosis. Disruption of the T helper 1 (Th1) and T helper 2 (Th2) immune systems by stimuli induced by house dust mites (HDM) and fine particulate matter leads to the secretion of various inflammatory cytokines, resulting in immune respiratory diseases characterized by airway inflammation. Chitooligosaccharides (COS) are known for their antioxidant and anti-inflammatory properties. Methods Human airway epithelial cells (BEAS-2B) were cultured in DMEM/F12 medium containing COS at concentrations of 25-100 µg/ml for 24 h. No intracellular toxicity was observed up to 1,000 µg/ml. Cell experiments were conducted at COS concentrations below 100 µg/ml, while animal experiments were performed at concentrations below 100 mg/kg body weight for 4 weeks. Samples of right lung tissue obtained from the experimental animals were used for gene and protein expression analysis, whereas samples of contralateral lung tissue were used for immunohistochemical analysis. Results COS regulated Th1 immunity by inhibiting major cytokines, including inflammatory tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), and interleukin-6 (IL-6), in BEAS-2B cells. In the HDM-induced allergic respiratory model, COS suppressed the infiltration of inflammatory cells around the airways and inhibited the mRNA expression of Th1 immune cytokines in lung tissues, while also reducing the expression of nuclear factor kappa B (NF-κB)-related proteins. Furthermore, the results confirmed the suppression of the levels of immunoglobulin E (IgE) in the blood secreted by mast cells activated by HDM, which led to a reduction in allergic mucus hypersecretion and airway sclerosis. Conclusion In summary, COS are thought to improve airway resistance by alleviating inflammatory allergic respiratory diseases caused by HDM and are regarded as substances that regulate the balance of the Th1 and Th2 immune systems in epithelial cells affected by mucus hypersecretion.
Collapse
Affiliation(s)
| | | | | | - Yeo Cho Yoon
- Healthcare & Nutrition Laboratory, Amicogen, Inc., Seongnam, Republic of Korea
| |
Collapse
|
73
|
Wang X, Shen W, Yao L, Li C, You H, Guo D. Current status and future prospects of molecular imaging in targeting the tumor immune microenvironment. Front Immunol 2025; 16:1518555. [PMID: 39911388 PMCID: PMC11794535 DOI: 10.3389/fimmu.2025.1518555] [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: 10/28/2024] [Accepted: 01/02/2025] [Indexed: 02/07/2025] Open
Abstract
Molecular imaging technologies have significantly transformed cancer research and clinical practice, offering valuable tools for visualizing and understanding the complex tumor immune microenvironment. These technologies allow for the non-invasive examination of key components within the tumor immune microenvironment, including immune cells, cytokines, and stromal cells, providing crucial insights into tumor biology and treatment responses. This paper reviews the latest advancements in molecular imaging, with a focus on its applications in assessing interactions within the tumor immune microenvironment. Additionally, the challenges faced by molecular imaging technologies are discussed, such as the need for highly sensitive and specific imaging agents, issues with data integration, and difficulties in clinical translation. The future outlook emphasizes the potential of molecular imaging to enhance personalized cancer treatment through the integration of artificial intelligence and the development of novel imaging probes. Addressing these challenges is essential to fully realizing the potential of molecular imaging in improving cancer diagnosis, treatment, and patient outcomes.
Collapse
Affiliation(s)
- Xiang Wang
- Department of Radiology, First People’s Hospital of Linping District, Hangzhou, China
- The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Weifen Shen
- Department of Radiology, First People’s Hospital of Linping District, Hangzhou, China
- The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Lingjun Yao
- Department of Radiology, First People’s Hospital of Linping District, Hangzhou, China
- The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Chao Li
- Department of Radiology, First People’s Hospital of Linping District, Hangzhou, China
- The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Huiming You
- Department of Radiology, First People’s Hospital of Linping District, Hangzhou, China
- The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Duancheng Guo
- Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
74
|
Basingab FS, Alshahrani OA, Alansari IH, Almarghalani NA, Alshelali NH, Alsaiary AH, Alharbi N, Zaher KA. From Pioneering Discoveries to Innovative Therapies: A Journey Through the History and Advancements of Nanoparticles in Breast Cancer Treatment. BREAST CANCER (DOVE MEDICAL PRESS) 2025; 17:27-51. [PMID: 39867813 PMCID: PMC11761866 DOI: 10.2147/bctt.s501448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Accepted: 01/03/2025] [Indexed: 01/28/2025]
Abstract
Nanoparticle technology has revolutionized breast cancer treatment by offering innovative solutions addressing the gaps in traditional treatment methods. This paper aimed to comprehensively explore the historical journey and advancements of nanoparticles in breast cancer treatment, highlighting their transformative impact on modern medicine. The discussion traces the evolution of nanoparticle-based therapies from their early conceptualization to their current applications and future potential. We initially explored the historical context of breast cancer treatment, highlighting the limitations of conventional therapies, such as surgery, radiation, and chemotherapy. The advent of nanotechnology has introduced a new era characterized by the development of various nanoparticles, including liposomes, dendrimers, and gold nanoparticles, designed to target cancer cells with remarkable precision. We further described the mechanisms of action for nanoparticles, including passive and active targeting, and reviewed significant breakthroughs and clinical trials that have validated their efficacy. Current applications of nanoparticles in breast cancer treatment have been examined, showcasing clinically approved therapies and comparing their effectiveness with traditional methods. This article also discusses the latest advancements in nanoparticle research, including drug delivery systems and combination therapy innovations, while addressing the current technical, biological, and regulatory challenges. The technical challenges include efficient and targeted delivery to tumor sites without affecting healthy tissue; biological, such as potential toxicity, immune system activation, or resistance mechanisms; economic, involving high production and scaling costs; and regulatory, requiring rigorous testing for safety, efficacy, and long-term effects to meet stringent approval standards. Finally, we have explored emerging trends, the potential for personalized medicine, and the ethical and social implications of this transformative technology. In conclusion, through comprehensive analysis and case studies, this paper underscores the profound impact of nanoparticles on breast cancer treatment and their future potential.
Collapse
Affiliation(s)
- Fatemah S Basingab
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, 21859, Saudi Arabia
- Immunology Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, 21859, Saudi Arabia
| | - Omniah A Alshahrani
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, 21859, Saudi Arabia
| | - Ibtehal H Alansari
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, 21859, Saudi Arabia
| | - Nada A Almarghalani
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, 21859, Saudi Arabia
| | - Nada H Alshelali
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, 21859, Saudi Arabia
| | - Abeer Hamad Alsaiary
- Biology Department, College of Sciences, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Najwa Alharbi
- Department of Biology Science, Faculty of Science, King Abdulaziz University, Jeddah, 21859, Saudi Arabia
| | - Kawther A Zaher
- Immunology Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, 21859, Saudi Arabia
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, 21859, Saudi Arabia
| |
Collapse
|
75
|
Picone F, Giudice V, Iside C, Venturini E, Di Pietro P, Vecchione C, Selleri C, Carrizzo A. Lymphocyte Subset Imbalance in Cardiometabolic Diseases: Are T Cells the Missing Link? Int J Mol Sci 2025; 26:868. [PMID: 39940640 PMCID: PMC11816853 DOI: 10.3390/ijms26030868] [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: 12/18/2024] [Revised: 01/09/2025] [Accepted: 01/18/2025] [Indexed: 02/16/2025] Open
Abstract
Cardiometabolic and cardiovascular diseases (CVDs) remain the leading cause of death worldwide, with well-established risk factors such as smoking, obesity, and diabetes contributing to plaque formation and chronic inflammation. However, emerging evidence suggests that the immune system plays a more significant role in the development and progression of CVD than previously thought. Specifically, the finely tuned regulation of lymphocyte subsets governs post-injury inflammation and tissue damage resolution and orchestrates the functions and activation of endothelial cells, cardiomyocytes, and fibroblasts in CVD-associated lesions (e.g., atherosclerotic plaques). A deeper understanding of the immune system's involvement in CVD development and progression will provide new insights into disease biology and uncover novel therapeutic targets aimed at re-establishing immune homeostasis. In this review, we summarize the current state of knowledge on the distribution and involvement of lymphocyte subsets in CVD, including atherosclerosis, diabetes, hypertension, myocardial infarction, and stroke.
Collapse
Affiliation(s)
- Francesca Picone
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, 84081 Baronissi, Italy; (F.P.); (C.I.); (P.D.P.); (C.V.); (C.S.)
| | - Valentina Giudice
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, 84081 Baronissi, Italy; (F.P.); (C.I.); (P.D.P.); (C.V.); (C.S.)
- Hematology and Transplant Center, University Hospital “San Giovanni di Dio e Ruggi d’Aragona”, 84131 Salerno, Italy
| | - Concetta Iside
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, 84081 Baronissi, Italy; (F.P.); (C.I.); (P.D.P.); (C.V.); (C.S.)
| | | | - Paola Di Pietro
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, 84081 Baronissi, Italy; (F.P.); (C.I.); (P.D.P.); (C.V.); (C.S.)
| | - Carmine Vecchione
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, 84081 Baronissi, Italy; (F.P.); (C.I.); (P.D.P.); (C.V.); (C.S.)
- Vascular Physiopathology Unit, IRCCS Neuromed, 86077 Pozzilli, Italy;
| | - Carmine Selleri
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, 84081 Baronissi, Italy; (F.P.); (C.I.); (P.D.P.); (C.V.); (C.S.)
- Hematology and Transplant Center, University Hospital “San Giovanni di Dio e Ruggi d’Aragona”, 84131 Salerno, Italy
| | - Albino Carrizzo
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, 84081 Baronissi, Italy; (F.P.); (C.I.); (P.D.P.); (C.V.); (C.S.)
- Vascular Physiopathology Unit, IRCCS Neuromed, 86077 Pozzilli, Italy;
| |
Collapse
|
76
|
Kour D, Bowen CA, Srivastava U, Nguyen HM, Kumari R, Kumar P, Brandelli AD, Bitarafan S, Tobin BR, Wood L, Seyfried NT, Wulff H, Rangaraju S. Identification of novel Kv1.3 channel-interacting proteins using proximity labelling in T-cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.16.633279. [PMID: 39868101 PMCID: PMC11760797 DOI: 10.1101/2025.01.16.633279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Potassium channels regulate membrane potential, calcium flux, cellular activation and effector functions of adaptive and innate immune cells. The voltage-activated Kv1.3 channel is an important regulator of T cell-mediated autoimmunity and microglia-mediated neuroinflammation. Kv1.3 channels, via protein-protein interactions, are localized with key immune proteins and pathways, enabling functional coupling between K+ efflux and immune mechanisms. To gain insights into proteins and pathways that interact with Kv1.3 channels, we applied a proximity-labeling proteomics approach to characterize protein interactors of the Kv1.3 channel in activated T-cells. Biotin ligase TurboID was fused to either N or C termini of Kv1.3, stably expressed in Jurkat T cells and biotinylated proteins in proximity to Kv1.3 were enriched and quantified by mass spectrometry. We identified over 1,800 Kv1.3 interactors including known interactors (beta-integrins, Stat1) although majority were novel. We found that the N-terminus of Kv1.3 preferentially interacts with protein synthesis and protein trafficking machinery, while the C-terminus interacts with immune signaling and cell junction proteins. T-cell Kv1.3 interactors included 335 cell surface, T-cell receptor complex, mitochondrial, calcium and cytokine-mediated signaling pathway and lymphocyte migration proteins. 178 Kv1.3 interactors in T-cells also represent genetic risk factors of T cell-mediated autoimmunity, including STIM1, which was further validated using co-immunoprecipitation. Our studies reveal novel proteins and molecular pathways that interact with Kv1.3 channels in adaptive (T-cell) and innate immune (microglia), providing a foundation for how Kv1.3 channels may regulate immune mechanisms in autoimmune and neurological diseases.
Collapse
Affiliation(s)
- Dilpreet Kour
- Department of Neurology, School of Medicine, Yale University, New Haven (CT), USA
| | - Christine A. Bowen
- Center for Neurodegenerative Diseases, Emory University, Atlanta (GA), USA
- Department of Biochemistry, Emory University, Atlanta (GA), USA
| | - Upasna Srivastava
- Department of Neurology, School of Medicine, Yale University, New Haven (CT), USA
| | - Hai M. Nguyen
- Department of Pharmacology, University of California – Davis, Davis (CA), USA
| | - Rashmi Kumari
- Department of Neurology, School of Medicine, Yale University, New Haven (CT), USA
| | - Prateek Kumar
- Department of Neurology, School of Medicine, Yale University, New Haven (CT), USA
| | - Amanda D. Brandelli
- Department of Neurology, School of Medicine, Yale University, New Haven (CT), USA
| | - Sara Bitarafan
- Parker H. Petit Institute for Bioengineering, Georgia Institute of Technology, Atlanta (GA), USA
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta (GA), USA
| | - Brendan R Tobin
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta (GA), USA
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta (GA), USA
| | - Levi Wood
- Parker H. Petit Institute for Bioengineering, Georgia Institute of Technology, Atlanta (GA), USA
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta (GA), USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta (GA), USA
| | - Nicholas T. Seyfried
- Center for Neurodegenerative Diseases, Emory University, Atlanta (GA), USA
- Department of Biochemistry, Emory University, Atlanta (GA), USA
| | - Heike Wulff
- Department of Pharmacology, University of California – Davis, Davis (CA), USA
| | - Srikant Rangaraju
- Department of Neurology, School of Medicine, Yale University, New Haven (CT), USA
| |
Collapse
|
77
|
Sardana K, Mathachan SR, Muddebihal A, Agrawal D, Ahuja A. Translating tissue expression of STAT 1, 3 and 6 in prurigo nodularis to clinical efficacy of oral tofacitinib - A prospective single-arm investigational study. Indian J Dermatol Venereol Leprol 2025; 0:1-6. [PMID: 39912139 DOI: 10.25259/ijdvl_1017_2024] [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: 07/11/2024] [Accepted: 10/15/2024] [Indexed: 02/07/2025]
Abstract
Background Interleukin (IL)-4, IL-13, IL-17, IL-22 and IL-3 are overexpressed in prurigo nodularis (PN). They mediate their action via the Janus Kinase (JAK) Signal transducer and activator of transcription (STAT) pathway. Objectives Our aim was to study the expression of tissue STAT1, STAT3, and STAT6, as well as the efficacy of the JAK-STAT inhibitor, tofacitinib, in PN. Methods A prospective study was conducted in a tertiary care hospital. Patients with PN were recruited after excluding secondary causes. Pruritus was graded using Pruritus Grading System Score (PGSS). All cases underwent histological assessment using immunohistochemical markers for STAT1, STAT3, and STAT6 in both lesional and perilesional skin. Tofacitinib was initiated at a dose of 5 mg twice daily or 11 mg once daily and then tapered to a maintenance dose. The final PGSS at the time of data evaluation, as well as the occurrence of remissions and relapses, was assessed. Results The majority of the 17 patients included in the study had moderate to severe disease. Immunohistochemical analysis revealed marked tissue expression of STAT6 in 13 and STAT3 in 10 patients, while STAT1 expression was seen in only 4 patients [p < 0.05], suggesting a Th2/Th17 tissue response. The mean onset of action of tofacitinib was 11.2 ± 6.44 days and the mean duration of treatment was 5.6 ± 2.2 months. A significant reduction in PGSS was noted after treatment (66.1%, P value 0.0004). Fourteen of the patients maintained remission on low-dose therapy (5 mg OD or A/D) while one patient experienced a relapse. No serious adverse effects were noted. Limitation We could not study the tissue cytokines and the expression of STATs after achieving clinical response on oral tofacitinib. Conclusion The efficacy of tofacitinib in PN is based on its inhibitory effect on Th2 and Th17 cytokines, which is dependent on STAT6 and STAT3.
Collapse
Affiliation(s)
- Kabir Sardana
- Department of Dermatology, Atal Bihari Vajpayee Institute of Medical Sciences & Dr. Ram Manohar Lohia Hospital, New Delhi, India
| | - Sinu Rose Mathachan
- Department of Dermatology, Aster DM Healthcare, Discovery Gardens, Dubai, United Arab Emirates
| | - Aishwarya Muddebihal
- Department of Dermatology, Atal Bihari Vajpayee Institute of Medical Sciences & Dr. Ram Manohar Lohia Hospital, New Delhi, India
| | - Diksha Agrawal
- Department of Dermatology, Venkateshwara Institute of Medical Sciences, Amroha, India
| | - Arvind Ahuja
- Department of Pathology, Atal Bihari Vajpayee Institute of Medical Sciences & Dr. Ram Manohar Lohia Hospital, Delhi, India
| |
Collapse
|
78
|
Basmenj ER, Pajhouh SR, Ebrahimi Fallah A, naijian R, Rahimi E, Atighy H, Ghiabi S, Ghiabi S. Computational epitope-based vaccine design with bioinformatics approach; a review. Heliyon 2025; 11:e41714. [PMID: 39866399 PMCID: PMC11761309 DOI: 10.1016/j.heliyon.2025.e41714] [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: 12/20/2024] [Accepted: 01/03/2025] [Indexed: 01/28/2025] Open
Abstract
The significance of vaccine development has gained heightened importance in light of the COVID-19 pandemic. In such critical circumstances, global citizens anticipate researchers in this field to swiftly identify a vaccine candidate to combat the pandemic's root cause. It is widely recognized that the vaccine design process is traditionally both time-consuming and costly. However, a specialized subfield within bioinformatics, known as "multi-epitope vaccine design" or "reverse vaccinology," has significantly decreased the time and costs of the vaccine design process. The methodology reverses itself in this subfield and finds a potential vaccine candidate by analyzing the pathogen's genome. Leveraging the tools available in this domain, we strive to pinpoint the most suitable antigen for crafting a vaccine against our target. Once the optimal antigen is identified, the next step involves uncovering epitopes within this antigen. The immune system recognizes particular areas of an antigen as epitopes. By characterizing these crucial segments, we gain the opportunity to design a vaccine centered around these epitopes. Subsequently, after identifying and assembling the vital epitopes with the assistance of linkers and adjuvants, our vaccine candidate can be formulated. Finally, employing computational techniques, we can thoroughly evaluate the designed vaccine. This review article comprehensively covers the entire multi-epitope vaccine development process, starting from obtaining the pathogen's genome to identifying the relevant vaccine candidate and concluding with an evaluation. Furthermore, we will delve into the essential tools needed at each stage, comparing and introducing them.
Collapse
Affiliation(s)
| | | | | | - Rafe naijian
- Student research committee, faculty of pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Elmira Rahimi
- Department of Biology, Central Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Hossein Atighy
- School of Pharmacy, Centro Escolar University, Manila, Philippines
| | - Shadan Ghiabi
- Faculty of Veterinary Medicine, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Shamim Ghiabi
- Tehran Azad University of Medical Sciences, Faculty of Pharmaceutical Sciences, Iran
| |
Collapse
|
79
|
Lin Y, Liao X, Cao X, Zhang Z, Wang X, He X, Liao H, Ju B, Qi F, Xu H, Ren Z, Wang Y, Hu Z, Yang J, Fu YX, Zhao J, Zhang Z, Peng H. Sequential intranasal booster triggers class switching from intramuscularly primed IgG to mucosal IgA against SARS-CoV-2. J Clin Invest 2025; 135:e175233. [PMID: 39808503 PMCID: PMC11870729 DOI: 10.1172/jci175233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 01/08/2025] [Indexed: 01/16/2025] Open
Abstract
The persistent emergence of COVID-19 variants and recurrent waves of infection worldwide underscores the urgent need for vaccines that effectively reduce viral transmission and prevent infections. Current intramuscular (IM) COVID-19 vaccines inadequately protect the upper respiratory mucosa. In response, we have developed a nonadjuvanted, IFN-armed SARS-CoV-2 fusion protein vaccine with IM priming and intranasal (IN) boost sequential immunization. Our study showed that this sequential vaccination strategy of the IM+IN significantly enhanced both upper respiratory and systemic antiviral immunity in a mouse model, characterized by the rapid increase in systemic and mucosal T and B cell responses, particularly the mucosal IgA antibody response. The IN boost triggered a swift secondary immune response, rapidly inducing antigen-specific IgA+ B cells. Further B cell receptor-seq (BCR-seq) analysis indicated that these IgA+ B cells primarily arose through direct class switching from preexisting IgG+ B cells in draining lymph nodes. Notably, our clinical studies revealed that the IN boost after IM vaccination elicited a robust systemic IgA antibody response in humans, as measured in serum. Thus, we believe that our cytokine-armed protein vaccine presents a promising strategy for inducing rapid and potent mucosal protection against respiratory viral infections.
Collapse
Affiliation(s)
- Yifan Lin
- Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, China
- Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Xuejiao Liao
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Xuezhi Cao
- Guangzhou Laboratory, Guangzhou International Bio-Island, Guangzhou, China
| | - Zhaoyong Zhang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Xiuye Wang
- Guangzhou Laboratory, Guangzhou International Bio-Island, Guangzhou, China
| | - Xiaomeng He
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | | | - Bin Ju
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Furong Qi
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Hairong Xu
- Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | | | - Yanqun Wang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | | | | | - Yang-Xin Fu
- Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, China
| | - Jincun Zhao
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, China
- Guangzhou Laboratory, Guangzhou International Bio-Island, Guangzhou, China
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Zheng Zhang
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Hua Peng
- Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- Guangzhou Laboratory, Guangzhou International Bio-Island, Guangzhou, China
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
80
|
Ye J, Yu Q, Chen Y, Huang C. Case report: Kikuchi-Fujimoto disease presenting with persistent fever and widespread lymphadenopathy in a young adult. Front Immunol 2025; 15:1519988. [PMID: 39867904 PMCID: PMC11757135 DOI: 10.3389/fimmu.2024.1519988] [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: 10/30/2024] [Accepted: 12/30/2024] [Indexed: 01/28/2025] Open
Abstract
Kikuchi-Fujimoto disease (KFD) is a rare, self-limiting condition typically characterized by fever and lymphadenopathy. The exact etiology remains unclear but is suspected to be associated with viral infections and autoimmune responses. This report presents the case of a 32-year-old Chinese male who was admitted with recurrent high fever, lymphadenopathy, and hepatosplenomegaly. Initial treatment was ineffective, and a lymph node biopsy subsequently confirmed the diagnosis of KFD, with evidence of cytomegalovirus infection. Following treatment with corticosteroids, the patient's symptoms improved rapidly, and no relapse was observed during follow-up after discharge. This case highlights the diagnostic challenges of KFD, particularly in distinguishing it from lymphoma and systemic lupus erythematosus. Accurate and timely diagnosis is crucial to avoid unnecessary treatments, and long-term follow-up is recommended to monitor for potential disease progression.
Collapse
Affiliation(s)
- Jing Ye
- Department of General Practice, The Affiliated Panyu Central Hospital, Guangzhou Medical University, Guangzhou, China
| | - Qian Yu
- Department of Pathology, The Affiliated Panyu Central Hospital, Guangzhou Medical University, Guangzhou, China
| | - Yan Chen
- Department of General Practice, The Affiliated Panyu Central Hospital, Guangzhou Medical University, Guangzhou, China
| | - Chunping Huang
- Department of General Practice, The Affiliated Panyu Central Hospital, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
81
|
Yu X, Pei W, Li B, Sun S, Li W, Wu Q. Immunosenescence, Physical Exercise, and their Implications in Tumor Immunity and Immunotherapy. Int J Biol Sci 2025; 21:910-939. [PMID: 39897036 PMCID: PMC11781184 DOI: 10.7150/ijbs.100948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Accepted: 12/28/2024] [Indexed: 02/04/2025] Open
Abstract
Aging is associated with a decline in immune function, termed immunosenescence, which compromises host defences and increases susceptibility to infections and cancer. Physical exercise is widely recognized for its myriad health benefits, including the potential to modulate the immune system. This review explores the bidirectional relationship between immunosenescence and physical exercise, focusing on their interplay in shaping antitumor immunity. We summarize the impact of aging on innate and adaptive immune cells, highlighting alterations that contribute to immunosenescence and cancer development. We further delineate the effects of exercise on immune cell function, demonstrating its potential to mitigate immunosenescence and enhance antitumor responses. We also discuss the implications of immunosenescence for the efficacy of immunotherapies, such as immune checkpoint inhibitors and adoptive T cell therapy, and explore the potential benefits of combining exercise with these interventions. Collectively, this review underscores the importance of understanding the complex relationship between immunosenescence, physical exercise, and antitumor immunity, paving the way for the development of innovative strategies to improve cancer outcomes in the aging population.
Collapse
Affiliation(s)
- Xin Yu
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, P. R. China
| | - Wei Pei
- Tongji University Cancer Center, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, P. R. China
| | - Bei Li
- Department of Pathology, Renmin Hospital of Wuhan University, Wuhan, Hubei, P. R. China
| | - Shengrong Sun
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, P. R. China
| | - Wenge Li
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, P. R. China
- Department of Oncology, Shanghai GoBroad Cancer Hospital, Shanghai, P. R. China
| | - Qi Wu
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, P. R. China
- Tongji University Cancer Center, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, P. R. China
| |
Collapse
|
82
|
Huang J, Zhang J, Song P, Huang J, Yang Z, Han J, Wu L, Guo X. p38α-eIF6-Nsun2 axis promotes ILC3's rapid response to protect host from intestinal inflammation. J Exp Med 2025; 222:e20240624. [PMID: 39589554 PMCID: PMC11602552 DOI: 10.1084/jem.20240624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 09/01/2024] [Accepted: 10/08/2024] [Indexed: 11/27/2024] Open
Abstract
Group 3 innate lymphoid cells (ILC3s) are important for maintaining gut homeostasis. Upon stimulation, ILC3s can rapidly produce cytokines to protect against infections and colitis. However, the regulation of ILC3 quick response is still unclear. Here, we find that eIF6 aggregates with Nsun2 and cytokine mRNA in ILC3s at steady state, which inhibits the methyltransferase activity of Nsun2 and the nuclear export of cytokine mRNA, resulting in the nuclear reservation of cytokine mRNA. Upon stimulation, phosphorylated p38α phosphorylates eIF6, which in turn releases Nsun2 activity, and promotes the nuclear export of cytokine mRNA and rapid cytokine production. Genetic disruption of p38α, Nsun2, or eIF6 in ILC3s influences the mRNA nuclear export and protein expression of the protective cytokines, thus leading to increased susceptibility to colitis. Together, our data identify a crucial role of the p38α-eIF6-Nsun2 axis in regulating rapid ILC3 immune response at the posttranscriptional level, which is critical for gut homeostasis maintenance and protection against gut inflammation.
Collapse
Affiliation(s)
- Jida Huang
- Institute for Immunology, Tsinghua University, Beijing, China
- School of Basic Medical Sciences, Tsinghua University, Beijing, China
- Beijing Key Lab for Immunological Research on Chronic Diseases, Tsinghua University, Beijing, China
- State Key Laboratory of Molecular Oncology, School of Basic Medical Sciences, Tsinghua University, Beijing, China
| | - Jing Zhang
- Institute for Immunology, Tsinghua University, Beijing, China
- School of Basic Medical Sciences, Tsinghua University, Beijing, China
- Beijing Key Lab for Immunological Research on Chronic Diseases, Tsinghua University, Beijing, China
- State Key Laboratory of Molecular Oncology, School of Basic Medical Sciences, Tsinghua University, Beijing, China
| | - Panwei Song
- Institute for Immunology, Tsinghua University, Beijing, China
- School of Basic Medical Sciences, Tsinghua University, Beijing, China
- Beijing Key Lab for Immunological Research on Chronic Diseases, Tsinghua University, Beijing, China
- State Key Laboratory of Molecular Oncology, School of Basic Medical Sciences, Tsinghua University, Beijing, China
| | - Jiaoyan Huang
- Institute for Immunology, Tsinghua University, Beijing, China
- School of Basic Medical Sciences, Tsinghua University, Beijing, China
- Beijing Key Lab for Immunological Research on Chronic Diseases, Tsinghua University, Beijing, China
| | - Zi Yang
- Protein Preparation and Identification Facilities at Technology Center for Protein Science, School of Life Sciences, Tsinghua University, Beijing, China
| | - Jiahuai Han
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Innovation Center for Cell Signaling Network, Xiamen University, Xiamen, China
| | - Li Wu
- Institute for Immunology, Tsinghua University, Beijing, China
- School of Basic Medical Sciences, Tsinghua University, Beijing, China
- Beijing Key Lab for Immunological Research on Chronic Diseases, Tsinghua University, Beijing, China
| | - Xiaohuan Guo
- Institute for Immunology, Tsinghua University, Beijing, China
- School of Basic Medical Sciences, Tsinghua University, Beijing, China
- Beijing Key Lab for Immunological Research on Chronic Diseases, Tsinghua University, Beijing, China
- State Key Laboratory of Molecular Oncology, School of Basic Medical Sciences, Tsinghua University, Beijing, China
- SXMU-Tsinghua Collaborative Innovation Center for Frontier Medicine, Shanxi Medical University, Taiyuan, China
| |
Collapse
|
83
|
Hoque M, Grigg JB, Ramlall T, Jones J, McGoldrick LL, Lin JC, Olson WC, Smith E, Franklin MC, Zhang T, Saotome K. Structural characterization of two γδ TCR/CD3 complexes. Nat Commun 2025; 16:318. [PMID: 39747888 PMCID: PMC11697310 DOI: 10.1038/s41467-024-55467-5] [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: 07/31/2024] [Accepted: 12/11/2024] [Indexed: 01/04/2025] Open
Abstract
The T-cell receptor (TCR)/CD3 complex plays an essential role in the immune response and is a key player in cancer immunotherapies. There are two classes of TCR/CD3 complexes, defined by their TCR chain usage (αβ or γδ). Recently reported structures have revealed the organization of the αβ TCR/CD3 complex, but similar studies regarding the γδ TCR/CD3 complex have lagged behind. Here, we report cryoelectron microscopy (cryoEM) structural analysis of two γδ TCRs, G115 (Vγ9 Vδ2) and 9C2 (Vγ5 Vδ1), in complex with CD3 subunits. Our results show that the overall subunit organization of the γδ TCR/CD3 complexes is similar to αβ TCRs. However, both γδ TCRs display highly mobile extracellular domains (ECDs), unlike αβ TCRs, which have TCR ECDs that are rigidly coupled to its transmembrane (TM) domains. We corroborate this finding in cells by demonstrating that a γδ T-cell specific antibody can bind a site that would be inaccessible in the more rigid αβ TCR/CD3 complex. Furthermore, we observed that the Vγ5 Vδ1 complex forms a TCR γ5 chain-mediated dimeric species whereby two TCR/CD3 complexes are assembled. Collectively, these data shed light on γδ TCR/CD3 complex formation and may aid the design of γδ TCR-based therapies.
Collapse
MESH Headings
- Receptors, Antigen, T-Cell, gamma-delta/immunology
- Receptors, Antigen, T-Cell, gamma-delta/metabolism
- Receptors, Antigen, T-Cell, gamma-delta/chemistry
- Cryoelectron Microscopy
- Humans
- CD3 Complex/immunology
- CD3 Complex/metabolism
- CD3 Complex/chemistry
- Receptors, Antigen, T-Cell, alpha-beta/chemistry
- Receptors, Antigen, T-Cell, alpha-beta/immunology
- Receptors, Antigen, T-Cell, alpha-beta/metabolism
- Models, Molecular
- Protein Domains
Collapse
Affiliation(s)
- Mohammed Hoque
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, 10591, USA.
| | | | - Trudy Ramlall
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, 10591, USA
| | - Jennifer Jones
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, 10591, USA
| | | | - John C Lin
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, 10591, USA
| | | | - Eric Smith
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, 10591, USA
| | | | - Tong Zhang
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, 10591, USA.
| | - Kei Saotome
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, 10591, USA.
| |
Collapse
|
84
|
Zhu K, Rohila D, Zhao Y, Shytikov D, Wu L, Zhao F, Hu S, Xu Q, Jin X, Lu L. Protein Phosphatase 2A Promotes CD8 + T Cell Effector Function through the Augmentation of CD28 Costimulation. RESEARCH (WASHINGTON, D.C.) 2025; 8:0545. [PMID: 39759159 PMCID: PMC11694323 DOI: 10.34133/research.0545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Revised: 10/05/2024] [Accepted: 11/14/2024] [Indexed: 01/07/2025]
Abstract
Protein phosphatase 2A (PP2A) is one of the most abundant serine/threonine phosphatases and plays critical roles in regulating cell fate and function. We previously showed that PP2A regulates the differentiation of CD4+ T cells and the development of thymocytes. Nevertheless, its role in CD8+ T cells remains elusive. By ablating the catalytic subunit α (Cα) of PP2A in CD8+ T cells, we revealed the essential role of PP2A in promoting the effector functions of CD8+ T cells. Notably, PP2A Cα-deficient CD8+ T cells exhibit reduced proliferation and decreased cytokine production upon stimulation in vitro. In vivo, mice lacking PP2A Cα in T cells displayed defective immune responses against lymphocytic choriomeningitis virus infection, associated with reduced CD8+ T cell expansion and decreased cytokine production. Consistently, the ablation of the PP2A Cα subunit in CD8+ T cells results in attenuated antitumor activity in mice. There is a notable decrease in the infiltration of PP2A Cα-deficient CD8+ T cells within the tumor microenvironment, and the cells that do infiltrate exhibit diminished effector functions. Mechanistically, PP2A Cα deficiency impedes CD28-induced AKT Ser473 phosphorylation, thus impairing CD8+ T cell costimulation signal. Collectively, our findings underscore the critical role of phosphatase PP2A as a propeller for CD28-mediated costimulation signaling in CD8+ T cell effector function by fine-tuning T cell activation.
Collapse
Affiliation(s)
- Kaixiang Zhu
- Department of Cardiology of The Second Affiliated Hospital,
Zhejiang University School of Medicine, Hangzhou 310009, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou 310009, China
- Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Hangzhou 310009, China
| | - Deepak Rohila
- Institute of Immunology, and Department of Rheumatology in Sir Run Run Shaw Hospital,
Zhejiang University School of Medicine, Hangzhou 310058, China
- Sanford Burnham Prebys Medical Discovery Institute, San Diego, CA, USA
| | - Yuanling Zhao
- Institute of Immunology, and Department of Rheumatology in Sir Run Run Shaw Hospital,
Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Dmytro Shytikov
- Zhejiang University–University of Edinburgh Institute, Zhejiang University School of Medicine, 314400 Haining, China
| | - Lize Wu
- Institute of Immunology, and Department of Rheumatology in Sir Run Run Shaw Hospital,
Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Fan Zhao
- Institute of Immunology, and Department of Rheumatology in Sir Run Run Shaw Hospital,
Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Shurong Hu
- Department of Gastroenterology, The Second Affiliated Hospital,
Zhejiang University School of Medicine, Hangzhou, China
| | - Qin Xu
- Cell Signaling and Immunity Section, Laboratory of Immune System Biology (LISB),
National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Xuexiao Jin
- Institute of Immunology, and Department of Rheumatology in Sir Run Run Shaw Hospital,
Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Linrong Lu
- Department of Cardiology of The Second Affiliated Hospital,
Zhejiang University School of Medicine, Hangzhou 310009, China
- Institute of Immunology, and Department of Rheumatology in Sir Run Run Shaw Hospital,
Zhejiang University School of Medicine, Hangzhou 310058, China
- Shanghai Immune Therapy Institute,
Shanghai Jiao Tong University School of Medicine Affiliated Renji Hospital, Shanghai 200025, China
| |
Collapse
|
85
|
Strober BJ, Zhang MJ, Amariuta T, Rossen J, Price AL. Fine-mapping causal tissues and genes at disease-associated loci. Nat Genet 2025; 57:42-52. [PMID: 39747598 DOI: 10.1038/s41588-024-01994-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 10/18/2024] [Indexed: 01/04/2025]
Abstract
Complex diseases often have distinct mechanisms spanning multiple tissues. We propose tissue-gene fine-mapping (TGFM), which infers the posterior inclusion probability (PIP) for each gene-tissue pair to mediate a disease locus by analyzing summary statistics and expression quantitative trait loci (eQTL) data; TGFM also assigns PIPs to non-mediated variants. TGFM accounts for co-regulation across genes and tissues and models uncertainty in cis-predicted expression models, enabling correct calibration. We applied TGFM to 45 UK Biobank diseases or traits using eQTL data from 38 Genotype-Tissue Expression (GTEx) tissues. TGFM identified an average of 147 PIP > 0.5 causal genetic elements per disease or trait, of which 11% were gene-tissue pairs. Causal gene-tissue pairs identified by TGFM reflected both known biology (for example, TPO-thyroid for hypothyroidism) and biologically plausible findings (for example, SLC20A2-artery aorta for diastolic blood pressure). Application of TGFM to single-cell eQTL data from nine cell types in peripheral blood mononuclear cells (PBMCs), analyzed jointly with GTEx tissues, identified 30 additional causal gene-PBMC cell type pairs.
Collapse
Affiliation(s)
- Benjamin J Strober
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
| | - Martin Jinye Zhang
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Computational Biology Department, School of Computer Science, Carnegie Mellon University, Pittsburgh, PA, USA
| | - Tiffany Amariuta
- Halıcıoğlu Data Science Institute, University of California San Diego, La Jolla, CA, USA
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Jordan Rossen
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Alkes L Price
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
| |
Collapse
|
86
|
Wik JA, Stromsnes K, Skålhegg BS. Using Seahorse Technology as an Efficient Way of Verifying T Cell Stimulation. Methods Mol Biol 2025; 2904:219-242. [PMID: 40220237 DOI: 10.1007/978-1-0716-4414-0_16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/14/2025]
Abstract
Antibody-dependent stimulation is commonly used to activate, differentiate, and/or to expand T cells for downstream analysis and functions. Such stimulations are inherently connected to endogenous phosphorylation which is commonly assessed using various methods including immunoblotting. However, antibody-dependent stimulation of T cells is also inherently connected to changes in endogenous metabolic activity. We describe methods used to stimulate T cells using soluble and immobilized antibodies in conjunction with immunosuppressive cyclic AMP (cAMP). The stimulations were assessed by downstream phosphorylation using immunoblotting and metabolic changes using Seahorse technology. We use phosphorylation of ERK 1/2, extracellular acidification rate (ECAR) and oxygen consumption rate (OCR) as readouts to represent the activation state of T cells. We also describe how these methods can be used to assess inhibitory stimuli exemplified by the cAMP analogue 8-(4-Chlorophenylthio) adenosine 3',5'-cyclic monophosphate (8-CPT-cAMP), but only if 8-CPT-cAMP is added before anti-CD3. Together the methods described in this chapter provide a comprehensive guideline to isolate, stimulate, and assess T cell stimulation. Moreover, we demonstrate the Seahorse technology as a time and work-efficient way of assessing the effects of T cell stimulation in real time.
Collapse
Affiliation(s)
- Jonas Aakre Wik
- Division of Molecular Nutrition, Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
- Department of Immunology and Transfusion Medicine, Oslo University Hospital, Oslo, Norway
- Hybrid Technology Hub - Centre of Excellence, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Kristine Stromsnes
- Division of Molecular Nutrition, Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Bjørn Steen Skålhegg
- Division of Molecular Nutrition, Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway.
| |
Collapse
|
87
|
Almaeen AH, Saad-Eldien HM, Gabr H. INF-γ/TGF-β1-primed umbilical cord mesenchymal stem cells boost the T-lymphocytes activity: Modulation of CD25 expression and IL-6 secretion. Int J Immunopathol Pharmacol 2025; 39:3946320251315007. [PMID: 39921228 PMCID: PMC11806471 DOI: 10.1177/03946320251315007] [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: 05/30/2024] [Accepted: 12/22/2024] [Indexed: 02/10/2025] Open
Abstract
BACKGROUND Mesenchymal stromal/stem cells (MSCs) have potent immunomodulatory abilities, particularly in a milieu of hyperactive immune system, through secreting a number of cytokines, growth factors, bioactive compounds and peptides, and by cell-cell contact. During viral infection, failure of immuno-neutralization of the viral particles, recruits T-lymphocytes (T-cells) that clear the virally-infected cells. MSCs greatly potentiate T-cells anti-viral activity. OBJECTIVE The objective of this study is to assess the ability of the cytokine-primed MSCs to activated T-cells, towards an antiviral application. METHOD Human umbilical cord MSCs (UC-MSCs) were isolated from Wharton Jelly of a consented donor. UC-MSCs were primed with interferon (INF)-γ and transforming growth factor (TGF)-β1. Peripheral blood T-cells were isolated using mini-max and CD3+ population was purified using anti-CD3 immuno-magnetic beads. Naïve or primed MSCs were co-cultured with naïve and phytohemagglutinin (PHA)-activated CD3+ T-cells. T-cell activation was evaluated by changes in their rate of proliferation by cell count, flowcytometric immuno-phenotyping for CD25 expression, and IL-6 secretion in the conditioned medium. RESULTS The findings revealed that CD3+ T-cells count nonsignificant differed comparing the five experimental groups; Naïve MSCs, Naïve T cells, coculture with naïve MSCs, coculture with primed MSCs, and upon phytohemagglutinin-activation, despite a nonsignificant reduction of proliferation in the last two groups' coculture. Only the coculture with the primed MSCs showed significant activation of T-cells assessed as CD25 expression compared to the other groups (p < 0.001 and p = 0.002, respectively). The undetectable levels of IL-6 in the conditioned medium of naïve MSCs, turned markedly high after their cytokine-priming (p < 0.001), reaching nonsignificant difference compared to naïve T-cells. Compared to naïve MSCs, naïve T-cells secreted considerable amounts of IL-6 (p < 0.001). Incubation of naïve MSCs with phytohemagglutinin-activated T-cells further the secretion of IL-6, to a level significantly higher than all of the aforementioned three groups; naïve MSCs, naïve T-cells and primed MSCs (p < 0.001, p = 0.0194, and p < 0.001, respectively). However, coculture of the cytokine-primed MSCs with phytohemagglutinin-activated T-cells dampened IL-6 secretion to a level that was significantly lower than that observed with naïve MSCs-phytohemagglutinin-activated T-cells coculture (p < 0.001). CONCLUSION The cytokine-primed UC-MSCs significantly upregulated CD25+ expression on T-cells, while hindering IL-6, without affecting their proliferation rate. This may point to potentially stronger antiviral effects, while alleviating the viral infection-induced cytokine storm.
Collapse
Affiliation(s)
| | - Heba M Saad-Eldien
- Department of Anatomy, College of Medicine, Jouf University, Sakaka, Saudi Arabia
| | - Hala Gabr
- Department of Hematology, Faculty of Medicine, Cairo University, Cairo, Egypt
| |
Collapse
|
88
|
Gossink EM, Coffer PJ, Cutilli A, Lindemans CA. Immunomodulation by galectin-9: Distinct role in T cell populations, current therapeutic avenues and future potential. Cell Immunol 2025; 407:104890. [PMID: 39571310 DOI: 10.1016/j.cellimm.2024.104890] [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: 07/13/2024] [Revised: 10/31/2024] [Accepted: 11/04/2024] [Indexed: 12/15/2024]
Abstract
Galectins, glycan-binding proteins, have been identified as critical regulators of the immune system. Recently, Galectin-9 (Gal-9) has emerged as biomarker that correlates with disease severity in a range of inflammatory conditions. However, Gal-9 has highly different roles in the context of immunoregulation, with the potential to either stimulate or suppress the immune response. Neutralizing antibodies targeting Gal-9 have been developed and are in early test phase investigating their therapeutic potential in cancer. Despite ongoing research, the mechanisms behind Gal-9 action remain not fully understood, and extrapolating the implications of targeting this molecule from previous studies is challenging. Here, we examine the pleiotropic function of Gal-9 focusing on conventional T lymphocytes, providing a current overview of its immunostimulatory and immunosuppressive roles. In particular, we highlight that Gal-9 differentially regulates immune responses depending on the context. Considering this complexity, further investigation of Gal-9's intricate biology is necessary to define therapeutic strategies in immune disorders and cancer treatment aimed at inducing or inhibiting Gal-9 signaling.
Collapse
Affiliation(s)
- Eva M Gossink
- Princess Máxima Center for Pediatric Oncology, 3584CS Utrecht, the Netherlands; Division of Pediatrics, University Medical Center Utrecht, 3584CX Utrecht, the Netherlands; Regenerative Medicine Center, University Medical Center Utrecht, 3584CT Utrecht, the Netherlands
| | - Paul J Coffer
- Division of Pediatrics, University Medical Center Utrecht, 3584CX Utrecht, the Netherlands; Regenerative Medicine Center, University Medical Center Utrecht, 3584CT Utrecht, the Netherlands; Center of Molecular Medicine, University Medical Center Utrecht, 3584CG Utrecht, the Netherlands
| | - Alessandro Cutilli
- Regenerative Medicine Center, University Medical Center Utrecht, 3584CT Utrecht, the Netherlands; Center of Molecular Medicine, University Medical Center Utrecht, 3584CG Utrecht, the Netherlands
| | - Caroline A Lindemans
- Princess Máxima Center for Pediatric Oncology, 3584CS Utrecht, the Netherlands; Division of Pediatrics, University Medical Center Utrecht, 3584CX Utrecht, the Netherlands; Regenerative Medicine Center, University Medical Center Utrecht, 3584CT Utrecht, the Netherlands.
| |
Collapse
|
89
|
Oh T, Woo Y, Kim G, Koo BS, Baek SH, Hwang EH, An YJ, Kim Y, Kim DY, Hong JJ. Spatiotemporal Cellular Dynamics of Germinal Center Reaction in Coronavirus Disease 2019 Lung-Draining Lymph Node Based on Imaging-Based Spatial Transcriptomics. J Transl Med 2025; 105:102180. [PMID: 39522760 DOI: 10.1016/j.labinv.2024.102180] [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: 07/03/2024] [Revised: 10/28/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024] Open
Abstract
Although lymph node structures may be compromised in severe SARS-CoV-2 infection, the extent and parameters of recovery in convalescing patients remain unclear. Therefore, this study aimed to elucidate the nuances of lymphoid structural recovery and their implications for immunologic memory in nonhuman primates infected with SARS-CoV-2. To do so, we utilized imaging-based spatial transcriptomics to delineate immune cell composition and tissue architecture formation in the lung-draining lymph nodes during primary infection, convalescence, and reinfection from COVID-19. We noted the establishment of a germinal center with memory B cell differentiation within lymphoid follicles during convalescence accompanied by contrasting transcriptome patterns indicative of the acquisition of follicular helper T cells versus the loss of regulatory T cells. Additionally, repopulation of germinal center-like B cells was observed in the medullary niche with accumulating plasma cells along with enhanced transcriptional expression of B cell-activating factor receptor over the course of reinfection. The spatial transcriptome atlas produced herein enhances our understanding of germinal center formation with immune cell dynamics during COVID-19 convalescence and lymphoid structural recovery with transcriptome dynamics following reinfection. These findings have the potential to inform the optimization of vaccine strategies and the development of precise therapeutic interventions in the spatial context.
Collapse
Affiliation(s)
- Taehwan Oh
- National Primate Research Centre, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, Chungcheongbuk, Republic of Korea
| | - YoungMin Woo
- National Primate Research Centre, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, Chungcheongbuk, Republic of Korea; KRIBB School of Bioscience, Korea University of Science & Technology (UST), Daejeon, Republic of Korea
| | - Green Kim
- National Primate Research Centre, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, Chungcheongbuk, Republic of Korea
| | - Bon-Sang Koo
- National Primate Research Centre, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, Chungcheongbuk, Republic of Korea
| | - Seung Ho Baek
- National Primate Research Centre, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, Chungcheongbuk, Republic of Korea
| | - Eun-Ha Hwang
- National Primate Research Centre, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, Chungcheongbuk, Republic of Korea
| | - You Jung An
- National Primate Research Centre, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, Chungcheongbuk, Republic of Korea
| | - Yujin Kim
- National Primate Research Centre, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, Chungcheongbuk, Republic of Korea
| | - Dong-Yeon Kim
- National Primate Research Centre, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, Chungcheongbuk, Republic of Korea
| | - Jung Joo Hong
- National Primate Research Centre, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, Chungcheongbuk, Republic of Korea; KRIBB School of Bioscience, Korea University of Science & Technology (UST), Daejeon, Republic of Korea.
| |
Collapse
|
90
|
Lee YS, Cheong MS, Lee J, Bang EK, Park SI, Park HJ, Bae SH, Yoon S, Roh G, Lee S, Cho Y, Ha D, Oh A, Lee SY, Choi EJ, Choi H, Jo S, Lee Y, Kim J, Kwak HW, Bang YJ, Lee D, Shim H, Park YK, Keum G, Nam JH, Kim W. Immunogenicity and protection of a triple repeat domain III mRNA vaccine against Zika virus. Vaccine 2025; 43:126518. [PMID: 39547049 DOI: 10.1016/j.vaccine.2024.126518] [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: 08/20/2024] [Revised: 11/04/2024] [Accepted: 11/05/2024] [Indexed: 11/17/2024]
Abstract
Zika virus (ZIKV) infection is primarily transmitted by mosquitoes and often asymptomatic in most individuals. Infection during pregnancy can lead to severe birth defects such as congenital microcephaly, and currently, there is no approved vaccine for ZIKV. Several studies are investigating the development of ZIKV vaccine using DNA and RNA as well as recombinant protein technologies; however, the outcomes thus far have not been consistently noteworthy. In this study, we designed an mRNA vaccine for ZIKV and evaluated its immunogenicity using a mouse model. Our vaccine, termed 3xEIII, encodes a triple repeat of domain III from the ZIKV E protein. We effectively encapsulated the mRNA within lipid nanoparticles (LNPs), administered 3xEIII to mice via two intramuscular injections, and assessed the induced humoral and cellular immune responses. Specifically, the vaccine elicited neutralizing antibodies that effectively eliminated ZIKV from the organs of challenged mice. Notably, 3xEIII conferred both protective effects and long-term immunity. In subsequent challenges conducted 40 weeks after boosting, immunized mice experienced temporary weight loss but showed significantly reduced viral titers in target organs by the 9th day post-infection. Conclusively from these findings, 3xEIII stands out as a promising noteworthy mRNA vaccine candidate for Zika virus.
Collapse
MESH Headings
- Animals
- Zika Virus Infection/prevention & control
- Zika Virus Infection/immunology
- Zika Virus/immunology
- Zika Virus/genetics
- Mice
- Antibodies, Neutralizing/blood
- Antibodies, Neutralizing/immunology
- Antibodies, Viral/blood
- Antibodies, Viral/immunology
- Viral Vaccines/immunology
- Viral Vaccines/administration & dosage
- Viral Vaccines/genetics
- Female
- mRNA Vaccines
- Nanoparticles
- Disease Models, Animal
- Mice, Inbred BALB C
- Immunity, Humoral
- Immunogenicity, Vaccine
- Immunity, Cellular
- Viral Envelope Proteins/immunology
- Viral Envelope Proteins/genetics
- Vaccines, Synthetic/immunology
- Vaccines, Synthetic/administration & dosage
- Injections, Intramuscular
- RNA, Messenger/genetics
- RNA, Messenger/immunology
- Liposomes
Collapse
Affiliation(s)
- Yu-Sun Lee
- Department of Medical and Biological Sciences, The Catholic University of Korea, Bucheon, Gyeonggi-do 14662, Republic of Korea; BK21 four Department of Biotechnology, The Catholic University of Korea, Gyeonggi-do, Bucheon, Republic of Korea
| | - Mi Sun Cheong
- GeneOne Life Science, Inc., 108 Yeoui-Daero, Yeongdeungpo-gu, Seoul 07335, Republic of Korea
| | - Jisun Lee
- Department of Medical and Biological Sciences, The Catholic University of Korea, Bucheon, Gyeonggi-do 14662, Republic of Korea
| | - Eun-Kyoung Bang
- Center for Brain Technology, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Sang In Park
- Department of Biomedical Laboratory Science, Daegu Haany University, Gyeongsan 38610, Republic of Korea
| | - Hyo-Jung Park
- Department of Medical and Biological Sciences, The Catholic University of Korea, Bucheon, Gyeonggi-do 14662, Republic of Korea; BK21 four Department of Biotechnology, The Catholic University of Korea, Gyeonggi-do, Bucheon, Republic of Korea
| | - Seo-Hyeon Bae
- Department of Medical and Biological Sciences, The Catholic University of Korea, Bucheon, Gyeonggi-do 14662, Republic of Korea; BK21 four Department of Biotechnology, The Catholic University of Korea, Gyeonggi-do, Bucheon, Republic of Korea
| | - Subin Yoon
- Department of Medical and Biological Sciences, The Catholic University of Korea, Bucheon, Gyeonggi-do 14662, Republic of Korea; BK21 four Department of Biotechnology, The Catholic University of Korea, Gyeonggi-do, Bucheon, Republic of Korea
| | - Gahyun Roh
- Department of Medical and Biological Sciences, The Catholic University of Korea, Bucheon, Gyeonggi-do 14662, Republic of Korea
| | - Seonghyun Lee
- Department of Medical and Biological Sciences, The Catholic University of Korea, Bucheon, Gyeonggi-do 14662, Republic of Korea; BK21 four Department of Biotechnology, The Catholic University of Korea, Gyeonggi-do, Bucheon, Republic of Korea
| | - Youngran Cho
- Department of Medical and Biological Sciences, The Catholic University of Korea, Bucheon, Gyeonggi-do 14662, Republic of Korea; BK21 four Department of Biotechnology, The Catholic University of Korea, Gyeonggi-do, Bucheon, Republic of Korea
| | - Dahyeon Ha
- Department of Medical and Biological Sciences, The Catholic University of Korea, Bucheon, Gyeonggi-do 14662, Republic of Korea; BK21 four Department of Biotechnology, The Catholic University of Korea, Gyeonggi-do, Bucheon, Republic of Korea
| | - Ayoung Oh
- Department of Medical and Biological Sciences, The Catholic University of Korea, Bucheon, Gyeonggi-do 14662, Republic of Korea; BK21 four Department of Biotechnology, The Catholic University of Korea, Gyeonggi-do, Bucheon, Republic of Korea
| | - Soo-Yeon Lee
- Department of Medical and Biological Sciences, The Catholic University of Korea, Bucheon, Gyeonggi-do 14662, Republic of Korea; BK21 four Department of Biotechnology, The Catholic University of Korea, Gyeonggi-do, Bucheon, Republic of Korea
| | - Eun-Jin Choi
- Department of Medical and Biological Sciences, The Catholic University of Korea, Bucheon, Gyeonggi-do 14662, Republic of Korea; BK21 four Department of Biotechnology, The Catholic University of Korea, Gyeonggi-do, Bucheon, Republic of Korea
| | - Huijeong Choi
- Department of Medical and Biological Sciences, The Catholic University of Korea, Bucheon, Gyeonggi-do 14662, Republic of Korea; BK21 four Department of Biotechnology, The Catholic University of Korea, Gyeonggi-do, Bucheon, Republic of Korea
| | - Sohee Jo
- Department of Medical and Biological Sciences, The Catholic University of Korea, Bucheon, Gyeonggi-do 14662, Republic of Korea; BK21 four Department of Biotechnology, The Catholic University of Korea, Gyeonggi-do, Bucheon, Republic of Korea
| | - Yeeun Lee
- Department of Medical and Biological Sciences, The Catholic University of Korea, Bucheon, Gyeonggi-do 14662, Republic of Korea; BK21 four Department of Biotechnology, The Catholic University of Korea, Gyeonggi-do, Bucheon, Republic of Korea
| | - Jungmin Kim
- Department of Medical and Biological Sciences, The Catholic University of Korea, Bucheon, Gyeonggi-do 14662, Republic of Korea; BK21 four Department of Biotechnology, The Catholic University of Korea, Gyeonggi-do, Bucheon, Republic of Korea
| | - Hye Won Kwak
- SML Biopharm, Inc., 17 Deokan-ro 104 beon-gil, Gwangmyeong-si, Gyeonggi-do 14353, Republic of Korea
| | - Yoo-Jin Bang
- SML Biopharm, Inc., 17 Deokan-ro 104 beon-gil, Gwangmyeong-si, Gyeonggi-do 14353, Republic of Korea
| | - Dabin Lee
- GeneOne Life Science, Inc., 108 Yeoui-Daero, Yeongdeungpo-gu, Seoul 07335, Republic of Korea
| | - Heeyoun Shim
- GeneOne Life Science, Inc., 108 Yeoui-Daero, Yeongdeungpo-gu, Seoul 07335, Republic of Korea
| | - Young Kun Park
- GeneOne Life Science, Inc., 108 Yeoui-Daero, Yeongdeungpo-gu, Seoul 07335, Republic of Korea
| | - Gyochang Keum
- Center for Brain Technology, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Jae-Hwan Nam
- Department of Medical and Biological Sciences, The Catholic University of Korea, Bucheon, Gyeonggi-do 14662, Republic of Korea; BK21 four Department of Biotechnology, The Catholic University of Korea, Gyeonggi-do, Bucheon, Republic of Korea; SML Biopharm, Inc., 17 Deokan-ro 104 beon-gil, Gwangmyeong-si, Gyeonggi-do 14353, Republic of Korea.
| | - Wonil Kim
- GeneOne Life Science, Inc., 108 Yeoui-Daero, Yeongdeungpo-gu, Seoul 07335, Republic of Korea..
| |
Collapse
|
91
|
Lv X, Zhu L, Feng S, Yang S, Li G, Zhan J, Tan Y, Liu Y, Zhang J, Wang Y, Cheng Y, Fu P, Xu Y, Zheng C. Hsa_circ_0109623 regulates the progression of autoimmune liver disease through Hsa_miR_146b-3p/Sortilin 1-mediated activation of CD4+ T cells. Hepatol Commun 2025; 9:e0607. [PMID: 39774281 PMCID: PMC11717529 DOI: 10.1097/hc9.0000000000000607] [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: 07/31/2024] [Accepted: 10/23/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Autoimmune hepatitis (AIH) is a chronic liver disease characterized by immune-mediated liver inflammation. Despite its global prevalence, the pathogenesis of AIH remains poorly understood, and there is a lack of specific biomarkers and targeted treatments. This study aimed to investigate the role of hsa_circ_0109623, hsa-miR-146b-3p, and Sortilin 1 (SORT1) in AIH and their potential as therapeutic targets. METHODS We collected liver tissue samples and peripheral blood mononuclear cells from patients with AIH and healthy controls and performed RT-PCR, western blotting, flow cytometry, and other molecular biology techniques to analyze the expression of hsa_circ_0109623, hsa-miR-146b-3p, and SORT1. We also used bioinformatics tools to predict the interaction between these molecules and conducted luciferase reporter assays to confirm their binding. RESULTS hsa_circ_0109623 was significantly upregulated in patients with AIH and positively correlated with inflammatory activity. We also found that hsa_circ_0109623 could enhance CD4+ T-cell activation and promote the expression of proinflammatory cytokines. Conversely, hsa-miR-146b-3p was downregulated in patients with AIH and negatively correlated with the expression of hsa_circ_0109623 and SORT1. In addition, hsa-miR-146b-3p acted as a sponge for hsa_circ_0109623, inhibiting CD4+ Th1 cell polarization and cytokine production. SORT1 was also upregulated in patients with AIH and acted as a sponge for hsa-miR-146b-3p, promoting CD4+ Th1 cell polarization and cytokine expression. Furthermore, hsa_miR_146b-3p/SORT1 can regulate the STAT1/STAT4 signaling pathway mediating the progression of AIH. CONCLUSIONS The hsa_circ_0109623/hsa-miR-146b-3p/SORT1 axis plays a crucial role in the pathogenesis of AIH by regulating CD4+ T-cell activation and cytokine production. These molecules may serve as potential biomarkers and therapeutic targets for AIH. Further research is needed to validate these findings and explore their clinical applications.
Collapse
Affiliation(s)
- Xinliang Lv
- Department of Rheumatology, Inner Mongolia Autonomous Region Hospital of Traditional Chinese Medicine, Hohhot, P.R. China
| | - Li Zhu
- Department of Endocrinology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, P.R. China
| | - Shijie Feng
- Department of Rheumatology, Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, P.R. China
| | - Siyu Yang
- Department of Radiology, The Third Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, P.R. China
| | - Guohua Li
- Department of Rheumatology, Inner Mongolia Autonomous Region Hospital of Traditional Chinese Medicine, Hohhot, P.R. China
| | - Jinqin Zhan
- Department of Ultrasound, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, P.R. China
| | - Yuchun Tan
- Department of Anesthesiology, Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, P.R. China
| | - Yuquan Liu
- Department of Rheumatology, Inner Mongolia Autonomous Region Hospital of Traditional Chinese Medicine, Hohhot, P.R. China
| | - Jinliang Zhang
- Department of Rheumatology, Inner Mongolia Autonomous Region Hospital of Traditional Chinese Medicine, Hohhot, P.R. China
| | - Yujin Wang
- Department of Cardiology Department, Chinese People’s Liberation Army General Hospital, Beijing, P.R. China
| | - Yucheng Cheng
- Department of Radiology, The Third Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, P.R. China
| | - Ping Fu
- Department of Rheumatology, Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, P.R. China
| | - Yushan Xu
- Department of Endocrinology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, P.R. China
| | - Chenhong Zheng
- Department of Rheumatology, Inner Mongolia Autonomous Region Hospital of Traditional Chinese Medicine, Hohhot, P.R. China
- Department of Endocrinology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, P.R. China
- Department of Cardiology Department, Chinese People’s Liberation Army General Hospital, Beijing, P.R. China
| |
Collapse
|
92
|
Predescu OR, Dinescu SC, Bita CE, Florescu A, Musetescu AE, Vreju F, Ciurea PL. The immunomodulatory potential of vitamin D on Th17 lymphocytes in systemic lupus erythematosus - a literature review. Med Pharm Rep 2025; 98:13-20. [PMID: 39949914 PMCID: PMC11817589 DOI: 10.15386/mpr-2752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Revised: 10/21/2024] [Accepted: 11/30/2024] [Indexed: 02/16/2025] Open
Abstract
This review offers insight into the complex interplay between cytokines and vitamin D, with focus on its role in systemic lupus erythematosus (SLE) pathogenesis. It offers a helpful resource for researchers and clinicians seeking to better understand and treat SLE and related autoimmune conditions. The pathogenesis of SLE is complex and involves a wide range of cytokines, primarily of the Th2 type; these cytokines mediate hyperactivity in B lymphocytes and antibody production. Notably, vitamin D is found to suppress the activity of critical Th17-related cytokines like IL-23 and IL-6, which is pivotal for Th17 cell development and function. This ultimately leads to reduced IL-17 production, an increase in regulatory T lymphocytes, and subsequent secretion of IL-10. Supplementation with vitamin D is seen to have positive effects on SLE, leading to lower disease activity scores, decreased levels of autoantibodies, and a reduction of fatigue.
Collapse
Affiliation(s)
| | | | - Cristina Elena Bita
- Department of Rheumatology, University of Medicine and Pharmacy of Craiova, Romania
| | - Alesandra Florescu
- Department of Rheumatology, University of Medicine and Pharmacy of Craiova, Romania
| | | | - Florentin Vreju
- Department of Rheumatology, University of Medicine and Pharmacy of Craiova, Romania
| | - Paulina Lucia Ciurea
- Department of Rheumatology, University of Medicine and Pharmacy of Craiova, Romania
| |
Collapse
|
93
|
Zhang L, Yang J, Wang L, Zhang T, Ju F, Wu Y. Perianal abscess complicated by histiocytic necrotizing lymphadenitis postoperatively: a case report. J Surg Case Rep 2025; 2025:rjaf007. [PMID: 39839205 PMCID: PMC11747751 DOI: 10.1093/jscr/rjaf007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 01/07/2025] [Indexed: 01/23/2025] Open
Abstract
Histiocytic necrotizing lymphadenitis (HNL) is a rare condition presenting with lymphadenopathy and fever. We report a case of a 61-year-old male with a history of perianal abscess surgery who developed HNL postoperatively. The patient was treated with corticosteroids, leading to resolution of fever and alleviation of symptoms. This case emphasizes the need for clinical vigilance for HNL in patients with a history of perianal abscess surgery and postoperative fever with lymph node enlargement.
Collapse
Affiliation(s)
- Liman Zhang
- Anorectal Department, Shijiazhuang Traditional Chinese Medical Hospital, 233 Zhongshan Road, Shijiazhuang 050001, China
| | - Jie Yang
- Anorectal Department, Shijiazhuang Traditional Chinese Medical Hospital, 233 Zhongshan Road, Shijiazhuang 050001, China
| | - Lili Wang
- Anorectal Department, Shijiazhuang Traditional Chinese Medical Hospital, 233 Zhongshan Road, Shijiazhuang 050001, China
| | - Tianpeng Zhang
- Anorectal Department, Shijiazhuang Traditional Chinese Medical Hospital, 233 Zhongshan Road, Shijiazhuang 050001, China
| | - Fei Ju
- Anorectal Department, Shijiazhuang Traditional Chinese Medical Hospital, 233 Zhongshan Road, Shijiazhuang 050001, China
| | - Yanjang Wu
- Anorectal Department, Shijiazhuang Traditional Chinese Medical Hospital, 233 Zhongshan Road, Shijiazhuang 050001, China
| |
Collapse
|
94
|
Li W, Liu H, Gao L, Hu Y, Zhang A, Li W, Liu G, Bai W, Xu Y, Xiao C, Deng J, Lei W, Chen G. In-depth human immune cellular profiling from newborn to frail. J Leukoc Biol 2024; 117:qiae046. [PMID: 38447557 DOI: 10.1093/jleuko/qiae046] [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: 12/20/2023] [Revised: 01/20/2024] [Accepted: 02/16/2024] [Indexed: 03/08/2024] Open
Abstract
Immune functional decline and remodeling accompany aging and frailty. It is still largely unknown how changes in the immune cellular composition differentiate healthy individuals from those who become frail at a relatively early age. Our aim in this exploratory study was to investigate immunological changes from newborn to frailty and the association between health statute and various immune cell subtypes. The participants analyzed in this study covered human cord blood cells and peripheral blood cells collected from young adults and healthy and frail old individuals. A total of 30 immune cell subsets were performed by flow cytometry based on the surface markers of immune cells. Furthermore, frailty was investigated for its relations with various leukocyte subpopulations. Frail individuals exhibited a higher CD4/CD8 ratio; a higher proportion of CD4+ central memory T cells, CD8+ effector memory T cells, CD27- switched memory B (BSM) cells, CD27+ BSM cells, age-associated B cells, and CD38-CD24- B cells; and a lower proportion of naïve CD8+ T cells and progenitor B cells. The frailty index score was found to be associated with naïve T cells, CD4/CD8 ratio, age-associated B cells, CD27- BSM cells, and CD4+ central memory T cells. Our findings conducted a relatively comprehensive and extensive atlas of age- and frailty-related changes in peripheral leukocyte subpopulations from newborn to frailty. The immune phenotypes identified in this study can contribute to a deeper understanding of immunosenescence in frailty and may provide a rationale for future interventions and diagnosis.
Collapse
Affiliation(s)
- Wangchun Li
- Intensive Care Unit, Affiliated Shunde Hospital, Jinan University, No.50, East Guizhou Avenue, Foshan 528000, China
| | - Hangyu Liu
- Institute of Geriatric Immunology, Department of Microbiology and Immunology, School of Medicine, Jinan University, No.601, West Huangpu Avenue, Tianhe District, Guangzhou 510632, China
- Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, Jinan University, No.601, West Huangpu Avenue, Tianhe District, Guangzhou 510632, China
| | - Lijuan Gao
- Institute of Geriatric Immunology, Department of Microbiology and Immunology, School of Medicine, Jinan University, No.601, West Huangpu Avenue, Tianhe District, Guangzhou 510632, China
- Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, Jinan University, No.601, West Huangpu Avenue, Tianhe District, Guangzhou 510632, China
| | - Yang Hu
- NHC Key Laboratory of Male Reproduction and Genetics, Guangdong Provincial Reproductive Science Institute, Guangdong Provincial Fertility Hospital, No.17, Meidong Road, Yuexiu District, Guangzhou 510632, China
| | - Anna Zhang
- Intensive Care Unit, Affiliated Shunde Hospital, Jinan University, No.50, East Guizhou Avenue, Foshan 528000, China
| | - Wenfeng Li
- Intensive Care Unit, Affiliated Shunde Hospital, Jinan University, No.50, East Guizhou Avenue, Foshan 528000, China
| | - Guolong Liu
- Department of Medical Oncology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, No.1, Panfu Road, Yuexiu District, Guangzhou 510180, China
| | - Weibin Bai
- Department of Food Science and Engineering, Guangdong Engineering Technology Center of Food Safety Molecular Rapid Detection, Institute of Food Safety and Nutrition, Jinan University, No.601, West Huangpu Avenue, Tianhe District, Guangzhou 510632, China
| | - Yudai Xu
- Institute of Geriatric Immunology, Department of Microbiology and Immunology, School of Medicine, Jinan University, No.601, West Huangpu Avenue, Tianhe District, Guangzhou 510632, China
- Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, Jinan University, No.601, West Huangpu Avenue, Tianhe District, Guangzhou 510632, China
| | - Chanchan Xiao
- Institute of Geriatric Immunology, Department of Microbiology and Immunology, School of Medicine, Jinan University, No.601, West Huangpu Avenue, Tianhe District, Guangzhou 510632, China
- Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, Jinan University, No.601, West Huangpu Avenue, Tianhe District, Guangzhou 510632, China
| | - Jieping Deng
- Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, Jinan University, No.601, West Huangpu Avenue, Tianhe District, Guangzhou 510632, China
- Department of Systems Biomedical Sciences, School of Medicine, Jinan University, No.601, West Huangpu Avenue, Tianhe District, Guangzhou 510632, China
| | - Wen Lei
- Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, Jinan University, No.601, West Huangpu Avenue, Tianhe District, Guangzhou 510632, China
- Integrated Chinese and Western Medicine Postdoctoral Research Station, Guangdong Second Provincial General Hospital, School of Medicine, Jinan University, No.466, Xingang Middle Road, Haizhu District, Guangzhou 510632, China
| | - Guobing Chen
- Institute of Geriatric Immunology, Department of Microbiology and Immunology, School of Medicine, Jinan University, No.601, West Huangpu Avenue, Tianhe District, Guangzhou 510632, China
- Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, Jinan University, No.601, West Huangpu Avenue, Tianhe District, Guangzhou 510632, China
- Key Laboratory of Viral Pathogenesis and Infection Prevention and Control, Jinan University, Ministry of Education, No.601, West Huangpu Avenue, Tianhe District, Guangzhou, 510632, China
| |
Collapse
|
95
|
Tsioulos G, Vallianou NG, Skourtis A, Dalamaga M, Kotsi E, Kargioti S, Adamidis N, Karampela I, Mourouzis I, Kounatidis D. Vaccination as a Promising Approach in Cardiovascular Risk Mitigation: Are We Ready to Embrace a Vaccine Strategy? Biomolecules 2024; 14:1637. [PMID: 39766344 PMCID: PMC11727084 DOI: 10.3390/biom14121637] [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: 11/25/2024] [Revised: 12/15/2024] [Accepted: 12/18/2024] [Indexed: 01/12/2025] Open
Abstract
Cardiovascular disease (CVD) remains a leading global health concern, with atherosclerosis being its principal cause. Standard CVD treatments primarily focus on mitigating cardiovascular (CV) risk factors through lifestyle changes and cholesterol-lowering therapies. As atherosclerosis is marked by chronic arterial inflammation, the innate and adaptive immune systems play vital roles in its progression, either exacerbating or alleviating disease development. This intricate interplay positions the immune system as a compelling therapeutic target. Consequently, immunomodulatory strategies have gained increasing attention, though none have yet reached widespread clinical adoption. Safety concerns, particularly the suppression of host immune defenses, remain a significant barrier to the clinical application of anti-inflammatory therapies. Recent decades have revealed the significant role of adaptive immune responses to plaque-associated autoantigens in atherogenesis, opening new perspectives for targeted immunological interventions. Preclinical models indicate that vaccines targeting specific atherosclerosis-related autoantigens can slow disease progression while preserving systemic immune function. In this context, numerous experimental studies have advanced the understanding of vaccine development by exploring diverse targeting pathways. Key strategies include passive immunization using naturally occurring immunoglobulin G (IgG) antibodies and active immunization targeting low-density lipoprotein cholesterol (LDL-C) and apolipoproteins, such as apolipoprotein B100 (ApoB100) and apolipoprotein CIII (ApoCIII). Other approaches involve vaccine formulations aimed at proteins that regulate lipoprotein metabolism, including proprotein convertase subtilisin/kexin type 9 (PCSK9), cholesteryl ester transfer protein (CETP), and angiopoietin-like protein 3 (ANGPTL3). Furthermore, the literature highlights the potential for developing non-lipid-related vaccines, with key targets including heat shock proteins (HSPs), interleukins (ILs), angiotensin III (Ang III), and a disintegrin and metalloproteinase with thrombospondin motifs 7 (ADAMTS-7). However, translating these promising findings into safe and effective clinical therapies presents substantial challenges. This review provides a critical evaluation of current anti-atherosclerotic vaccination strategies, examines their proposed mechanisms of action, and discusses key challenges that need to be overcome to enable clinical translation.
Collapse
Affiliation(s)
- Georgios Tsioulos
- Fourth Department of Internal Medicine, Medical School, Attikon General University Hospital, National and Kapodistrian University of Athens, 12462 Athens, Greece;
| | - Natalia G. Vallianou
- First Department of Internal Medicine, Sismanogleio General Hospital, 15126 Athens, Greece; (S.K.); (N.A.)
| | - Alexandros Skourtis
- Department of Internal Medicine, Evangelismos General Hospital, 10676 Athens, Greece;
| | - Maria Dalamaga
- Department of Biological Chemistry, National and Kapodistrian University of Athens, 75 Mikras Asias Str., 11527 Athens, Greece;
| | - Evangelia Kotsi
- Second Department of Internal Medicine, Medical School, National and Kapodistrian University of Athens, Hippokratio General Hospital, 11527 Athens, Greece;
| | - Sofia Kargioti
- First Department of Internal Medicine, Sismanogleio General Hospital, 15126 Athens, Greece; (S.K.); (N.A.)
| | - Nikolaos Adamidis
- First Department of Internal Medicine, Sismanogleio General Hospital, 15126 Athens, Greece; (S.K.); (N.A.)
| | - Irene Karampela
- Second Department of Critical Care, Medical School, Attikon General University Hospital, University of Athens, 12461 Athens, Greece;
| | - Iordanis Mourouzis
- Department of Pharmacology, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Dimitris Kounatidis
- Diabetes Center, First Department of Propaedeutic Internal Medicine, Medical School, National and Kapodistrian University of Athens, Laiko General Hospital, 11527 Athens, Greece;
| |
Collapse
|
96
|
Bukhari I, Li M, Li G, Xu J, Zheng P, Chu X. Pinpointing the integration of artificial intelligence in liver cancer immune microenvironment. Front Immunol 2024; 15:1520398. [PMID: 39759506 PMCID: PMC11695355 DOI: 10.3389/fimmu.2024.1520398] [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: 10/31/2024] [Accepted: 12/02/2024] [Indexed: 01/07/2025] Open
Abstract
Liver cancer remains one of the most formidable challenges in modern medicine, characterized by its high incidence and mortality rate. Emerging evidence underscores the critical roles of the immune microenvironment in tumor initiation, development, prognosis, and therapeutic responsiveness. However, the composition of the immune microenvironment of liver cancer (LC-IME) and its association with clinicopathological significance remain unelucidated. In this review, we present the recent developments related to the use of artificial intelligence (AI) for studying the immune microenvironment of liver cancer, focusing on the deciphering of complex high-throughput data. Additionally, we discussed the current challenges of data harmonization and algorithm interpretability for studying LC-IME.
Collapse
Affiliation(s)
- Ihtisham Bukhari
- Department of Oncology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Marshall B. J. Medical Research Center, Zhengzhou University, Zhengzhou, Henan, China
| | - Mengxue Li
- Marshall B. J. Medical Research Center, Zhengzhou University, Zhengzhou, Henan, China
| | - Guangyuan Li
- Department of Oncology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jixuan Xu
- Department of Gastrointestinal & Thyroid Surgery, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Pengyuan Zheng
- Marshall B. J. Medical Research Center, Zhengzhou University, Zhengzhou, Henan, China
| | - Xiufeng Chu
- Department of Oncology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Marshall B. J. Medical Research Center, Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
97
|
Burrello J, Goi J, Burrello A, Vacchi E, Rendon-Angel A, Lazzarini E, Bianco G, Limongelli V, Vassalli G, Cereda CW, Monticone S, Mulatero P, Bussolati B, Alimonti A, Camici GG, Melli G, Osto E, Pedrazzini G, Lucio B. Age- and sex-related variations in extracellular vesicle profiling for the assessment of cardiovascular risk: the EVaging index. NPJ AGING 2024; 10:63. [PMID: 39702460 DOI: 10.1038/s41514-024-00189-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 11/29/2024] [Indexed: 12/21/2024]
Abstract
Extracellular vesicles (EVs) offer valuable diagnostic and prognostic insights for cardiovascular (CV) diseases, but the influence of age-related chronic inflammation ("inflammaging") and sex differences on EV profiles linked to CV risk remains unclear. This study aimed to use EV profiling to predict age and stratify patients by CV risk. We developed an EVaging index by analyzing surface antigen profiles of serum EVs from 625 participants, aged 20 to 94 years, across varying CV risk groups. The EVaging index was associated with age in healthy individuals and distinguished CV risk profiles in patients, correlating with CV outcomes and likelihood of fatal CV events according to the European Society of Cardiology (ESC) SCORE, and reflecting age-associated comorbidities. While changes in disease-related EV fingerprint adds complexity in CV patients, EV profiling may help assess biological aging and CV risk, emphasizing EVs' roles in inflammaging.
Collapse
Affiliation(s)
- Jacopo Burrello
- Cardiovascular Theranostics, Istituto Cardiocentro Ticino, Laboratories for Translational Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
- Division of Internal Medicine and Hypertension Unit, Department of Medical Sciences, University of Torino, Torino, Italy
| | - Jessica Goi
- Division of Internal Medicine and Hypertension Unit, Department of Medical Sciences, University of Torino, Torino, Italy
| | - Alessio Burrello
- Department of Control and Computer Engineering, Politecnico di Torino, Torino, Italy
| | - Elena Vacchi
- Neurodegenerative Diseases Group, Laboratories for Translational Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera italiana (USI), Lugano, Switzerland
| | - Azucena Rendon-Angel
- Cardiovascular Theranostics, Istituto Cardiocentro Ticino, Laboratories for Translational Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera italiana (USI), Lugano, Switzerland
| | - Edoardo Lazzarini
- Cardiovascular Theranostics, Istituto Cardiocentro Ticino, Laboratories for Translational Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
- Euler Institute, Faculty of Biomedical Sciences, Università della Svizzera italiana (USI), Lugano, Switzerland
| | - Giovanni Bianco
- Neurology Department, Neurocenter of Southern Switzerland, Ente Ospedaliero Cantonale, Lugano, Switzerland
| | - Vittorio Limongelli
- Euler Institute, Faculty of Biomedical Sciences, Università della Svizzera italiana (USI), Lugano, Switzerland
| | - Giuseppe Vassalli
- Faculty of Biomedical Sciences, Università della Svizzera italiana (USI), Lugano, Switzerland
- Cellular and Molecular Cardiology Laboratory, Cardiocentro Ticino Institute, Bellinzona, Switzerland
| | - Carlo W Cereda
- Faculty of Biomedical Sciences, Università della Svizzera italiana (USI), Lugano, Switzerland
- Neurology Department, Neurocenter of Southern Switzerland, Ente Ospedaliero Cantonale, Lugano, Switzerland
| | - Silvia Monticone
- Division of Internal Medicine and Hypertension Unit, Department of Medical Sciences, University of Torino, Torino, Italy
| | - Paolo Mulatero
- Division of Internal Medicine and Hypertension Unit, Department of Medical Sciences, University of Torino, Torino, Italy
| | | | - Andrea Alimonti
- Faculty of Biomedical Sciences, Università della Svizzera italiana (USI), Lugano, Switzerland
- Institute of Oncology Research (IOR), Oncology Institute of Southern Switzerland, Bellinzona, Switzerland
- Department of Health Sciences and Technology (D-HEST), ETH Zurich, Zurich, Switzerland
| | - Giovanni G Camici
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
- Department of Research and Education, University Hospital Zurich, Zurich, Switzerland
| | - Giorgia Melli
- Neurodegenerative Diseases Group, Laboratories for Translational Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera italiana (USI), Lugano, Switzerland
- Neurology Department, Neurocenter of Southern Switzerland, Ente Ospedaliero Cantonale, Lugano, Switzerland
| | - Elena Osto
- Division of Physiology and Pathophysiology, Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Medical University of Graz, Graz, Austria
- Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Giovanni Pedrazzini
- Faculty of Biomedical Sciences, Università della Svizzera italiana (USI), Lugano, Switzerland
- Division of Cardiology, Istituto Cardiocentro Ticino, Ente Ospedaliero Cantonale Lugano Switzerland, Zurich, Switzerland
| | - Barile Lucio
- Cardiovascular Theranostics, Istituto Cardiocentro Ticino, Laboratories for Translational Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland.
- Faculty of Biomedical Sciences, Università della Svizzera italiana (USI), Lugano, Switzerland.
- Euler Institute, Faculty of Biomedical Sciences, Università della Svizzera italiana (USI), Lugano, Switzerland.
| |
Collapse
|
98
|
Pitaro M, Antonini G, Arcovito A, Buccisano F, De Lauro A, Irno Consalvo M, Gallo V, Giacon N, Mangiatordi GF, Pacelli M, Pitaro MT, Polticelli F, Sorrenti M, Venditti A. Development of a recombinant human IgG1 monoclonal antibody against the TRBV5-1 segment of the T cell receptor for the treatment of mature T cell neoplasms. Front Immunol 2024; 15:1520103. [PMID: 39742266 PMCID: PMC11686114 DOI: 10.3389/fimmu.2024.1520103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Accepted: 11/21/2024] [Indexed: 01/03/2025] Open
Abstract
Background Mature T-cell neoplasms arise from the neoplastic transformation of a single T lymphocyte, and all cells in a neoplastic clone share the same V segment in the beta chain of the T-cell receptor (TCR). These segments may represent an innovative target for the development of targeted therapies. Methods A specific V segment of the TCR beta chain (TRBV5-1) was analyzed using bioinformatic tools, identifying three potential antigenic peptides. One of these peptides, selected for synthesis, was used to screen a library of human single-chain variable fragments (scFv) through phage display. One fragment demonstrated high affinity and specificity for the antigen and was used to produce a human monoclonal antibody of the IgG1 class. Results Surface plasmon resonance (SPR) studies confirmed the high affinity of the monoclonal antibody for the antigen in the nanomolar range. Flow cytometry analysis on patients' samples demonstrated that the antibody, conjugated with a fluorochrome, selectively binds to tumor T lymphocytes expressing TRBV5-1, without binding to other lymphocytes or blood cell components. Conclusions The development of fully human IgG1 monoclonal antibodies targeting specific V segments of the TCR beta chain represents a potential therapeutic option for patients with mature T-cell neoplasms.
Collapse
Affiliation(s)
- Michele Pitaro
- INBB – Istituto Nazionale Biostrutture e Biosistemi, Rome, Italy
| | - Giovanni Antonini
- INBB – Istituto Nazionale Biostrutture e Biosistemi, Rome, Italy
- Dipartimento di Scienze, Università di Roma Tre, Rome, Italy
| | - Alessandro Arcovito
- Dipartimento di Scienze Biotecnologiche di Base, Cliniche, Intensivologiche e Perioperatorie, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Francesco Buccisano
- Dipartimento di Biomedicina e Prevenzione, Università di Roma Tor Vergata, Rome, Italy
| | | | - Maria Irno Consalvo
- Dipartimento di Biomedicina e Prevenzione, Università di Roma Tor Vergata, Rome, Italy
| | - Valentina Gallo
- Dipartimento di Scienze, Università di Roma Tre, Rome, Italy
| | - Noah Giacon
- Dipartimento di Scienze Biotecnologiche di Base, Cliniche, Intensivologiche e Perioperatorie, Università Cattolica del Sacro Cuore, Rome, Italy
| | | | | | | | | | | | - Adriano Venditti
- Dipartimento di Biomedicina e Prevenzione, Università di Roma Tor Vergata, Rome, Italy
| |
Collapse
|
99
|
Zhang Y, Guo J, Chen Z, Chang Y, Zhang X, Liu Z, Li X, Zha X, Sun G, Li Y. Triclocarban disrupts the activation and differentiation of human CD8 + T cells by suppressing the vitamin D receptor signaling. JOURNAL OF HAZARDOUS MATERIALS 2024; 480:136096. [PMID: 39383692 DOI: 10.1016/j.jhazmat.2024.136096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 10/04/2024] [Accepted: 10/05/2024] [Indexed: 10/11/2024]
Abstract
Triclocarban (TCC) is a widely applied environmental endocrine-disrupting chemical (EDC). Similar to most of EDCs, TCC potentially damages the immunity of various species. However, whether and how TCC impacts the adaptive immunity in mammals has yet to be determined. Herein, we discovered that TCC disrupts the activation and differentiation of CD8+ T cells in primary human peripheral blood samples, purified CD8+ T cells, and in mice in vivo. Mechanistically, TCC might block the activation of the vitamin D receptor (VDR) and reduce the synthesis of cholesterol, a precursor of vitamin D, resulting in inhibition of VDR signaling due to the suppression of both its ligand and the receptor itself by TCC. Our findings elucidate the hazard and potential mechanisms of TCC in mammalian adaptive immunity and highlighted VDR as a potential therapeutic target for the immunodeficiency caused by TCC.
Collapse
Affiliation(s)
- Yikai Zhang
- Guangdong Provincial Key Laboratory of Spine and Spinal Cord Reconstruction, The Fifth Affiliated Hospital of Jinan University (Heyuan Shenhe People's Hospital), Jinan University, Heyuan 517000, China; Department of Hematology, First Affiliated Hospital of Jinan University, Guangzhou 510632, China; Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, Jinan University, Guangzhou 510632, China
| | - Jiafan Guo
- Department of Clinical Laboratory, First Affiliated Hospital of Jinan University, Guangzhou 510632, China
| | - Zhixi Chen
- Department of Hematology, First Affiliated Hospital of Jinan University, Guangzhou 510632, China; Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, Jinan University, Guangzhou 510632, China
| | - Yiming Chang
- Department of Pediatrics, First Affiliated Hospital of Jinan University, Guangzhou 510632, China
| | - Xingwei Zhang
- Department of Clinical Laboratory, First Affiliated Hospital of Jinan University, Guangzhou 510632, China
| | - Zirui Liu
- Department of Hematology, First Affiliated Hospital of Jinan University, Guangzhou 510632, China; Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, Jinan University, Guangzhou 510632, China
| | - Xinye Li
- Department of Hematology, First Affiliated Hospital of Jinan University, Guangzhou 510632, China; Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, Jinan University, Guangzhou 510632, China
| | - Xianfeng Zha
- Department of Clinical Laboratory, First Affiliated Hospital of Jinan University, Guangzhou 510632, China.
| | - Guodong Sun
- Guangdong Provincial Key Laboratory of Spine and Spinal Cord Reconstruction, The Fifth Affiliated Hospital of Jinan University (Heyuan Shenhe People's Hospital), Jinan University, Heyuan 517000, China.
| | - Yangqiu Li
- Department of Hematology, First Affiliated Hospital of Jinan University, Guangzhou 510632, China; Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, Jinan University, Guangzhou 510632, China.
| |
Collapse
|
100
|
Dong L, Qiu X, Li Z, Ge W, Tang X, Zhou R, Chen W, Xu X, Wang K. Potential crosstalk between Naïve CD4 + T cells and SPP1 + Macrophages is associated with clinical outcome and therapeutic response in hepatocellular carcinoma. Int Immunopharmacol 2024; 142:113231. [PMID: 39332093 DOI: 10.1016/j.intimp.2024.113231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 09/06/2024] [Accepted: 09/19/2024] [Indexed: 09/29/2024]
Abstract
BACKGROUND The highly heterogeneity of the tumor microenvironment (TME) in hepatocellular carcinoma (HCC) results in diverse clinical outcomes and therapeutic responses. This study aimed to investigate potential intercellular crosstalk and its impact on clinical outcomes and therapeutic responses. METHODS Single-cell RNA sequencing (scRNA-seq), spatial transcriptomics (ST) and bulk RNA sequencing (RNA-seq) datasets were integrated to comprehensively analyze the intercellular interactions within the TME. Multiplex immunohistochemistry was conducted to validate the intercellular interactions. A machine learning-based integrative procedure was used in bulk RNA-seq datasets to generate a risk model to predict prognosis and therapeutic responses. RESULTS Survival analyses based on the bulk RNA-seq datasets revealed the negative impact of the naïve Cluster of Differentiation 4+ (CD4) T cells and Secreted Phosphoprotein 1+ (SPP1) macrophages on prognosis. Furthermore, their intricate intercellular crosstalk and spatial colocalization were also observed by scRNA-seq and ST analyses. Based on this crosstalk, a machine learning model, termed the naïve CD4+ T cell and SPP1+ macrophage prognostic score (TMPS), was established in the bulk-RNA seq datasets for prognostic prediction. The TMPS achieved C-index values of 0.785, 0.715, 0.692 and 0.857, respectively, across 4 independent cohorts. A low TMPS was associated with a significantly increased survival rates, improved response to immunotherapy and reduced infiltration of immunosuppressive cells, such as. regulatory T cells. Finally, 8 potential sensitive drugs and 6 potential targets were predicted for patients based on their TMPS. CONCLUSION The crosstalk between naïve CD4+ T cells and SPP1+ macrophages play a crucial role in the TME. TMPS can reflect this crosstalk and serve as a valuable tool for prognostic stratification and guiding clinical decision-making.
Collapse
Affiliation(s)
- Libin Dong
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310058, Zhejiang, China
| | - Xun Qiu
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310058, Zhejiang, China
| | - Zekuan Li
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang, China
| | - Wenwen Ge
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310058, Zhejiang, China
| | - Xiao Tang
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310058, Zhejiang, China
| | - Ruhong Zhou
- Institute of Quantitative Biology, Shanghai Institute for Advanced Study, College of Life Sciences, Zhejiang University, Hangzhou 310027, Zhejiang, China
| | - Wei Chen
- Department of Cell Biology, Zhejiang University School of Medicine, Hangzhou 310058, Zhejiang, China
| | - Xiao Xu
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310058, Zhejiang, China; School of Clinical Medicine, Hangzhou Medical College, Hangzhou 310059, Zhejiang, China.
| | - Kai Wang
- School of Clinical Medicine, Hangzhou Medical College, Hangzhou 310059, Zhejiang, China.
| |
Collapse
|