1
|
Sato Y. Immune Aging and Its Implication for Age-Related Disease Progression. Physiology (Bethesda) 2025; 40:0. [PMID: 39887318 DOI: 10.1152/physiol.00051.2024] [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: 11/11/2024] [Revised: 12/18/2024] [Accepted: 01/25/2025] [Indexed: 02/01/2025] Open
Abstract
As life expectancy increases globally, the prevalence and severity of age-related diseases have risen, significantly impacting patients' quality of life and increasing dependency on the healthcare system. Age-related diseases share several pathological commonalities, and emerging evidence suggests that targeting these biological processes ameliorates multiple age-related diseases. Immune aging plays a critical role in the pathogenesis of age-related diseases, given its involvement not only in controlling infection and cancer but also in facilitating tissue homeostasis and repair. Aging causes compositional and functional changes in both innate and adaptive immune cells, thereby significantly contributing to the pathogenesis of age-related disease and systemic low-grade inflammation, termed "inflammaging." This review article aims to describe the current understanding of immune aging and its impact on age-related diseases with particular emphasis on kidney and autoimmune diseases. In addition, this review highlights tertiary lymphoid structures (TLS) as a hallmark of immune aging, exploring their roles in inflammation, tissue damage, and potential therapeutic targeting.
Collapse
Affiliation(s)
- Yuki Sato
- Department of Medicine, Mayo Clinic College of Medicine and Science, Rochester, Minnesota, United States
- Department of Immunology, Mayo Clinic College of Medicine and Science, Rochester, Minnesota, United States
- Department of Nephrology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
2
|
Shi Y, Luo Q, Duan J, Tang B, Guan Q. The rules of different B cell subtypes in colorectal cancer: friends or foes? Future Oncol 2025:1-12. [PMID: 40491002 DOI: 10.1080/14796694.2025.2511588] [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: 11/15/2024] [Accepted: 05/23/2025] [Indexed: 06/11/2025] Open
Abstract
Tumor-infiltrating B cells (TIBs) are the most important cell type involved in the immune response. TIBs display considerable intratumor heterogeneity due to genetic variation, epigenetic differences and transcriptional plasticity in the tumor microenvironment (TME). Owing to the unique anatomical location of CRC, the B cell subpopulation exhibits more extensive heterogeneity. Many studies have shown that TIBs have gradually become a key predictor of immunotherapy for malignant cancers, including CRC. TIBs have essential functions, including antigen presentation and antibody secretion, and they promote T-cell activation and myeloid chemotaxis. However, owing to the complex TME, TIBs not only promote the antitumor immune response but also inhibit the immune response. With the in-depth study of tumor-infiltrating T cells, tumor-associated myeloid cells and the interactions among these cells in the TME, the special role of immune cells in the TME has gradually become clear. However, the influence of TIBs in the TME and their interactions with nonimmune cells in the TME remain unclear. Here, we summarize the current progress in TIBs based on single-cell RNA sequencing in CRC in recent years, focusing on specific effector or regulatory characteristics of different B cell subclusters in the CRC TME.
Collapse
Affiliation(s)
- Yuanchao Shi
- The First Clinical Academy of Lanzhou University, Lanzhou University, Lanzhou, Gansu, China
- Department of General Surgery and Gastrointestinal Oncology Surgery, Lanzhou University First Hospital, Lanzhou, Gansu, China
| | - Qianwen Luo
- The First Clinical Academy of Lanzhou University, Lanzhou University, Lanzhou, Gansu, China
- Department of General Surgery and Gastrointestinal Oncology Surgery, Lanzhou University First Hospital, Lanzhou, Gansu, China
| | - Jingwei Duan
- Emergency Department, Peking University Third Hospital, Beijing, China
| | - Bo Tang
- Department of General Surgery and Center of Minimal Invasive Gastrointestinal Surgery, Third Military Medical University Southwest Hospital, Chongqing, China
| | - Quanlin Guan
- The First Clinical Academy of Lanzhou University, Lanzhou University, Lanzhou, Gansu, China
- Department of General Surgery and Gastrointestinal Oncology Surgery, Lanzhou University First Hospital, Lanzhou, Gansu, China
| |
Collapse
|
3
|
Lu Z, Du W, Jiao X, Wang Y, Shi J, Shi Y, Shu Y, Niu Z, Hara H, Wu J, Hsu CH, Van Cutsem E, Brock MV, Zhang Z, Ding N, Zhang Y, Shen Z, Shen L. NOTCH1 Mutation and Survival Analysis of Tislelizumab in Advanced or Metastatic Esophageal Squamous Cell Carcinoma: A Biomarker Analysis From the Randomized, Phase III, RATIONALE-302 Trial. J Clin Oncol 2025; 43:1898-1909. [PMID: 40179324 PMCID: PMC12118624 DOI: 10.1200/jco-24-01818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 01/07/2025] [Accepted: 02/04/2025] [Indexed: 04/05/2025] Open
Abstract
PURPOSE Although multiple agents targeting PD-1 have been approved as second-line treatment for esophageal squamous cell carcinoma (ESCC), only a fraction of patients derive long-term survival. Hence, reliable predictive biomarkers are urgently needed. METHODS Comprehensive tumor genomic profiling and transcriptome sequencing were performed on samples from the RATIONALE-302 study. We also conducted single-cell RNA sequencing analysis on Notch1 knockdown ESCC murine models to further explore the potential molecular mechanisms underlying anti-PD-1 benefit. RESULTS We identified NOTCH1 mutation as a potential predictive biomarker for longer overall survival (OS) with tislelizumab versus chemotherapy (18.4 months v 5.3 months; hazard ratio, 0.35 [95% CI, 0.17 to 0.71]). At the transcriptional level, type I IFN (IFN-I)/toll-like receptor expression signatures were positively associated with OS benefit of tislelizumab, whereas B-cell and neutrophil signatures predicted unfavorable OS. Exploratory analyses showed that the presence of NOTCH1 mutation correlated with enrichment of IFN-I signatures and reduced infiltration of B cells and neutrophils. In murine models, comparative single-cell transcriptome analyses further revealed that Notch1 deficiency facilitated a more immunologically activated tumor microenvironment which potentiated anti-PD-1 treatment. CONCLUSION Our data provide novel insights for anti-PD-1 treatment selection using NOTCH1 mutations and may provide a rationale for combination therapy in ESCC.
Collapse
Affiliation(s)
- Zhihao Lu
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, China
| | - Wenting Du
- Clinical Biomarker, BeiGene (Shanghai) Co, Ltd, Shanghai, China
| | - Xi Jiao
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, China
| | - Yanni Wang
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, China
| | - Jingwen Shi
- Clinical Biomarker, BeiGene (Beijing) Co, Ltd, Beijing, China
| | - Yang Shi
- Clinical Biomarker, BeiGene (Beijing) Co, Ltd, Beijing, China
| | - Yongqian Shu
- The First Affiliated Hospital of Nanjing Medical University (Jiangsu Province Hospital)—Cancer Center, Nanjing, China
| | - Zuoxing Niu
- Shandong Cancer Hospital—Oncology, Jinan, China
| | - Hiroki Hara
- Saitama Cancer Center—Gastroenterology, Kitaadachi-gun, Japan
| | - Jun Wu
- The First People's Hospital of Changzhou—Oncology, Changzhou, China
| | - Chih-Hung Hsu
- National Taiwan University Hospital, Taipei, Republic of China
| | - Eric Van Cutsem
- University Hospitals Gasthuisberg/Leuven & KU Leuven, Leuven, Belgium
| | - Malcolm V. Brock
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Zhang Zhang
- Statistics, BeiGene (Beijing) Co, Ltd, Beijing, China
| | - Ningning Ding
- Clinical Development, BeiGene (Beijing) Co, Ltd, Beijing, China
| | - Yun Zhang
- Clinical Biomarker, BeiGene (Beijing) Co, Ltd, Beijing, China
| | - Zhirong Shen
- Clinical Biomarker, BeiGene (Beijing) Co, Ltd, Beijing, China
| | - Lin Shen
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, China
- Department of Gastrointestinal Oncology, State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Peking University Cancer Hospital & Institute, Beijing, China
| |
Collapse
|
4
|
Zheng P, Xiao Y, Wu Z, Wang Q, Lv Y, Niu W, Zhang Y, Xiao J, Cao J, Li M, Lei P, Huang T. Tumor-secreted galectin-3 suppresses antitumor response by inducing IL-10 + B cells. J Immunother Cancer 2025; 13:e011445. [PMID: 40449954 DOI: 10.1136/jitc-2024-011445] [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] [Accepted: 05/17/2025] [Indexed: 06/03/2025] Open
Abstract
BACKGROUND Breast cancer is the leading cause of cancer-related deaths among women globally. Tumor-secreted proteins foster an immunosuppressive environment, enabling immune evasion and reducing the effectiveness of antitumor immunity. Interleukin (IL)-10, an immune-regulatory cytokine, is upregulated in breast cancer (BRCA), but the underlying mechanism remains unclear. METHODS In silico analysis was used to identify candidate genes associated with IL-10 expression. Mouse model of BRCA and in vitro co-culture assays were conducted to identify immune cells responsible for IL-10 upregulation. The efficacy of galectin-3 (Gal-3) inhibition combined with chemotherapy was also evaluated. RESULTS Tumor-secreted Gal-3 served as a key immunosuppressive factor in BRCA. Gal-3 promoted IL-10 production in B cells, thus impairing antitumor immunity. Mechanistically, Gal-3 triggered CD45 polymerization, reducing its phosphatase activity and subsequently activating STAT3 to promote IL-10 production. Knockdown of STAT3 or its blockade abrogated Gal-3-induced IL-10 upregulation. Furthermore, Gal-3 inhibition combined with chemotherapy significantly reduced tumor growth and enhanced antitumor immunity. CONCLUSIONS Gal-3 is a crucial regulator of immune evasion in BRCA. Targeting Gal-3 may improve the efficacy of BRCA therapies.
Collapse
Affiliation(s)
- Peng Zheng
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yunxiao Xiao
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Zhenghao Wu
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Qianheng Wang
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yibing Lv
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Wenhao Niu
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yirui Zhang
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Junyi Xiao
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jing Cao
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Mingxuan Li
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Ping Lei
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Tao Huang
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
5
|
Zheng Y, Cao Y, Wang W, Tong Y, Wang S, Li C, Zhao M, Song Y, Wang YGS, Qi J, Wu C, Yang J, Zheng J, Gao J, Wang J, Yang Q, Liu G, Zhao J, Zhang Y, Xiao H, Zhang YY, Tang YD. Dusp14-Mediated Dephosphorylation of MLKL Protects Against Cardiomyocyte Necroptosis in Hypothyroidism-Induced Heart Failure. Circulation 2025. [PMID: 40357546 DOI: 10.1161/circulationaha.125.074353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Accepted: 04/03/2025] [Indexed: 05/15/2025]
Abstract
BACKGROUND Hypothyroidism leads to multiple organ dysfunction, with the heart the most affected. However, the pathologic mechanism of hypothyroidism-induced heart failure remains to be completely elucidated. Thyroid hormone replacement therapy enhances myocardium systolic function but increases the occurrence of arrythmias. There is an urgent need to explore these mechanisms in detail and to discover and develop drugs that can target and manage heart failure in patients with hypothyroidism. METHODS In this study, a mouse model of hypothyroidism-induced heart failure was established through the administration of propylthiouracil. Dusp14 knockout mice were generated, and adeno-associated virus-mediated cardiomyocyte-specific overexpression of Dusp14 (dual specificity phosphatase 14) was used in combination with related cellular experiments to investigate the protective effects of Dusp14 on hypothyroidism-induced heart failure. Further analyses confirmed the crucial involvement of necroptosis in the pathogenesis of hypothyroidism-induced heart failure, and demonstrated the protective role of Dusp14 in modulating necroptosis. In addition, a novel small molecule compound that effectively regulates Dusp14 activity in vitro was identified through molecular docking, providing a potential therapeutic avenue. RESULTS Dusp14 regulates necroptosis and mitigates hypothyroidism-induced heart failure. Myocardial tissue sections from mice in the hypothyroidism group showed positive Evans blue dye staining, and the serum levels of the myocardial injury marker lactate dehydrogenase were significantly higher compared with the euthyroid group (n=8). In addition, phosphorylation levels of the necroptosis marker MLKL (mixed lineage kinase domain-like protein) were significantly elevated, indicating the activation of necroptosis (n=8). These findings suggest that myocardial necroptosis is activated during hypothyroidism. Myocardial-specific overexpression of Dusp14 reduced myocardial necroptosis and improved myocardial contractile function in hypothyroid mice (n=8). In contrast, Dusp14 knockout exacerbated myocardial contractile dysfunction and necroptosis in these mice (n=5-7). These results indicate that Dusp14 alleviates hypothyroidism-induced heart failure by inhibiting necroptosis. P077-0472, a small molecule compound, was identified as an activator of Dusp14, which could inhibit cardiomyocyte necroptosis from hypothyroidism (n=6). CONCLUSIONS Dusp14 inhibits cardiomyocyte necroptosis from hypothyroidism and consequently rescues damaged cardiomyocytes. P077-0472, a novel small molecule compound that activates the dephosphorylation function of Dusp14, could inhibit cardiomyocyte necroptosis from hypothyroidism.
Collapse
Affiliation(s)
- Yitian Zheng
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China (Y. Zheng, Y.C., W.W., Y.T., C.L., M.Z., Y.S., Y.-g.-s.W., J.Q., C.W., J.Y., J. Zheng, J.G., J.W., Y. Zhang, H.X., Y.-Y.Z., Y.-D.T.)
- Beijing Key Laboratory of Clinical Evaluation of Cardiovascular-Kidney-Metabolic and Immuno-Inflammatory Innovative Drugs and Medical Devices, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China (Y. Zheng, Y.C., W.W., Y.T., C.L., M.Z., Y.S., Y.-g.-s.W., J.Q., C.W., J.Y., J. Zheng, J.G., J.W., Y. Zhang, H.X., Y.-Y.Z., Y.-D.T.)
- Research Unit of Medical Science Research Management/Basic and Clinical Research of Metabolic Cardiovascular Diseases, Chinese Academy of Medical Sciences, Beijing, China (Y. Zheng, W.W., H.X., Y.-Y.Z., Y.-D.T.)
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (Y. Zheng)
| | - Yueyue Cao
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China (Y. Zheng, Y.C., W.W., Y.T., C.L., M.Z., Y.S., Y.-g.-s.W., J.Q., C.W., J.Y., J. Zheng, J.G., J.W., Y. Zhang, H.X., Y.-Y.Z., Y.-D.T.)
- Beijing Key Laboratory of Clinical Evaluation of Cardiovascular-Kidney-Metabolic and Immuno-Inflammatory Innovative Drugs and Medical Devices, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China (Y. Zheng, Y.C., W.W., Y.T., C.L., M.Z., Y.S., Y.-g.-s.W., J.Q., C.W., J.Y., J. Zheng, J.G., J.W., Y. Zhang, H.X., Y.-Y.Z., Y.-D.T.)
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China (Y.C.)
| | - Wenyao Wang
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China (Y. Zheng, Y.C., W.W., Y.T., C.L., M.Z., Y.S., Y.-g.-s.W., J.Q., C.W., J.Y., J. Zheng, J.G., J.W., Y. Zhang, H.X., Y.-Y.Z., Y.-D.T.)
- Beijing Key Laboratory of Clinical Evaluation of Cardiovascular-Kidney-Metabolic and Immuno-Inflammatory Innovative Drugs and Medical Devices, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China (Y. Zheng, Y.C., W.W., Y.T., C.L., M.Z., Y.S., Y.-g.-s.W., J.Q., C.W., J.Y., J. Zheng, J.G., J.W., Y. Zhang, H.X., Y.-Y.Z., Y.-D.T.)
- Research Unit of Medical Science Research Management/Basic and Clinical Research of Metabolic Cardiovascular Diseases, Chinese Academy of Medical Sciences, Beijing, China (Y. Zheng, W.W., H.X., Y.-Y.Z., Y.-D.T.)
| | - Yicheng Tong
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China (Y. Zheng, Y.C., W.W., Y.T., C.L., M.Z., Y.S., Y.-g.-s.W., J.Q., C.W., J.Y., J. Zheng, J.G., J.W., Y. Zhang, H.X., Y.-Y.Z., Y.-D.T.)
- Beijing Key Laboratory of Clinical Evaluation of Cardiovascular-Kidney-Metabolic and Immuno-Inflammatory Innovative Drugs and Medical Devices, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China (Y. Zheng, Y.C., W.W., Y.T., C.L., M.Z., Y.S., Y.-g.-s.W., J.Q., C.W., J.Y., J. Zheng, J.G., J.W., Y. Zhang, H.X., Y.-Y.Z., Y.-D.T.)
| | - Shuaixing Wang
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China (S.W.)
| | - Chen Li
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China (Y. Zheng, Y.C., W.W., Y.T., C.L., M.Z., Y.S., Y.-g.-s.W., J.Q., C.W., J.Y., J. Zheng, J.G., J.W., Y. Zhang, H.X., Y.-Y.Z., Y.-D.T.)
- Beijing Key Laboratory of Clinical Evaluation of Cardiovascular-Kidney-Metabolic and Immuno-Inflammatory Innovative Drugs and Medical Devices, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China (Y. Zheng, Y.C., W.W., Y.T., C.L., M.Z., Y.S., Y.-g.-s.W., J.Q., C.W., J.Y., J. Zheng, J.G., J.W., Y. Zhang, H.X., Y.-Y.Z., Y.-D.T.)
| | - Mingming Zhao
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China (Y. Zheng, Y.C., W.W., Y.T., C.L., M.Z., Y.S., Y.-g.-s.W., J.Q., C.W., J.Y., J. Zheng, J.G., J.W., Y. Zhang, H.X., Y.-Y.Z., Y.-D.T.)
- Beijing Key Laboratory of Clinical Evaluation of Cardiovascular-Kidney-Metabolic and Immuno-Inflammatory Innovative Drugs and Medical Devices, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China (Y. Zheng, Y.C., W.W., Y.T., C.L., M.Z., Y.S., Y.-g.-s.W., J.Q., C.W., J.Y., J. Zheng, J.G., J.W., Y. Zhang, H.X., Y.-Y.Z., Y.-D.T.)
| | - Yao Song
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China (Y. Zheng, Y.C., W.W., Y.T., C.L., M.Z., Y.S., Y.-g.-s.W., J.Q., C.W., J.Y., J. Zheng, J.G., J.W., Y. Zhang, H.X., Y.-Y.Z., Y.-D.T.)
- Beijing Key Laboratory of Clinical Evaluation of Cardiovascular-Kidney-Metabolic and Immuno-Inflammatory Innovative Drugs and Medical Devices, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China (Y. Zheng, Y.C., W.W., Y.T., C.L., M.Z., Y.S., Y.-g.-s.W., J.Q., C.W., J.Y., J. Zheng, J.G., J.W., Y. Zhang, H.X., Y.-Y.Z., Y.-D.T.)
| | - Yuan-Geng-Shuo Wang
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China (Y. Zheng, Y.C., W.W., Y.T., C.L., M.Z., Y.S., Y.-g.-s.W., J.Q., C.W., J.Y., J. Zheng, J.G., J.W., Y. Zhang, H.X., Y.-Y.Z., Y.-D.T.)
- Beijing Key Laboratory of Clinical Evaluation of Cardiovascular-Kidney-Metabolic and Immuno-Inflammatory Innovative Drugs and Medical Devices, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China (Y. Zheng, Y.C., W.W., Y.T., C.L., M.Z., Y.S., Y.-g.-s.W., J.Q., C.W., J.Y., J. Zheng, J.G., J.W., Y. Zhang, H.X., Y.-Y.Z., Y.-D.T.)
| | - Jiating Qi
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China (Y. Zheng, Y.C., W.W., Y.T., C.L., M.Z., Y.S., Y.-g.-s.W., J.Q., C.W., J.Y., J. Zheng, J.G., J.W., Y. Zhang, H.X., Y.-Y.Z., Y.-D.T.)
- Beijing Key Laboratory of Clinical Evaluation of Cardiovascular-Kidney-Metabolic and Immuno-Inflammatory Innovative Drugs and Medical Devices, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China (Y. Zheng, Y.C., W.W., Y.T., C.L., M.Z., Y.S., Y.-g.-s.W., J.Q., C.W., J.Y., J. Zheng, J.G., J.W., Y. Zhang, H.X., Y.-Y.Z., Y.-D.T.)
| | - Chao Wu
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China (Y. Zheng, Y.C., W.W., Y.T., C.L., M.Z., Y.S., Y.-g.-s.W., J.Q., C.W., J.Y., J. Zheng, J.G., J.W., Y. Zhang, H.X., Y.-Y.Z., Y.-D.T.)
- Beijing Key Laboratory of Clinical Evaluation of Cardiovascular-Kidney-Metabolic and Immuno-Inflammatory Innovative Drugs and Medical Devices, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China (Y. Zheng, Y.C., W.W., Y.T., C.L., M.Z., Y.S., Y.-g.-s.W., J.Q., C.W., J.Y., J. Zheng, J.G., J.W., Y. Zhang, H.X., Y.-Y.Z., Y.-D.T.)
| | - Jie Yang
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China (Y. Zheng, Y.C., W.W., Y.T., C.L., M.Z., Y.S., Y.-g.-s.W., J.Q., C.W., J.Y., J. Zheng, J.G., J.W., Y. Zhang, H.X., Y.-Y.Z., Y.-D.T.)
- Beijing Key Laboratory of Clinical Evaluation of Cardiovascular-Kidney-Metabolic and Immuno-Inflammatory Innovative Drugs and Medical Devices, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China (Y. Zheng, Y.C., W.W., Y.T., C.L., M.Z., Y.S., Y.-g.-s.W., J.Q., C.W., J.Y., J. Zheng, J.G., J.W., Y. Zhang, H.X., Y.-Y.Z., Y.-D.T.)
| | - Jilin Zheng
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China (Y. Zheng, Y.C., W.W., Y.T., C.L., M.Z., Y.S., Y.-g.-s.W., J.Q., C.W., J.Y., J. Zheng, J.G., J.W., Y. Zhang, H.X., Y.-Y.Z., Y.-D.T.)
- Beijing Key Laboratory of Clinical Evaluation of Cardiovascular-Kidney-Metabolic and Immuno-Inflammatory Innovative Drugs and Medical Devices, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China (Y. Zheng, Y.C., W.W., Y.T., C.L., M.Z., Y.S., Y.-g.-s.W., J.Q., C.W., J.Y., J. Zheng, J.G., J.W., Y. Zhang, H.X., Y.-Y.Z., Y.-D.T.)
| | - Jun Gao
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China (Y. Zheng, Y.C., W.W., Y.T., C.L., M.Z., Y.S., Y.-g.-s.W., J.Q., C.W., J.Y., J. Zheng, J.G., J.W., Y. Zhang, H.X., Y.-Y.Z., Y.-D.T.)
- Beijing Key Laboratory of Clinical Evaluation of Cardiovascular-Kidney-Metabolic and Immuno-Inflammatory Innovative Drugs and Medical Devices, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China (Y. Zheng, Y.C., W.W., Y.T., C.L., M.Z., Y.S., Y.-g.-s.W., J.Q., C.W., J.Y., J. Zheng, J.G., J.W., Y. Zhang, H.X., Y.-Y.Z., Y.-D.T.)
| | - Jingjia Wang
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China (Y. Zheng, Y.C., W.W., Y.T., C.L., M.Z., Y.S., Y.-g.-s.W., J.Q., C.W., J.Y., J. Zheng, J.G., J.W., Y. Zhang, H.X., Y.-Y.Z., Y.-D.T.)
- Beijing Key Laboratory of Clinical Evaluation of Cardiovascular-Kidney-Metabolic and Immuno-Inflammatory Innovative Drugs and Medical Devices, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China (Y. Zheng, Y.C., W.W., Y.T., C.L., M.Z., Y.S., Y.-g.-s.W., J.Q., C.W., J.Y., J. Zheng, J.G., J.W., Y. Zhang, H.X., Y.-Y.Z., Y.-D.T.)
| | - Qing Yang
- Department of Cardiology, Tianjin Medical University General Hospital, China (Q.Y.)
| | - Gang Liu
- Department of Cardiology, the First Hospital of Hebei Medical University, Shijiazhuang, Hebei, China (G.L.)
| | - Jiajun Zhao
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China (J. Zhao)
| | - Yan Zhang
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China (Y. Zheng, Y.C., W.W., Y.T., C.L., M.Z., Y.S., Y.-g.-s.W., J.Q., C.W., J.Y., J. Zheng, J.G., J.W., Y. Zhang, H.X., Y.-Y.Z., Y.-D.T.)
- Beijing Key Laboratory of Clinical Evaluation of Cardiovascular-Kidney-Metabolic and Immuno-Inflammatory Innovative Drugs and Medical Devices, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China (Y. Zheng, Y.C., W.W., Y.T., C.L., M.Z., Y.S., Y.-g.-s.W., J.Q., C.W., J.Y., J. Zheng, J.G., J.W., Y. Zhang, H.X., Y.-Y.Z., Y.-D.T.)
| | - Han Xiao
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China (Y. Zheng, Y.C., W.W., Y.T., C.L., M.Z., Y.S., Y.-g.-s.W., J.Q., C.W., J.Y., J. Zheng, J.G., J.W., Y. Zhang, H.X., Y.-Y.Z., Y.-D.T.)
- Beijing Key Laboratory of Clinical Evaluation of Cardiovascular-Kidney-Metabolic and Immuno-Inflammatory Innovative Drugs and Medical Devices, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China (Y. Zheng, Y.C., W.W., Y.T., C.L., M.Z., Y.S., Y.-g.-s.W., J.Q., C.W., J.Y., J. Zheng, J.G., J.W., Y. Zhang, H.X., Y.-Y.Z., Y.-D.T.)
- Research Unit of Medical Science Research Management/Basic and Clinical Research of Metabolic Cardiovascular Diseases, Chinese Academy of Medical Sciences, Beijing, China (Y. Zheng, W.W., H.X., Y.-Y.Z., Y.-D.T.)
| | - You-Yi Zhang
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China (Y. Zheng, Y.C., W.W., Y.T., C.L., M.Z., Y.S., Y.-g.-s.W., J.Q., C.W., J.Y., J. Zheng, J.G., J.W., Y. Zhang, H.X., Y.-Y.Z., Y.-D.T.)
- Beijing Key Laboratory of Clinical Evaluation of Cardiovascular-Kidney-Metabolic and Immuno-Inflammatory Innovative Drugs and Medical Devices, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China (Y. Zheng, Y.C., W.W., Y.T., C.L., M.Z., Y.S., Y.-g.-s.W., J.Q., C.W., J.Y., J. Zheng, J.G., J.W., Y. Zhang, H.X., Y.-Y.Z., Y.-D.T.)
- Research Unit of Medical Science Research Management/Basic and Clinical Research of Metabolic Cardiovascular Diseases, Chinese Academy of Medical Sciences, Beijing, China (Y. Zheng, W.W., H.X., Y.-Y.Z., Y.-D.T.)
| | - Yi-Da Tang
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China (Y. Zheng, Y.C., W.W., Y.T., C.L., M.Z., Y.S., Y.-g.-s.W., J.Q., C.W., J.Y., J. Zheng, J.G., J.W., Y. Zhang, H.X., Y.-Y.Z., Y.-D.T.)
- Beijing Key Laboratory of Clinical Evaluation of Cardiovascular-Kidney-Metabolic and Immuno-Inflammatory Innovative Drugs and Medical Devices, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China (Y. Zheng, Y.C., W.W., Y.T., C.L., M.Z., Y.S., Y.-g.-s.W., J.Q., C.W., J.Y., J. Zheng, J.G., J.W., Y. Zhang, H.X., Y.-Y.Z., Y.-D.T.)
- Research Unit of Medical Science Research Management/Basic and Clinical Research of Metabolic Cardiovascular Diseases, Chinese Academy of Medical Sciences, Beijing, China (Y. Zheng, W.W., H.X., Y.-Y.Z., Y.-D.T.)
- University of Health and Rehabilitation Sciences, Qingdao, China (Y.-D.T.)
| |
Collapse
|
6
|
Gracie CJ, Mitchell R, Johnstone JC, Clarke AJ. The unusual metabolism of germinal center B cells. Trends Immunol 2025; 46:416-428. [PMID: 40221291 DOI: 10.1016/j.it.2025.02.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 02/21/2025] [Accepted: 02/27/2025] [Indexed: 04/14/2025]
Abstract
In the germinal center (GC), B cells undergo rounds of somatic hypermutation (SHM), proliferation, and positive selection to develop into high-affinity, long-lived plasma cells and memory B cells. It is well established that, upon activation, B cells significantly alter their metabolism, but until recently little was understood about their metabolism within the GC. In this review we discuss novel in vivo models in which GC B cell (GCBC) metabolism is disrupted; these have greatly increased our understanding of B cell metabolic phenotype. GCBCs are unusual in that, unlike almost all other rapidly proliferating immune cells, they use little glycolysis but prefer fatty acid oxidation (FAO) to fuel ATP synthesis, whilst preferentially utilizing glucose and amino acids as carbon and nitrogen sources for biosynthetic pathways.
Collapse
Affiliation(s)
- Caitlin J Gracie
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Robert Mitchell
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | | | | |
Collapse
|
7
|
Lai Z, Kong D, Li Q, Wang Y, Li K, Duan X, Shao J, Xie Y, Chen J, Zhang T, Feng Y, Deng H, Wang J, Wang C, Shu K, Zhao H, Du H, Jia C, Dai H, Xie L, Liu J, Luo X, Wang L, Xu L, Zhu Z, Lei X, Wang Y, Yang Y, Liu Y, Liang Y, Yang Y, Xie J, Liu B, Deng Z, Liu X. Single-cell spatial transcriptomics of tertiary lymphoid organ-like structures in human atherosclerotic plaques. NATURE CARDIOVASCULAR RESEARCH 2025; 4:547-566. [PMID: 40295810 DOI: 10.1038/s44161-025-00639-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 03/20/2025] [Indexed: 04/30/2025]
Abstract
Tertiary lymphoid organs have been identified in the arterial adventitia in both mouse models of atherosclerosis and patients with atherosclerosis, yet their role in the disease remains insufficiently explored. Here we present a spatially resolved single-cell transcriptome atlas of human atherosclerotic plaques, identifying 14 distinct cell types and providing evidence of plaque tertiary lymphoid organs (PTLOs). The development of PTLOs was associated with the expression of lymphangiogenic chemokine genes and the adhesion molecule gene in fibroblast-like smooth muscle cells. PTLOs harbor abundant B cells with expanded and diversified B cell receptors, suggesting substantial immune involvement. We also observed that B cells may be exchanged between PTLOs and perivascular adipose tissues. The presence of PTLO-like structures correlates with cerebrovascular events, which may be mediated by PTLO-derived IgG antibodies enhancing macrophage functional activity. Our findings suggest the existence and characteristics of PTLOs in human atherosclerosis, elucidating their cellular functions and clinical implications and offering avenues for understanding, diagnosing and treating this condition.
Collapse
Affiliation(s)
- Zhichao Lai
- Department of Vascular Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Deqiang Kong
- Department of Vascular Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | | | - Yue Wang
- BGI Research, Beijing, China
- Shanxi Medical University-BGI Collaborative Center for Future Medicine, Shanxi Medical University, Taiyuan, China
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Kang Li
- Department of Vascular Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Xiaohan Duan
- BGI Research, Beijing, China
- 4+4 Medical Doctor Program, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jiang Shao
- Department of Vascular Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Yiyun Xie
- Department of Vascular Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Junye Chen
- Department of Vascular Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Department of Pathophysiology, Peking Union Medical College, Beijing, China
| | - Tianjing Zhang
- Department of Vascular Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Yuyao Feng
- Department of Vascular Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | | | - Jiaxian Wang
- Department of Vascular Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Chaonan Wang
- Department of Hemangiomas and Vascular Malformations, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Keqiang Shu
- Department of Vascular Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Hongmei Zhao
- 4+4 Medical Doctor Program, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hanze Du
- Department of Endocrinology, Key Laboratory of Endocrinology of National Health Commission, Translation Medicine Centre, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Congwei Jia
- Department of Pathology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Huanyu Dai
- Department of Vascular Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Lizhi Xie
- Department of Vascular Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | | | | | - Lin Wang
- 4+4 Medical Doctor Program, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Leyin Xu
- Department of Vascular Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Zhan Zhu
- Department of Vascular Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Xiangling Lei
- Department of Vascular Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Yuru Wang
- Department of Vascular Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Yixuan Yang
- Department of Vascular Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Yanan Liu
- Department of Vascular Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | | | | | - Jun Xie
- Shanxi Medical University-BGI Collaborative Center for Future Medicine, Shanxi Medical University, Taiyuan, China
| | - Bao Liu
- Department of Vascular Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China.
| | | | - Xin Liu
- BGI Research, Beijing, China.
- Shanxi Medical University-BGI Collaborative Center for Future Medicine, Shanxi Medical University, Taiyuan, China.
| |
Collapse
|
8
|
Bindu S, Bibi R, Pradeep R, Sarkar K. The evolving role of B cells in malignancies. Hum Immunol 2025; 86:111301. [PMID: 40132250 DOI: 10.1016/j.humimm.2025.111301] [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: 08/06/2024] [Revised: 03/07/2025] [Accepted: 03/19/2025] [Indexed: 03/27/2025]
Abstract
B cells play diverse roles in different pathological circumstances, such as neoplastic diseases, autoimmune disorders, and neurological maladies. B cells, which are essential elements of the adaptive immune system, demonstrate exceptional functional variety, including the generation of antibodies, the presentation of antigens, and the secretion of cytokines. Within the field of oncology, B cells display a multifaceted nature in the tumor microenvironment, simultaneously manifesting both tumor-promoting and tumor-suppressing characteristics. Studies have found that the existence of tertiary lymphoid structures, which consist of B cells, is linked to better survival rates in different types of cancers. This article examines the involvement of B cells in different types of malignancies, emphasizing their importance in the development of the diseases and their potential as biomarkers. Additionally, the review also examines the crucial role of B cells in autoimmune illnesses and their potential as targets for therapy. The article also analyses the role of B cells in immunization and exploring their potential uses in cancer immunotherapy. This analysis highlights the intricate and occasionally contradictory roles of B cells, underlining the necessity for additional research to clarify their varied actions in various illness scenarios.
Collapse
Affiliation(s)
- Soham Bindu
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Chennai, Tamil Nadu 603203, India
| | - Roshni Bibi
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Chennai, Tamil Nadu 603203, India
| | - R Pradeep
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Chennai, Tamil Nadu 603203, India
| | - Koustav Sarkar
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Chennai, Tamil Nadu 603203, India.
| |
Collapse
|
9
|
Yang JI, Moresco P, Fearon D, Yao M. Identification of B cell antigens in solid cancer: initial insights and functional implications. Front Immunol 2025; 16:1571570. [PMID: 40356924 PMCID: PMC12066463 DOI: 10.3389/fimmu.2025.1571570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Accepted: 04/01/2025] [Indexed: 05/15/2025] Open
Abstract
Cancer antigen discovery has mostly focused on T cell antigens, while antigens driving B cell responses have been largely overlooked despite representing another important branch of adaptive immune responses in cancer. Traditional B cell antigens in cancer have been studied using serological approaches analyzing polyclonal antibodies in serum. With recent technological advances in single-cell sequencing, a few studies have begun to investigate single B cell antigen specificity in the tumor microenvironment using immunoglobulin single-cell sequencing, recombinant monoclonal antibody production, cancer binding screening, and antigen identification. In this review, we highlight the initial insights into B cell directed cancer antigens and categorize them into cancer-associated viral antigens and non-viral antigens, with the latter featuring autoantigens. We will further discuss the functions of B cells in cancer in the context of their antigen specificity, and categorize their functions into antibody effector function, T cell activation, and B cell secretion. Lastly, we will provide perspectives on the challenges and opportunities in the identification of new B cell cancer antigens and highlight their translational potential.
Collapse
Affiliation(s)
- Jung-In Yang
- Cancer Center, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, United States
| | - Philip Moresco
- Cancer Center, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, United States
- Graduate Program in Genetics, Stony Brook University, Stony Brook, NY, United States
- Medical Scientist Training Program, Stony Brook University Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, United States
| | - Douglas Fearon
- Cancer Center, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, United States
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY, United States
| | - Min Yao
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY, United States
- Sanders Tri-Institutional Therapeutics Discovery Institute, New York, NY, United States
| |
Collapse
|
10
|
Yu Z, Xu Z, Fu T, Liu S, Cui J, Zhang B, Liang J, Pang C, Ke Y, Wang R, Tang Z, Gao Y, Du B, Feng Y, Zhao H, Xue G, Yan C, Gan L, Feng J, Fan Z, Yang Y, Huang L, Zhao S, Ying S, Gu Q, Yuan J. Parallel comparison of T cell and B cell subpopulations of adenoid hypertrophy and tonsil hypertrophy of children. Nat Commun 2025; 16:3516. [PMID: 40229254 PMCID: PMC11997228 DOI: 10.1038/s41467-025-58094-w] [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: 05/09/2024] [Accepted: 03/12/2025] [Indexed: 04/16/2025] Open
Abstract
The adenoids and tonsils are important immune organs of the nasopharynx that often become hypertrophic in childhood because of recurrent pathogen infection. However, the differences in the immune microenvironment of adenoid hypertrophy (AH) and tonsil hypertrophy (TH) are unclear. Here, we show the epidemiological characteristics and peripheral blood cell indices of 1209 pediatric patients (1-15 years old) diagnosed with AH, and find that AH is often accompanied by TH and characterized by specific changes in immune cell types. Single-cell RNA sequencing analysis show that 12 paired AH and TH samples contain large numbers of B, T cells and some exhausted effector memory CD4+ T cells. Compared with matched TH, AH have more naïve B cells and regulatory CD4+ T cells and less plasma B cells. Weaker antigen presentation and more significant immunosuppression are also observed in AH. In contrast, the number and cytotoxicity of cytotoxic CD8+ T cells decrease with AH grade. These findings will help our understanding of the immune response to nasopharyngeal infection.
Collapse
Affiliation(s)
- Zihui Yu
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, 100020, China
| | - Ziying Xu
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, 100020, China
| | - Tongtong Fu
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, 100020, China
| | - Shiyu Liu
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, 100020, China
- Military supplies and energy quality supervision station of Bejing, Beijing, 100071, China
| | - Jinghua Cui
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, 100020, China
| | - Bing Zhang
- Department of Otolaryngology, Capital Center For Children's Health, Capital Medical University, Beijing, 100020, China
| | - Jieqiong Liang
- Department of Otolaryngology, Capital Center For Children's Health, Capital Medical University, Beijing, 100020, China
| | - Chong Pang
- Department of Otolaryngology, Capital Center For Children's Health, Capital Medical University, Beijing, 100020, China
| | - Yuehua Ke
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, 100020, China
| | - Ruikun Wang
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, 100020, China
- Capital Institute of Pediatrics-Peking University Teaching Hospital, Beijing, 100020, China
| | - Zhijie Tang
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, 100020, China
- Capital Institute of Pediatrics-Peking University Teaching Hospital, Beijing, 100020, China
| | - Yagang Gao
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, 100020, China
| | - Bing Du
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, 100020, China
| | - Yanling Feng
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, 100020, China
| | - Hanqing Zhao
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, 100020, China
| | - Guanhua Xue
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, 100020, China
| | - Chao Yan
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, 100020, China
| | - Lin Gan
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, 100020, China
| | - Junxia Feng
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, 100020, China
| | - Zheng Fan
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, 100020, China
| | - Yang Yang
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, 100020, China
| | - Lijuan Huang
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, 100020, China
| | - Shuo Zhao
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, 100020, China
| | - Sun Ying
- School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Qinglong Gu
- Department of Otolaryngology, Capital Center For Children's Health, Capital Medical University, Beijing, 100020, China.
| | - Jing Yuan
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, 100020, China.
| |
Collapse
|
11
|
Liu GM, Guo R, Xu JW. A bibliometric and visual analysis based on immune checkpoint inhibitors for hepatocellular carcinoma: 2014 - 2024. Front Pharmacol 2025; 16:1520055. [PMID: 40260385 PMCID: PMC12009821 DOI: 10.3389/fphar.2025.1520055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Accepted: 03/24/2025] [Indexed: 04/23/2025] Open
Abstract
Background Immune checkpoint inhibitors (ICIs) have changed the treatment landscape of hepatocellular carcinoma (HCC), especially those with unresectable advanced stages. The field has progressed rapidly, and the research hotspots have significantly changed compared to previous years. The study aims to comprehensively review and analyze the development history, knowledge structure, current research focus, and emerging trends in ICIs for HCC. Materials and methods Reviews and articles published in English from The Web of Science Core Collection (WoSCC) database from 2014 to 2024 were systemically retrieved. Citespace, VOSviewer, and Bibliometrix R package were used for further bibliometric analysis and visualization for countries, institutions, authors, references, and keywords. Results 2,941 records were included for analysis. The literature on ICIs for HCC has continued to grow steadily over the past decade. Three major research centers have emerged: North America, Europe, and East Asia. The Chinese institution has the highest publication volume, but Kudo Masatoshi from Japan has the highest number of publications. At the same time, Richard S. Finn from the United States leads in citations and co-citations. The most prolific journal is "Cancers". The clustering and Timeline view of critical literature and keywords indicated that research on ICIs for HCC is rapidly advancing toward a more evidence-based, personalized, and multimodal approach. Immune evasion mechanisms, predictive biomarkers, and high-quality clinical trials focusing on Novel combination, conversion, and perioperative therapies, including ICIs, are emerging hotspots. Conclusion This study highlights the groundbreaking advancements of ICIs in treating HCC and shows a trend rapidly advancing towards a more evidence-based, personalized, and multimodal approach. The study updated the current understanding of ICIs in hepatocellular carcinoma and identified vital future directions for research, such as the exploration of mechanisms of immune evasion, developing predictive biomarkers, and combining therapy strategies.
Collapse
Affiliation(s)
- Gao-Min Liu
- Meizhou Clinical Medical College of Shantou University Medical College, Meizhou, China
- Department of Hepatobiliary Surgery, Meizhou People’s Hospital, Meizhou, China
| | - Rui Guo
- Department of Hepatobiliary Surgery, Meizhou People’s Hospital, Meizhou, China
| | - Ji-Wei Xu
- Meizhou Clinical Medical College of Shantou University Medical College, Meizhou, China
- Department of Hepatobiliary Surgery, Meizhou People’s Hospital, Meizhou, China
| |
Collapse
|
12
|
Jiao M, Guo Y, Zhang H, Wen H, Chen P, Wang Z, Yu B, Zhuma K, Zhang Y, Qie J, Xing Y, Zhao P, Pan Z, Wang L, Zhang D, Li F, Ren Y, Chen C, Chu Y, Gu J, Liu R. ACAT1 regulates tertiary lymphoid structures and correlates with immunotherapy response in non-small cell lung cancer. J Clin Invest 2025; 135:e181517. [PMID: 40166933 PMCID: PMC11957694 DOI: 10.1172/jci181517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 01/08/2025] [Indexed: 04/02/2025] Open
Abstract
Tertiary lymphoid structures (TLS) in the tumor microenvironment (TME) are emerging solid-tumor indicators of prognosis and response to immunotherapy. Considering that tumorigenesis requires metabolic reprogramming and subsequent TME remodeling, the discovery of TLS metabolic regulators is expected to produce immunotherapeutic targets. To identify such metabolic regulators, we constructed a metabolism-focused sgRNA library and performed an in vivo CRISPR screening in an orthotopic lung tumor mouse model. Combined with The Cancer Genome Atlas database analysis of TLS-related metabolic hub genes, we found that the loss of Acat1 in tumor cells sensitized tumors to anti-PD1 treatment, accompanied by increased TLS in the TME. Mechanistic studies revealed that ACAT1 resulted in mitochondrial protein hypersuccinylation in lung tumor cells and subsequently enhanced mitochondrial oxidative metabolism, which impeded TLS formation. Elimination of ROS by NAC or Acat1 knockdown promoted B cell aggregation and TLS construction. Consistently, data from tissue microassays of 305 patients with lung cancer showed that TLS were more abundant in non-small cell lung cancer (NSCLC) tissues with lower ACAT1 expression. Intratumoral ACAT1 expression was associated with poor immunotherapy outcomes in patients with NSCLC. In conclusion, our results identified ACAT1 as a metabolic regulator of TLS and a promising immunotherapeutic target in NSCLC.
Collapse
Affiliation(s)
- Mengxia Jiao
- Shanghai Fifth People’s Hospital, and Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Yifan Guo
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Hongyu Zhang
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Haoyu Wen
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Thoracic Surgery, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Peng Chen
- Shanghai Fifth People’s Hospital, and Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Zhiqiang Wang
- Shanghai Fifth People’s Hospital, and Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
- Department of Neurology, Children’s Hospital of Fudan University, Shanghai, China
| | - Baichao Yu
- Department of Immunology, School of Basic Medical Sciences, and MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Fudan University, Shanghai, China
| | - Kameina Zhuma
- Department of Immunology, School of Basic Medical Sciences, and MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Fudan University, Shanghai, China
| | - Yuchen Zhang
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jingbo Qie
- Shanghai Fifth People’s Hospital, and Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Yun Xing
- Shanghai Fifth People’s Hospital, and Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Pengyuan Zhao
- Shanghai Fifth People’s Hospital, and Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Zihe Pan
- Shanghai Fifth People’s Hospital, and Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Luman Wang
- Department of Immunology, School of Basic Medical Sciences, and MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Fudan University, Shanghai, China
| | - Dan Zhang
- Shanghai Fifth People’s Hospital, and Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Fei Li
- Department of Pathology and Frontier Innovation Center, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Yijiu Ren
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Chang Chen
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yiwei Chu
- Shanghai Fifth People’s Hospital, and Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
- Department of Immunology, School of Basic Medical Sciences, and MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Fudan University, Shanghai, China
| | - Jie Gu
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Ronghua Liu
- Shanghai Fifth People’s Hospital, and Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| |
Collapse
|
13
|
Cen X, Lan Y, Zou J, Chen R, Hu C, Tong Y, Zhang C, Chen J, Wang Y, Zhou R, He W, Lu T, Dubee F, Jovic D, Dong W, Gao Q, Ma M, Lu Y, Xue Y, Cheng X, Li Y, Yang H. Pan-cancer analysis shapes the understanding of cancer biology and medicine. Cancer Commun (Lond) 2025. [PMID: 40120098 DOI: 10.1002/cac2.70008] [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: 09/10/2024] [Revised: 02/13/2025] [Accepted: 02/16/2025] [Indexed: 03/25/2025] Open
Abstract
Advances in multi-omics datasets and analytical methods have revolutionized cancer research, offering a comprehensive, pan-cancer perspective. Pan-cancer studies identify shared mechanisms and unique traits across different cancer types, which are reshaping diagnostic and treatment strategies. However, continued innovation is required to refine these approaches and deepen our understanding of cancer biology and medicine. This review summarized key findings from pan-cancer research and explored their potential to drive future advancements in oncology.
Collapse
Affiliation(s)
- Xiaoping Cen
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, P. R. China
- HIM-BGI Omics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), Hangzhou, Zhejiang, P. R. China
- BGI Research, Shenzhen, Guangdong, P. R. China
- Guangzhou National Laboratory, Guangzhou, Guangdong, P. R. China
| | - Yuanyuan Lan
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, P. R. China
- HIM-BGI Omics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), Hangzhou, Zhejiang, P. R. China
| | - Jiansheng Zou
- HIM-BGI Omics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), Hangzhou, Zhejiang, P. R. China
- College of Information Engineering, Zhejiang University of Technology, Hangzhou, Zhejiang, P. R. China
| | - Ruilin Chen
- HIM-BGI Omics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), Hangzhou, Zhejiang, P. R. China
- College of Information Engineering, Zhejiang University of Technology, Hangzhou, Zhejiang, P. R. China
| | - Can Hu
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, P. R. China
| | - Yahan Tong
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, P. R. China
| | - Chen Zhang
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, P. R. China
- HIM-BGI Omics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), Hangzhou, Zhejiang, P. R. China
- BGI Research, Shenzhen, Guangdong, P. R. China
| | - Jingyue Chen
- HIM-BGI Omics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), Hangzhou, Zhejiang, P. R. China
- Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, Zhejiang, P. R. China
| | - Yuanmei Wang
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, P. R. China
- HIM-BGI Omics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), Hangzhou, Zhejiang, P. R. China
- BGI Research, Shenzhen, Guangdong, P. R. China
| | - Run Zhou
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, P. R. China
- HIM-BGI Omics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), Hangzhou, Zhejiang, P. R. China
- BGI Research, Shenzhen, Guangdong, P. R. China
| | - Weiwei He
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, P. R. China
- HIM-BGI Omics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), Hangzhou, Zhejiang, P. R. China
| | - Tianyu Lu
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, P. R. China
- BGI Research, Shenzhen, Guangdong, P. R. China
| | - Fred Dubee
- BGI Research, Shenzhen, Guangdong, P. R. China
| | | | - Wei Dong
- HIM-BGI Omics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), Hangzhou, Zhejiang, P. R. China
- Clin Lab, BGI Genomics, Beijing, P. R. China
| | - Qingqing Gao
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, P. R. China
- HIM-BGI Omics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), Hangzhou, Zhejiang, P. R. China
- BGI Research, Shenzhen, Guangdong, P. R. China
| | - Man Ma
- HIM-BGI Omics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), Hangzhou, Zhejiang, P. R. China
| | - Youyong Lu
- Laboratory of Molecular Oncology, Peking University Cancer Hospital and Institute, Beijing, P. R. China
| | - Yu Xue
- MOE Key Laboratory of Molecular Biophysics, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Center for Artificial Intelligence Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, P. R. China
| | - Xiangdong Cheng
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, P. R. China
| | - Yixue Li
- Guangzhou National Laboratory, Guangzhou, Guangdong, P. R. China
- GZMU-GIBH Joint School of Life Sciences, The Guangdong-Hong Kong-Macau Joint Laboratory for Cell Fate Regulation and Diseases, Guangzhou Medical University, Guangzhou, Guangdong, P. R. China
| | - Huanming Yang
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, P. R. China
- BGI, Shenzhen, Guangdong, P. R. China
- James D. Watson Institute of Genome Sciences, Hangzhou, Zhejiang, P. R. China
| |
Collapse
|
14
|
Zhang H, Pang Y, Yi L, Wang X, Wei P, Wang H, Lin S. Epigenetic regulators combined with tumour immunotherapy: current status and perspectives. Clin Epigenetics 2025; 17:51. [PMID: 40119465 PMCID: PMC11929245 DOI: 10.1186/s13148-025-01856-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Accepted: 03/03/2025] [Indexed: 03/24/2025] Open
Abstract
Immunotherapy, particularly immune checkpoint inhibitor therapy, has demonstrated clinical benefits in solid tumours. Despite its satisfactory clinical efficacy, it still faces several issues, such as limited eligibility, low response rates and cytotoxicity. Cancer epigenetics implies that tumour cells exhibit unique phenotypes because of their unique characteristics, thus reprogramming of the epigenome holds promise for cancer therapy. Epigenetic regulation plays an important role in regulating gene expression during tumour development and maintenance. Epigenetic regulators induce cancer cell cycle arrest, apoptosis and differentiation of cancer cells, thereby exerting anti-tumour effects. Recent studies have revealed a significant correlation between epigenetic regulatory factors and immune checkpoint therapy. Epigenetics can modulate various aspects of the tumour immune microenvironment and immune response to enhance the sensitivity of immunotherapy, such as lowering the concentration required and mitigating cytotoxicity. This review primarily discusses DNA methyltransferase inhibitors, histone deacetylase inhibitors, enhancer of zeste homolog 2 inhibitors and lysine-specific demethylase 1 inhibitors, which are associated with transcriptional repression. This repression alters the expression of genes involved in the immune checkpoint, thereby enhancing the effectiveness of immunotherapy. We also discuss the potential and challenges of tumour immunotherapy and highlight its advantages, application challenges and clinical research on integrating epigenetic regulatory factors with tumour immunotherapy.
Collapse
Affiliation(s)
- Huan Zhang
- Department of Gastroenterology, Beijing Chest Hospital, Beijing Tuberculosis and Thoracic Tumor Research Institute, Capital Medical University, Beijing, 101149, China
| | - Yutong Pang
- Department of Gastroenterology, Beijing Chest Hospital, Beijing Tuberculosis and Thoracic Tumor Research Institute, Capital Medical University, Beijing, 101149, China
| | - Ling Yi
- Cancer Research Center, Beijing Chest Hospital, Beijing Tuberculosis and Thoracic Tumor Research Institute, Capital Medical University, Beijing, 101149, China
| | - Xiaojue Wang
- Cancer Research Center, Beijing Chest Hospital, Beijing Tuberculosis and Thoracic Tumor Research Institute, Capital Medical University, Beijing, 101149, China
| | - Panjian Wei
- Cancer Research Center, Beijing Chest Hospital, Beijing Tuberculosis and Thoracic Tumor Research Institute, Capital Medical University, Beijing, 101149, China
| | - Haichao Wang
- Institute of Resources and Environment, Beijing Academy of Science and Technology, Beijing, 100089, China.
| | - Shuye Lin
- Department of Gastroenterology, Beijing Chest Hospital, Beijing Tuberculosis and Thoracic Tumor Research Institute, Capital Medical University, Beijing, 101149, China.
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, 101149, China.
| |
Collapse
|
15
|
Ma L, Luan Y, Lu L. Analyze the Diversity and Function of Immune Cells in the Tumor Microenvironment From the Perspective of Single-Cell RNA Sequencing. Cancer Med 2025; 14:e70622. [PMID: 40062730 PMCID: PMC11891933 DOI: 10.1002/cam4.70622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 12/14/2024] [Accepted: 01/09/2025] [Indexed: 05/13/2025] Open
Abstract
BACKGROUND Cancer development is closely associated with complex alterations in the tumor microenvironment (TME). Among these, immune cells within the TME play a huge role in personalized tumor diagnosis and treatment. OBJECTIVES This review aims to summarize the diversity of immune cells in the TME, their impact on patient prognosis and treatment response, and the contributions of single-cell RNA sequencing (scRNA-seq) in understanding their functional heterogeneity. METHODS We analyzed recent studies utilizing scRNA-seq to investigate immune cell populations in the TME, focusing on their interactions and regulatory mechanisms. RESULTS ScRNA-seq reveals the functional heterogeneity of immune cells, enhances our understanding of their role in tumor antibody responses, and facilitates the construction of immune cell interaction networks. These insights provide guidance for the development of cancer immunotherapies and personalized treatment approaches. CONCLUSION Applying scRNA-seq to immune cell analysis in the TME offers a novel pathway for personalized cancer treatment. Despite its promise, several challenges remain, highlighting the need for further advancements to fully integrate scRNA-seq into clinical applications.
Collapse
Affiliation(s)
- Lujuan Ma
- Department of Medical Oncology, Guangzhou First People's Hospital, School of MedicineSouth China University of TechnologyGuangzhouGuangdongChina
| | - Yu Luan
- Department of Medical Oncology, Guangzhou First People's Hospital, School of MedicineSouth China University of TechnologyGuangzhouGuangdongChina
| | - Lin Lu
- Department of Medical Oncology, Guangzhou First People's Hospital, School of MedicineSouth China University of TechnologyGuangzhouGuangdongChina
| |
Collapse
|
16
|
Yin Z, Li L, Zhang Q, Zhang X, Shi R, Xia X, Wang Z, Li S, Ye M, Liu Y, Tan W, Chen Z. PerC B-Cells Activation via Thermogenetics-Based CXCL12 Generator for Intraperitoneal Immunity Against Metastatic Disseminated Tumor Cells. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2411731. [PMID: 39865939 DOI: 10.1002/adma.202411731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 01/04/2025] [Indexed: 01/28/2025]
Abstract
During cancer peritoneal metastasis (PM), conventional antigen-presenting cells (dendritic cells, macrophages) promote tumorigenesis and immunosuppression in peritoneal cavity. While intraperitoneal immunotherapy (IPIT) has been used in clinical investigations to relieve PM, the limited knowledge of peritoneal immunocytes has hindered the development of therapeutic IPIT. Here, a dendritic cell-independent, next-generation IPIT is described that activates peritoneal cavity B (PerC B) cell subsets for intraperitoneal anti-tumor immunity via exogenous antigen presentation. The PerC B-cell-involved IPIT framework consists of an isotropic-porous, cell-fitting, thermogenetics-based CXCL12 generator. Such nanoscale thermal-confined generator can programmatically fine-tune the expression of CXCL12 to recruit disseminated tumor cells (DTCs) through CXCL12-CXCR4 axis while avoiding cytokine storm, subsequently release DTC-derived antigen to trigger PerC B-cell-involved immunity. Notably, antigen-presenting B-cell cluster, expressing the regulatory signaling molecules Ptpn6, Ms4a1, and Cd52, is identified playing the key role in the IPIT via single-cell RNA sequencing. Moreover, such IPIT availably assuages peritoneal effusion and PM in an orthotopic gastric cancer and metastatic model. Overall, this work offers a perspective on PerC B-cell-involved antigen-presenting in intraperitoneal immunity and provides a configurable strategy for activating anti-DTC immunity for next-generation IPIT.
Collapse
Affiliation(s)
- Zhiwei Yin
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Environmental Science and Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, 410082, China
| | - Ling Li
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Environmental Science and Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, 410082, China
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310018, China
| | - Qiang Zhang
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Environmental Science and Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, 410082, China
- Department of Chemistry, College of Sciences, Northeastern University, Shenyang, 110819, China
| | - Xiaoshen Zhang
- Department of Respiratory Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Rui Shi
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Environmental Science and Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, 410082, China
| | - Xin Xia
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Environmental Science and Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, 410082, China
| | - Zhaoxin Wang
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Environmental Science and Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, 410082, China
| | - Shengkai Li
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Environmental Science and Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, 410082, China
| | - Mao Ye
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Environmental Science and Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, 410082, China
| | - Yanlan Liu
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Environmental Science and Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, 410082, China
| | - Weihong Tan
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Environmental Science and Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, 410082, China
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310018, China
- Institute of Molecular Medicine (IMM), Renji Hospital, Shanghai Jiao Tong University School of Medicine and College of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Zhuo Chen
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Environmental Science and Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, 410082, China
| |
Collapse
|
17
|
Tang M, Xu Y, Pan M. Integrative Analysis of scRNA-Seq and Bulk RNA-Seq Identifies Plasma Cell Related Genes and Constructs a Prognostic Model for Hepatocellular Carcinoma. J Hepatocell Carcinoma 2025; 12:427-444. [PMID: 40040881 PMCID: PMC11878290 DOI: 10.2147/jhc.s509749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Accepted: 02/21/2025] [Indexed: 03/06/2025] Open
Abstract
Purpose The complexity and heterogeneity of the tumor immune microenvironment (TIME) are linked to the development and poor prognosis of hepatocellular carcinoma (HCC). However, the cell type within the TIME that is most closely associated with HCC development remains unclear. Herein, we aimed to identify cell clusters that significantly contribute to HCC development and their underlying mechanisms. Method and Results Using single-cell RNA sequencing (scRNA-seq), we analyzed changes in the TIME of normal and tumor tissues, identifying plasma cells as the key cluster in HCC development. Based on plasma cell-related genes (PCRGs), we constructed and validated an eight-gene prognostic model (ST6GALNAC4, SEC61A1, SSR3, RPN2, PRDX4, TRAM1, SPCS2, CD79A) using internal and external datasets and a nomogram. Functional enrichment, miRNA network construction, and transcriptional regulation analyses were performed to explore underlying mechanisms. TIDE scores and the GDSC database were used to predict immunotherapy and chemotherapy sensitivity in different risk groups. Finally, SSR3's biological function was validated in vitro in HCC cell lines. Conclusion Plasma cells are key clusters in HCC development. A prognostic model based on the PCRGs can accurately predict the prognosis of patients with HCC and guide clinical treatment.
Collapse
Affiliation(s)
- Mingyang Tang
- General Surgery Center, Department of Hepatobiliary Surgery II, Zhujiang Hospital, Southern Medical University, Guangzhou, 510000, People’s Republic of China
| | - Yuyan Xu
- General Surgery Center, Department of Hepatobiliary Surgery II, Zhujiang Hospital, Southern Medical University, Guangzhou, 510000, People’s Republic of China
| | - Mingxin Pan
- General Surgery Center, Department of Hepatobiliary Surgery II, Zhujiang Hospital, Southern Medical University, Guangzhou, 510000, People’s Republic of China
| |
Collapse
|
18
|
Neo SY, Shuen TWH, Khare S, Chong J, Lau M, Shirgaonkar N, Chua L, Zhao J, Lee K, Tan C, Ba R, Lim J, Chua J, Cheong HS, Chai HM, Chan CY, Chung AYF, Cheow PC, Jeyaraj PR, Teo JY, Koh YX, Chok AY, Chow PKH, Goh B, Wan WK, Leow WQ, Loh TJZ, Tang PY, Karunanithi J, Ngo NT, Lim TKH, Xu S, Dasgupta R, Toh HC, Lam KP. Atypical memory B cells acquire Breg phenotypes in hepatocellular carcinoma. JCI Insight 2025; 10:e187025. [PMID: 39998891 PMCID: PMC11981623 DOI: 10.1172/jci.insight.187025] [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: 02/27/2025] Open
Abstract
The functional plasticity of tumor-infiltrating lymphocyte B-cells (TIL-B) spans from antitumor responses to noncanonical immune suppression. Yet, how the tumor microenvironment (TME) influences TIL-B development is still underappreciated. Our current study integrated single-cell transcriptomics and B cell receptor (BCR) sequencing to profile TIL-B phenotypes and clonalities in hepatocellular carcinoma (HCC). Using trajectory and gene regulatory network analysis, we were able to characterize plasma cells and memory and naive B cells within the HCC TME and further revealed a downregulation of BCR signaling genes in plasma cells and a subset of inflammatory TNF+ memory B cells. Within the TME, a nonswitched memory B cell subset acquired an age-associated B cell phenotype (TBET+CD11c+) and expressed higher levels of PD-L1, CD25, and granzyme B. We further demonstrated that the presence of HCC tumor cells could confer suppressive functions on peripheral blood B cells that in turn, dampen T cell costimulation. To the best of our knowledge, these findings represent novel mechanisms of noncanonical immune suppression in HCC. While previous studies identified atypical memory B cells in chronic hepatitis and across several solid cancer types, we further highlighted their potential role as regulatory B cells (Bregs) within both the TME and peripheral blood of HCC patients.
Collapse
Affiliation(s)
- Shi Yong Neo
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore
- Department of Oncology and Pathology, Karolinska Institute, Stockholm, Sweden
| | | | - Shruti Khare
- Laboratory of Precision Oncology and Cancer Evolution, Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), Singapore
| | - Joni Chong
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore
| | - Maichan Lau
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore
| | - Niranjan Shirgaonkar
- Laboratory of Precision Oncology and Cancer Evolution, Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), Singapore
| | - Levene Chua
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore
| | - Junzhe Zhao
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore
| | - Keene Lee
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore
| | - Charmaine Tan
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore
| | - Rebecca Ba
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore
| | - Janice Lim
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore
| | - Joelle Chua
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore
| | - Hui Shi Cheong
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore
| | - Hui Min Chai
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore
| | - Chung Yip Chan
- Department of Hepato-pancreato-biliary and Transplant Surgery, Singapore General Hospital, Singapore
| | - Alexander Yaw Fui Chung
- Department of Hepato-pancreato-biliary and Transplant Surgery, Singapore General Hospital, Singapore
| | - Peng Chung Cheow
- Department of Hepato-pancreato-biliary and Transplant Surgery, Singapore General Hospital, Singapore
| | - Prema Raj Jeyaraj
- Department of Hepato-pancreato-biliary and Transplant Surgery, Singapore General Hospital, Singapore
| | - Jin Yao Teo
- Department of Hepato-pancreato-biliary and Transplant Surgery, Singapore General Hospital, Singapore
| | - Ye Xin Koh
- Department of Hepato-pancreato-biliary and Transplant Surgery, Singapore General Hospital, Singapore
| | - Aik Yong Chok
- Department of Hepato-pancreato-biliary and Transplant Surgery, Singapore General Hospital, Singapore
| | - Pierce Kah Hoe Chow
- Department of Hepato-pancreato-biliary and Transplant Surgery, Singapore General Hospital, Singapore
- Division of Surgery and Surgical Oncology, National Cancer Centre Singapore, Singapore
| | - Brian Goh
- Department of Hepato-pancreato-biliary and Transplant Surgery, Singapore General Hospital, Singapore
| | - Wei Keat Wan
- Department of Anatomical Pathology, Singapore General Hospital, Singapore
| | - Wei Qiang Leow
- Department of Anatomical Pathology, Singapore General Hospital, Singapore
| | - Tracy Jie Zhen Loh
- Department of Anatomical Pathology, Singapore General Hospital, Singapore
| | - Po Yin Tang
- Department of Anatomical Pathology, Singapore General Hospital, Singapore
| | | | - Nye Thane Ngo
- Department of Anatomical Pathology, Singapore General Hospital, Singapore
| | - Tony Kiat Hon Lim
- Department of Anatomical Pathology, Singapore General Hospital, Singapore
| | - Shengli Xu
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Ramanuj Dasgupta
- Laboratory of Precision Oncology and Cancer Evolution, Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), Singapore
| | - Han Chong Toh
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore
| | - Kong-Peng Lam
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore
- School of Biological Sciences, Nanyang Technological University, Singapore
- Department of Microbiology & Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| |
Collapse
|
19
|
Ruffin AT, Casey AN, Kunning SR, MacFawn IP, Liu Z, Arora C, Rohatgi A, Kemp F, Lampenfeld C, Somasundaram A, Rappocciolo G, Kirkwood JM, Duvvuri U, Seethala R, Bao R, Huang Y, Cillo AR, Ferris RL, Bruno TC. Dysfunctional CD11c -CD21 - extrafollicular memory B cells are enriched in the periphery and tumors of patients with cancer. Sci Transl Med 2025; 17:eadh1315. [PMID: 39970232 DOI: 10.1126/scitranslmed.adh1315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Revised: 02/07/2024] [Accepted: 01/16/2025] [Indexed: 02/21/2025]
Abstract
Many patients with recurrent and metastatic cancer fail to produce a durable response to immunotherapy, highlighting the need for additional therapeutic targets to improve the immune landscape in tumors. Recent studies have highlighted the importance of B cells in the antitumor response, with memory B cells (MBCs) being prognostic in a variety of solid tumors. MBCs are a heterogenous B cell subset and can be generated through both germinal center reactions and extrafollicular (EF) responses. EF-derived MBCs have been recently linked to poor prognosis and treatment resistance in solid tumors and thus may represent candidate biomarkers or immunotherapy targets. EF-derived MBCs, termed "double-negative" (DN) MBCs may be further classified on the basis of surface expression of CD11c and CD21 into DN1, DN2, and DN3 MBCs. CD11c-CD21+ DN1 MBCs and CD11c+CD21- DN2 MBCs have been well studied across inflammatory diseases; however, the biology and clinical relevance of CD11c-CD21- DN3 MBCs remain unknown. Here, we report an accumulation of DN3 MBCs in the blood and tumors of patients with head and neck squamous cell carcinoma (HNSCC) and an increase in DN3 MBCs in locally advanced HNSCC tumors. Circulating and intratumoral DN3 MBCs were hyporesponsive to antigen stimulation, had low antibody production, and failed to differentiate into antibody-secreting cells. Moreover, DN3 MBCs accumulated selectively outside of tertiary lymphoid structures. Last, circulating DN3 MBCs correlated with poor therapeutic response, advanced disease, and worse outcomes in patients with HNSCC and melanoma, supporting further assessment of EF-derived MBCs as potential biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Ayana T Ruffin
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Tumor Microenvironment Center, Hillman Cancer Center, University of Pittsburgh, PA 15213, USA
- Program in Microbiology and Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
- UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA
| | - Allison N Casey
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Tumor Microenvironment Center, Hillman Cancer Center, University of Pittsburgh, PA 15213, USA
- Molecular Genetics and Developmental Biology Graduate Program, Pittsburgh, PA 15213, USA
| | - Sheryl R Kunning
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Tumor Microenvironment Center, Hillman Cancer Center, University of Pittsburgh, PA 15213, USA
- UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA
| | - Ian P MacFawn
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Tumor Microenvironment Center, Hillman Cancer Center, University of Pittsburgh, PA 15213, USA
- UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA
| | - Zhentao Liu
- UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Charu Arora
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Tumor Microenvironment Center, Hillman Cancer Center, University of Pittsburgh, PA 15213, USA
- UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA
| | - Anjali Rohatgi
- Tumor Microenvironment Center, Hillman Cancer Center, University of Pittsburgh, PA 15213, USA
- UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA
| | - Felicia Kemp
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Program in Microbiology and Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Caleb Lampenfeld
- Tumor Microenvironment Center, Hillman Cancer Center, University of Pittsburgh, PA 15213, USA
- UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA
| | - Ashwin Somasundaram
- Tumor Microenvironment Center, Hillman Cancer Center, University of Pittsburgh, PA 15213, USA
- UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA
| | | | - John M Kirkwood
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Umamaheswar Duvvuri
- Department of Otolaryngology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Raja Seethala
- UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA
- Department of Otolaryngology, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Riyue Bao
- UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Yufei Huang
- UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Department of Electrical and Computer Engineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Department of Pharmaceutical Sciences, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Anthony R Cillo
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Tumor Microenvironment Center, Hillman Cancer Center, University of Pittsburgh, PA 15213, USA
- UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA
- Cancer Immunology and Immunotherapy Program, UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA
- Center for Systems Immunology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Robert L Ferris
- Tumor Microenvironment Center, Hillman Cancer Center, University of Pittsburgh, PA 15213, USA
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Department of Otolaryngology, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Cancer Immunology and Immunotherapy Program, UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA
| | - Tullia C Bruno
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Tumor Microenvironment Center, Hillman Cancer Center, University of Pittsburgh, PA 15213, USA
- UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA
- Cancer Immunology and Immunotherapy Program, UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA
| |
Collapse
|
20
|
Xiong X, Zheng LW, Ding Y, Chen YF, Cai YW, Wang LP, Huang L, Liu CC, Shao ZM, Yu KD. Breast cancer: pathogenesis and treatments. Signal Transduct Target Ther 2025; 10:49. [PMID: 39966355 PMCID: PMC11836418 DOI: 10.1038/s41392-024-02108-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Revised: 10/27/2024] [Accepted: 12/08/2024] [Indexed: 02/20/2025] Open
Abstract
Breast cancer, characterized by unique epidemiological patterns and significant heterogeneity, remains one of the leading causes of malignancy-related deaths in women. The increasingly nuanced molecular subtypes of breast cancer have enhanced the comprehension and precision treatment of this disease. The mechanisms of tumorigenesis and progression of breast cancer have been central to scientific research, with investigations spanning various perspectives such as tumor stemness, intra-tumoral microbiota, and circadian rhythms. Technological advancements, particularly those integrated with artificial intelligence, have significantly improved the accuracy of breast cancer detection and diagnosis. The emergence of novel therapeutic concepts and drugs represents a paradigm shift towards personalized medicine. Evidence suggests that optimal diagnosis and treatment models tailored to individual patient risk and expected subtypes are crucial, supporting the era of precision oncology for breast cancer. Despite the rapid advancements in oncology and the increasing emphasis on the clinical precision treatment of breast cancer, a comprehensive update and summary of the panoramic knowledge related to this disease are needed. In this review, we provide a thorough overview of the global status of breast cancer, including its epidemiology, risk factors, pathophysiology, and molecular subtyping. Additionally, we elaborate on the latest research into mechanisms contributing to breast cancer progression, emerging treatment strategies, and long-term patient management. This review offers valuable insights into the latest advancements in Breast Cancer Research, thereby facilitating future progress in both basic research and clinical application.
Collapse
Affiliation(s)
- Xin Xiong
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Cancer Institute, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P. R. China
| | - Le-Wei Zheng
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Cancer Institute, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P. R. China
| | - Yu Ding
- Department of Breast and Thyroid, Guiyang Maternal and Child Health Care Hospital & Guiyang Children's Hospital, Guiyang, P. R. China
- Department of Clinical Medicine, Guizhou Medical University, Guiyang, P. R. China
| | - Yu-Fei Chen
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Cancer Institute, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P. R. China
| | - Yu-Wen Cai
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Cancer Institute, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P. R. China
| | - Lei-Ping Wang
- Department of Breast and Urologic Medical Oncology, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P. R. China
| | - Liang Huang
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Cancer Institute, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P. R. China
| | - Cui-Cui Liu
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Cancer Institute, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P. R. China
| | - Zhi-Ming Shao
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Cancer Institute, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P. R. China
| | - Ke-Da Yu
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Cancer Institute, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P. R. China.
| |
Collapse
|
21
|
Samiea A, Celis G, Yadav R, Rodda LB, Moreau JM. B cells in non-lymphoid tissues. Nat Rev Immunol 2025:10.1038/s41577-025-01137-6. [PMID: 39910240 DOI: 10.1038/s41577-025-01137-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/16/2025] [Indexed: 02/07/2025]
Abstract
B cells have long been understood to be drivers of both humoral and cellular immunity. Recent advances underscore this importance but also indicate that in infection, inflammatory disease and cancer, B cells function directly at sites of inflammation and form tissue-resident memory populations. The spatial organization and cellular niches of tissue B cells have profound effects on their function and on disease outcome, as well as on patient response to therapy. Here we review the role of B cells in peripheral tissues in homeostasis and disease, and discuss the newly identified cellular and molecular signals that are involved in regulating their activity. We integrate emerging data from multi-omic human studies with experimental models to propose a framework for B cell function in tissue inflammation and homeostasis.
Collapse
Affiliation(s)
- Abrar Samiea
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, OR, USA
- Cancer Early Detection Advanced Research Center, Oregon Health & Science University, Portland, OR, USA
| | - George Celis
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, OR, USA
| | - Rashi Yadav
- Cancer Early Detection Advanced Research Center, Oregon Health & Science University, Portland, OR, USA
| | - Lauren B Rodda
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, OR, USA.
| | - Joshua M Moreau
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, OR, USA.
- Cancer Early Detection Advanced Research Center, Oregon Health & Science University, Portland, OR, USA.
- Department of Dermatology, Oregon Health & Science University, Portland, OR, USA.
- Division of Oncological Sciences, Oregon Health & Science University, Portland, OR, USA.
| |
Collapse
|
22
|
Yan J, Jiang Z, Zhang S, Yu Q, Lu Y, Miao R, Tang Z, Fan J, Wu L, Duda DG, Zhou J, Yang X. Spatial‒temporal heterogeneities of liver cancer and the discovery of the invasive zone. Clin Transl Med 2025; 15:e70224. [PMID: 39924620 PMCID: PMC11807767 DOI: 10.1002/ctm2.70224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Accepted: 01/19/2025] [Indexed: 02/11/2025] Open
Abstract
Solid tumours are intricate and highly heterogeneous ecosystems, which grow in and invade normal organs. Their progression is mediated by cancer cells' interaction with different cell types, such as immune cells, stromal cells and endothelial cells, and with the extracellular matrix. Owing to its high incidence, aggressive growth and resistance to local and systemic treatments, liver cancer has particularly high mortality rates worldwide. In recent decades, spatial heterogeneity has garnered significant attention as an unfavourable biological characteristic of the tumour microenvironment, prompting extensive research into its role in liver tumour development. Advances in spatial omics have facilitated the detailed spatial analysis of cell types, states and cell‒cell interactions, allowing a thorough understanding of the spatial and temporal heterogeneities of tumour microenvironment and informing the development of novel therapeutic approaches. This review illustrates the latest discovery of the invasive zone, and systematically introduced specific macroscopic spatial heterogeneities, pathological spatial heterogeneities and tumour microenvironment heterogeneities of liver cancer.
Collapse
Affiliation(s)
- Jiayan Yan
- Department of Liver Surgery & TransplantationLiver Cancer InstituteZhongshan HospitalFudan UniversityShanghaiChina
- Key Laboratory of Carcinogenesis and Cancer InvasionMinistry of EducationShanghaiChina
- Zhongshan‐BGI Precision Medical CenterZhongshan HospitalFudan UniversityShanghaiChina
| | - Zhifeng Jiang
- Department of Liver Surgery & TransplantationLiver Cancer InstituteZhongshan HospitalFudan UniversityShanghaiChina
- Key Laboratory of Carcinogenesis and Cancer InvasionMinistry of EducationShanghaiChina
- Zhongshan‐BGI Precision Medical CenterZhongshan HospitalFudan UniversityShanghaiChina
| | - Shiyu Zhang
- Department of Liver Surgery & TransplantationLiver Cancer InstituteZhongshan HospitalFudan UniversityShanghaiChina
- Key Laboratory of Carcinogenesis and Cancer InvasionMinistry of EducationShanghaiChina
- Zhongshan‐BGI Precision Medical CenterZhongshan HospitalFudan UniversityShanghaiChina
| | - Qichao Yu
- College of Life SciencesUniversity of Chinese Academy of SciencesBeijingChina
- BGI‐ShenzhenBeishan Industrial ZoneShenzhenChina
| | - Yijun Lu
- Department of Liver Surgery & TransplantationLiver Cancer InstituteZhongshan HospitalFudan UniversityShanghaiChina
- Key Laboratory of Carcinogenesis and Cancer InvasionMinistry of EducationShanghaiChina
- Zhongshan‐BGI Precision Medical CenterZhongshan HospitalFudan UniversityShanghaiChina
| | - Runze Miao
- Department of Liver Surgery & TransplantationLiver Cancer InstituteZhongshan HospitalFudan UniversityShanghaiChina
- Key Laboratory of Carcinogenesis and Cancer InvasionMinistry of EducationShanghaiChina
- Zhongshan‐BGI Precision Medical CenterZhongshan HospitalFudan UniversityShanghaiChina
| | - Zhaoyou Tang
- Department of Liver Surgery & TransplantationLiver Cancer InstituteZhongshan HospitalFudan UniversityShanghaiChina
- Key Laboratory of Carcinogenesis and Cancer InvasionMinistry of EducationShanghaiChina
| | - Jia Fan
- Department of Liver Surgery & TransplantationLiver Cancer InstituteZhongshan HospitalFudan UniversityShanghaiChina
- Key Laboratory of Carcinogenesis and Cancer InvasionMinistry of EducationShanghaiChina
| | - Liang Wu
- BGI‐ShenzhenBeishan Industrial ZoneShenzhenChina
| | - Dan G. Duda
- Steele Laboratories for Tumor BiologyDepartment of Radiation OncologyMassachusetts General Hospital and Harvard Medical SchoolBostonMassachusettsUSA
| | - Jian Zhou
- Department of Liver Surgery & TransplantationLiver Cancer InstituteZhongshan HospitalFudan UniversityShanghaiChina
- Key Laboratory of Carcinogenesis and Cancer InvasionMinistry of EducationShanghaiChina
| | - Xinrong Yang
- Department of Liver Surgery & TransplantationLiver Cancer InstituteZhongshan HospitalFudan UniversityShanghaiChina
- Key Laboratory of Carcinogenesis and Cancer InvasionMinistry of EducationShanghaiChina
| |
Collapse
|
23
|
Song S, Wang C, Chen Y, Zhou X, Han Y, Zhang H. Dynamic roles of tumor-infiltrating B lymphocytes in cancer immunotherapy. Cancer Immunol Immunother 2025; 74:92. [PMID: 39891668 PMCID: PMC11787113 DOI: 10.1007/s00262-024-03936-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 12/27/2024] [Indexed: 02/03/2025]
Abstract
The amazing diversity of B cells within the tumor microenvironment is the basis for the diverse development of B cell-based immunotherapies. Here, we focus on elucidating the mechanisms of tumor intervention mediated by four tumor-infiltrating B lymphocytes. Naive B cells present the initial antigen, germinal center B cell subsets enhance antibody affinity, and immunoglobulin subtypes exert multiple immune effects, while regulatory B cells establish immune tolerance. Together they reflect the complexity of the changing dynamics of cancer immunity. Additionally, we have investigated the dynamic effects of tumor-infiltrating B lymphocytes in immunotherapy and their relationship to prognosis, providing new insights into potential treatment strategies for patients.
Collapse
Affiliation(s)
- Shishengnan Song
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, China
| | - Chong Wang
- Department of Thoracic Surgery, Beijing Chest Hospital Affiliated to Capital Medical University (Beijing Tuberculosis and Thoracic Tumor Research Institute), 9 Beiguan Street, Tongzhou, 101149, Beijing, China
| | - Yangchao Chen
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, 999077, NT, China
| | - Xiaorong Zhou
- Department of Immunology, Medical School of Nantong University, 19 Qixiu Road, Nantong, 226001, Jiangsu, China.
| | - Yi Han
- Department of Thoracic Surgery, Beijing Chest Hospital Affiliated to Capital Medical University (Beijing Tuberculosis and Thoracic Tumor Research Institute), 9 Beiguan Street, Tongzhou, 101149, Beijing, China.
| | - Haijian Zhang
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, China.
| |
Collapse
|
24
|
Bowley TY, Ortiz MC, Lagutina IV, Steinkamp MP, Fahy BN, Tawfik B, Harari-Turquie M, Marchetti D. A Melanoma Brain Metastasis CTC Signature and CTC:B-cell Clusters Associate with Secondary Liver Metastasis: A Melanoma Brain-Liver Metastasis Axis. CANCER RESEARCH COMMUNICATIONS 2025; 5:295-308. [PMID: 39831781 PMCID: PMC11816052 DOI: 10.1158/2767-9764.crc-24-0498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 11/20/2024] [Accepted: 01/15/2025] [Indexed: 01/22/2025]
Abstract
SIGNIFICANCE This study provides important insights into the relevance of prometastatic CTC:B-cell clusters in melanoma progression, extends the importance of the CTC RPL/RPS gene signature beyond primary metastasis/melanoma brain metastasis driving targeted organ specificity for liver metastasis ("metastasis of metastasis"), and identifies new targets for clinical melanoma metastasis therapies.
Collapse
Affiliation(s)
- Tetiana Y. Bowley
- Division of Molecular Medicine, Department of Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Mireya C. Ortiz
- Division of Molecular Medicine, Department of Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Irina V. Lagutina
- Animal Models Shared Resource, The University of New Mexico Comprehensive Cancer Center, Albuquerque, New Mexico
| | - Mara P. Steinkamp
- Department of Pathology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Bridget N. Fahy
- Division of Surgical Oncology and Palliative Medicine, University of New Mexico Comprehensive Cancer Center, Albuquerque, New Mexico
| | - Bernard Tawfik
- Division of Hematology and Oncology, Department of Internal Medicine, University of New Mexico Comprehensive Cancer Center, Albuquerque, New Mexico
| | - Moises Harari-Turquie
- Division of Hematology and Oncology, Department of Internal Medicine, University of New Mexico Comprehensive Cancer Center, Albuquerque, New Mexico
| | - Dario Marchetti
- Division of Molecular Medicine, Department of Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
- Department of Pathology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| |
Collapse
|
25
|
Ricci JE. Tumor-induced metabolic immunosuppression: Mechanisms and therapeutic targets. Cell Rep 2025; 44:115206. [PMID: 39798090 DOI: 10.1016/j.celrep.2024.115206] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 12/04/2024] [Accepted: 12/23/2024] [Indexed: 01/15/2025] Open
Abstract
Metabolic reprogramming in both immune and cancer cells plays a crucial role in the antitumor immune response. Recent studies indicate that cancer metabolism not only sustains carcinogenesis and survival via altered signaling but also modulates immune cell function. Metabolic crosstalk within the tumor microenvironment results in nutrient competition and acidosis, thereby hindering immune cell functionality. Interestingly, immune cells also undergo metabolic reprogramming that enables their proliferation, differentiation, and effector functions. This review highlights the regulation of antitumor immune responses through metabolic reprogramming in cancer and immune cells and explores therapeutic strategies that target these metabolic pathways in cancer immunotherapy, including using chimeric antigen receptor (CAR)-T cells. We discuss innovative combinations of immunotherapy, cellular therapies, and metabolic interventions that could optimize the efficacy of existing treatment protocols.
Collapse
Affiliation(s)
- Jean-Ehrland Ricci
- Université Côte d'Azur, INSERM, C3M, Nice, France; Équipe labellisée LIGUE Contre le Cancer, Nice, France.
| |
Collapse
|
26
|
Asashima H, Akao S, Matsumoto I. Emerging roles of checkpoint molecules on B cells. Immunol Med 2025:1-12. [PMID: 39819449 DOI: 10.1080/25785826.2025.2454045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 01/07/2025] [Indexed: 01/19/2025] Open
Abstract
Immune checkpoint molecules, including both co-inhibitory molecules and co-stimulatory molecules, are known to play critical roles in regulating T-cell responses. During the last decades, immunotherapies targeting these molecules (such as programmed cell death 1 (PD-1), and lymphocyte activation gene 3 (LAG-3)) have provided clinical benefits in many cancers. It is becoming apparent that not only T cells, but also B cells have a capacity to express some checkpoint molecules. These were originally thought to be only the markers for regulatory B cells which produce IL-10, but recent studies suggest that these molecules (especially T-cell immunoglobulin and mucin domain 1 (TIM-1), T cell immunoreceptor with Ig and ITIM domains (TIGIT), and PD-1) can regulate intrinsic B-cell activation and functions. Here, we focus on these molecules and summarize their characteristics, ligands, and functions on B cells.
Collapse
Affiliation(s)
- Hiromitsu Asashima
- Department of Rheumatology, Institute of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Satoshi Akao
- Department of Rheumatology, Institute of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Isao Matsumoto
- Department of Rheumatology, Institute of Medicine, University of Tsukuba, Ibaraki, Japan
| |
Collapse
|
27
|
Yang Z, Chen Y, Miao Y, Yan H, Chen K, Xu Y, Su L, Zhang L, Yan Y, Chi H, Fu J, Wang L. Elucidating stearoyl metabolism and NCOA4-mediated ferroptosis in gastric cancer liver metastasis through multi-omics single-cell integrative mendelian analysis: advancing personalized immunotherapy strategies. Discov Oncol 2025; 16:46. [PMID: 39812999 PMCID: PMC11735723 DOI: 10.1007/s12672-025-01769-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Accepted: 01/02/2025] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND The metabolism of stearoyl-GPE plays a key role in the liver metastasis of gastric cancer. This investigation delves into the mechanisms underlying the intricate tumor microenvironment (TME) heterogeneity triggered by stearoyl metabolism in gastric cancer with liver metastasis (LMGC), offering novel perspectives for LMGC. OBJECTIVE Utilizing Mendelian randomization, we determined that stearoyl metabolism significantly contributes to the progression of gastric cancer (GC). Following this, bulk transcriptome analyses and single-cell multiomics techniques to investigate the roles of stearoyl-GPE metabolism-related genes, particularly NCOA4, in regulating LMGC TME. RESULTS Our analysis highlights the crucial role of stearoyl metabolism in modulating the complex microenvironment of LMGC, particularly impacting monocyte cells. Through single-cell sequencing and spatial transcriptomics, we have identified key metabolic genes specific to stearoyl metabolism within the monocyte cell population, including NCOA4. Regarding the relationship between ferroptosis, stearoyl metabolism, and LMGC findings, it is plausible that stearoyl metabolism and LMGC pathways intersect with mechanisms involved in ferroptosis. Ferroptosis, characterized by iron-dependent lipid peroxidation, represents a regulated form of cell death. The activity of Stearoyl-CoA desaturase (SCD), a critical enzyme in stearoyl metabolism, has been associated with the modulation of lipid composition and susceptibility to ferroptosis. Furthermore, the LMGC is integral to cellular processes related to oxidative stress and lipid metabolism, both of which are significant factors in the context of ferroptosis. CONCLUSION This study enhances the understanding of the relationship between stearoyl metabolism and ferroptosis in promoting liver metastasis of gastric cancer and its role in the regulation of tumor heterogeneity. In addition, this study contributes to a deeper understanding of the dynamics of gastric cancer tumor microenvironment (TME) and provides a basis for the development of better interventions to combat cancer metastasis.
Collapse
Affiliation(s)
- Zhongqiu Yang
- Department of General Surgery, Dazhou Central Hospital, Dazhou, 635000, China
| | - Yuquan Chen
- School of Public Health and Preventive Medicine, Faculty of Medicine, Nursing & Health Sciences, Monash University, Victoria, 3004, Australia
| | - Yaping Miao
- General Hospital of Ningxia Medical University, Yinchuan, 750000, Ningxia, China
- Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Haisheng Yan
- General Hospital of Ningxia Medical University, Yinchuan, 750000, Ningxia, China
- Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Kexin Chen
- General Hospital of Ningxia Medical University, Yinchuan, 750000, Ningxia, China
- Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Yaoqin Xu
- General Hospital of Ningxia Medical University, Yinchuan, 750000, Ningxia, China
- Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Lanqian Su
- School of Clinical Medicine, Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Lanyue Zhang
- School of Clinical Medicine, Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Yalan Yan
- School of Clinical Medicine, Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Hao Chi
- School of Clinical Medicine, Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China.
- Western Institute of Digital-Intelligent Medicine, 401329, Chongqing, China.
| | - Jin Fu
- Department of Laboratory Medicine, Chonggang General Hospital, Chongqing, 400080, China.
| | - Lexin Wang
- Western Institute of Digital-Intelligent Medicine, 401329, Chongqing, China.
| |
Collapse
|
28
|
Huang QF, Wang GF, Zhang YM, Zhang C, Ran YQ, He JZ, Wang G, Xu XE, Wang SH, Wu JY, Li EM, Xu LY. Lympho-myeloid aggregate-infiltrating CD20 + B cells display a double-negative phenotype and correlate with poor prognosis in esophageal squamous cell carcinoma. Transl Res 2025; 275:48-61. [PMID: 39536938 DOI: 10.1016/j.trsl.2024.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 09/19/2024] [Accepted: 11/10/2024] [Indexed: 11/16/2024]
Abstract
According to morphological features, tumor-infiltrating B cells (TIL-Bs) can be classified as lympho-myeloid aggregates (LMAs) and tertiary lymphoid structures (TLSs). As a disease with high incidence and mortality, research on esophageal squamous cell carcinoma (ESCC) TIL-Bs is still unclear. Thus, we aimed to investigate the prognostic value and functional involvement of TIL-Bs in ESCC. Based on CD20 immunohistochemical staining of 147 ESCC samples, the TIL-Bs at different anatomic subregions (intra-tumor (T), invasive margin (IM) and peri-tumor (P)) were quantified and correlated with survival by Kaplan-Meier analyses. We found that LMAs were widely distributed throughout the whole section and were associated with poor prognosis, especially those located in the T subregion, which was contrary to the positive clinical significance of TLSs. Based on the number of LMAs and TLSs, a four-level immune type was constructed as an independent predictor for survival. Using multiplexed immunofluorescence (mIF) staining, we found that the main phenotype of infiltrating B cells in LMAs was CD20+IgD-CD27- double-negative (DN) B cells. DN B cells were abundant in ESCC tumor tissue, and their high expression was related to shortened overall survival time. Subsequently, we demonstrate a close relationship between DN B cells and regulatory T cells (Tregs) using single cell RNA-seq data, bulk RNA-seq data and flow cytometry, and verified the spatial proximity of DN B cells and Tregs by mIF staining. Trajectory analysis and flow cytometry revealed that DN B cells highly expressed genes involved in the antigen processing and presentation pathway, such as HLA-DR. The abundance of DN B cells and LMAs in ESCC provides novel potential targets for optimal immunotherapy against ESCC.
Collapse
Affiliation(s)
- Qing-Feng Huang
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Institute of Oncologic Pathology, Shantou University Medical College, Shantou 515041, Guangdong, PR China; Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, Guangdong, PR China
| | - Ge-Fei Wang
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Institute of Oncologic Pathology, Shantou University Medical College, Shantou 515041, Guangdong, PR China
| | - Yi-Meng Zhang
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Institute of Oncologic Pathology, Shantou University Medical College, Shantou 515041, Guangdong, PR China; Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, Guangdong, PR China
| | - Cong Zhang
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Institute of Oncologic Pathology, Shantou University Medical College, Shantou 515041, Guangdong, PR China; Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, Guangdong, PR China
| | - Ying-Qi Ran
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Institute of Oncologic Pathology, Shantou University Medical College, Shantou 515041, Guangdong, PR China; Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, Guangdong, PR China
| | - Jian-Zhong He
- Department of Pathology, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai 519000, Guangdong Province, PR China
| | - Geng Wang
- Department of Thoracic Surgery, Cancer Hospital of Shantou University Medical College, Shantou 515041, Guangdong, PR China
| | - Xiu-E Xu
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Institute of Oncologic Pathology, Shantou University Medical College, Shantou 515041, Guangdong, PR China; Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, Guangdong, PR China
| | - Shao-Hong Wang
- Departments of Pathology, Shantou Central Hospital, Shantou 515041, Guangdong, PR China
| | - Jian-Yi Wu
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Institute of Oncologic Pathology, Shantou University Medical College, Shantou 515041, Guangdong, PR China; Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, Guangdong, PR China
| | - En-Min Li
- Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, Guangdong, PR China; Shantou Academy Medical Sciences, Shantou 515041, Guangdong, PR China; Guangdong Esophageal Cancer Research Institute, Shantou Sub-center, Cancer Research Center, Shantou University Medical College, Shantou 515041, Guangdong, PR China.
| | - Li-Yan Xu
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Institute of Oncologic Pathology, Shantou University Medical College, Shantou 515041, Guangdong, PR China; Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, Guangdong, PR China; Guangdong Esophageal Cancer Research Institute, Shantou Sub-center, Cancer Research Center, Shantou University Medical College, Shantou 515041, Guangdong, PR China.
| |
Collapse
|
29
|
Lin L, Wen J, Xu T, Si Y. TM4SF4 is a diagnostic biomarker accelerating progression of papillary thyroid cancer via AKT pathway. Cancer Biol Ther 2024; 25:2424570. [PMID: 39497261 PMCID: PMC11540099 DOI: 10.1080/15384047.2024.2424570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Revised: 10/27/2024] [Accepted: 10/28/2024] [Indexed: 11/08/2024] Open
Abstract
The incidence of papillary thyroid cancer (PTC) has been steadily rising, though the underlying mechanism remains unclear. This study aims to elucidate the biological role of TM4SF4 in the PTC progression. Our differential expression analysis indicated that TM4SF4 was significantly upregulated in PTC, which was corroborated in both our local cohort and the data from Human Protein Atlas. Additionally, clinical characteristics analysis and receiver operating characteristic curves (ROC) demonstrated that TM4SF4 served as a significant diagnostic marker for PTC. Correlation and enrichment analysis of TM4SF4-related partners suggested that it was involved in cell junction and cohesion processes. Furthermore, immune infiltration analysis revealed a positive correlation between TM4SF4 expression and the immune activation in PTC. Importantly, in vitro experiments demonstrated that TM4SF4 downregulation suppressed the proliferation and metastasis of PTC cell lines while inducing apoptosis. We further discovered that the AKT activator SC79 was able to reverse the malignant behaviors suppression caused by TM4SF4 knockdown, suggesting that TM4SF4 may promote PTC progression via the AKT pathway. In conclusion, our study highlights the oncogenic role of TM4SF4 in PTC and identifies it as a novel biomarker for diagnosis and treatment.
Collapse
Affiliation(s)
- Lizhi Lin
- Department of Surgery, Clinical Sciences Lund, Lund University, Lund, Sweden
- Department of Gastrointestinal Surgery, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jialiang Wen
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, PR China
| | - Tiansheng Xu
- Department of Anorectal Surgery, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People’s Hospital, Quzhou, Zhejiang, PR China
| | - Yuhao Si
- Department of Anorectal Surgery, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People’s Hospital, Quzhou, Zhejiang, PR China
- Department of Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, PR China
| |
Collapse
|
30
|
Huang CX, Lao XM, Wang XY, Ren YZ, Lu YT, Shi W, Wang YZ, Wu CY, Xu L, Chen MS, Gao Q, Liu L, Wei Y, Kuang DM. Pericancerous cross-presentation to cytotoxic T lymphocytes impairs immunotherapeutic efficacy in hepatocellular carcinoma. Cancer Cell 2024; 42:2082-2097.e10. [PMID: 39547231 DOI: 10.1016/j.ccell.2024.10.012] [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: 06/11/2024] [Revised: 09/09/2024] [Accepted: 10/21/2024] [Indexed: 11/17/2024]
Abstract
Hyperprogressive disease can occur in cancer patients receiving immune checkpoint blockade (ICB) therapy, but whether and how reactive cytotoxic T lymphocytes (CTLs) exert protumorigenic effects in this context remain elusive. Herein, our study reveals that pericancerous macrophages cross-present antigens to CD103+ CTLs in hepatocellular carcinoma (HCC) via the endoplasmic reticulum (ER)-associated degradation machinery-mediated cytosolic pathway. This process leads to the retention of CD103+ CTLs in the pericancerous area, whereby they activate NLRP3 inflammasome in macrophages, promoting hepatoma progression and resistance to immunotherapy. Our single-cell RNA sequencing (scRNA-seq) and spatial transcriptomics analysis of HCC patients shows that despite their tissue-resident effector phenotype, the aggregation of CD103+ CTLs predicts unfavorable clinical outcomes for HCC patients receiving multiple types of treatment. Correspondingly, therapeutic strategies that redistribute CD103+ CTLs can disrupt this pathogenic interplay with macrophages, enhancing the efficacy of ICB treatment against HCC.
Collapse
MESH Headings
- Carcinoma, Hepatocellular/immunology
- Carcinoma, Hepatocellular/therapy
- Carcinoma, Hepatocellular/pathology
- Liver Neoplasms/immunology
- Liver Neoplasms/therapy
- Liver Neoplasms/pathology
- T-Lymphocytes, Cytotoxic/immunology
- Humans
- Immunotherapy/methods
- Macrophages/immunology
- Mice
- Animals
- Integrin alpha Chains/metabolism
- Integrin alpha Chains/immunology
- Cross-Priming/immunology
- Antigens, CD/metabolism
- Antigens, CD/immunology
- Immune Checkpoint Inhibitors/therapeutic use
- Immune Checkpoint Inhibitors/pharmacology
- NLR Family, Pyrin Domain-Containing 3 Protein/metabolism
- NLR Family, Pyrin Domain-Containing 3 Protein/immunology
- Inflammasomes/immunology
- Inflammasomes/metabolism
- Tumor Microenvironment/immunology
- Cell Line, Tumor
- Mice, Inbred C57BL
Collapse
Affiliation(s)
- Chun-Xiang Huang
- Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, MOE Key Laboratory of Gene Function and Regulation, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, China
| | - Xiang-Ming Lao
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Sun Yat-sen University, Guangzhou 510060, China
| | - Xu-Yan Wang
- Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, MOE Key Laboratory of Gene Function and Regulation, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, China
| | - Yi-Zheng Ren
- Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, MOE Key Laboratory of Gene Function and Regulation, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, China
| | - Yi-Tong Lu
- Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, MOE Key Laboratory of Gene Function and Regulation, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, China
| | - Wei Shi
- Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, MOE Key Laboratory of Gene Function and Regulation, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, China
| | - Ying-Zhe Wang
- Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, MOE Key Laboratory of Gene Function and Regulation, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, China
| | - Cai-Yuan Wu
- Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, MOE Key Laboratory of Gene Function and Regulation, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, China
| | - Li Xu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Sun Yat-sen University, Guangzhou 510060, China
| | - Min-Shan Chen
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Sun Yat-sen University, Guangzhou 510060, China
| | - Qiang Gao
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Lianxin Liu
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Yuan Wei
- Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, MOE Key Laboratory of Gene Function and Regulation, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, China; Innovation Center of the Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510275, China.
| | - Dong-Ming Kuang
- Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, MOE Key Laboratory of Gene Function and Regulation, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, China; Innovation Center of the Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510275, China.
| |
Collapse
|
31
|
Bod L, Shalapour S. B cells spatial organization defines their phenotype and function in cancer "Tell me with whom you consort, and I will tell you who you are" - Goethe. Curr Opin Immunol 2024; 91:102504. [PMID: 39547092 DOI: 10.1016/j.coi.2024.102504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 10/15/2024] [Accepted: 10/21/2024] [Indexed: 11/17/2024]
Abstract
The presence of B cells and their subtypes in the tumor environment has been recognized a for very long time. Immunoglobulins specific for more than thousands of tumor-associated antigens were detected in the sera of patients with cancer; however, antibody-mediated cancer cell killing is usually impaired. The role of humoral immune response remained elusive until recently, with new discoveries regarding their contribution in regulating antitumor immunity, particularly during immunotherapy. Humoral immunity has been described to promote or attenuate tumorigenesis and can have opposing effects on therapeutic outcome in different tumor entities. The antagonism effect of B cells depends on their subtypes and immunoglobulin isotypes and is regulated by their spatial distribution and localization. In this short review, we will focus on how the spatial organization of B cells within the tumor microenvironment, tumor-associated lymph nodes, and tertiary lymphoid structures define their fate and function and contribute to the regulation of antitumor immunity.
Collapse
Affiliation(s)
- Lloyd Bod
- Department of Medicine, Krantz Family Center for Cancer Research, Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Shabnam Shalapour
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA.
| |
Collapse
|
32
|
Yang Y, Li M, Ding L, Zhang Y, Liu K, Liu M, Li Y, Luo H, Zuo X, Zhang H, Guo M. EZH2 promotes B-cell autoimmunity in primary Sjogren's syndrome via METTL3-mediated m6A modification. J Autoimmun 2024; 149:103341. [PMID: 39577129 DOI: 10.1016/j.jaut.2024.103341] [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: 08/08/2024] [Revised: 11/05/2024] [Accepted: 11/14/2024] [Indexed: 11/24/2024]
Abstract
OBJECTIVE Enhancer of zeste homologue 2 (EZH2) plays an important role in promoting B-cell activation and differentiation. This study aimed to elucidate the role of EZH2 in the B-cell autoimmune response in primary Sjögren's syndrome (pSS) and to explore the therapeutic potential of inhibiting EZH2 in pSS. METHODS Single-cell RNA sequencing analysis of B cells in peripheral blood from pSS patients was conducted to identify abnormal expression of EZH2 and METTL3 in B-cell subsets. The levels of EZH2 were further validated across multiple B-cell subsets and the salivary glands (SGs) of pSS patients, as well as three different mouse models of Sjögren's syndrome (SS). Correlation analyses were performed to explore the relationship between the expression of EZH2 and clinical features of pSS patients. Following EZH2 inhibition, SS-like signs and antibody production were assessed in an experimental Sjögren syndrome (ESS) mouse model. RNA sequencing (RNA-seq) and chromatin immunoprecipitation sequencing (ChIP-seq) data post-EZH2 inhibition were bioinformatically analyzed to identify the EZH2 targets in pSS. ChIP-qPCR was performed to validate the binding of H3K27me3 to the CDKN1A promoter. Flow cytometric apoptosis analysis and Carboxy Fluorescein Succinimidyl Ester (CFSE) assay were used to assess the impact of an EZH2 inhibitor on B-cell apoptosis and proliferation. Additionally, METTL3 expression and its correlation with disease activity were analyzed in pSS patients. EZH2 expression was examined after METTL3 knockdown. METTL3-RNA immunoprecipitation (RIP) and actinomycin D assays were conducted to confirm the direct binding of METTL3 to EZH2 mRNA and its impact on mRNA stability. M6A-RIP-qPCR was performed to validate the presence of m6A modifications on EZH2 mRNA. RESULTS EZH2 was found upregulated in multiple B-cell subsets from the peripheral blood and SGs of pSS patients, as well as in three different animal models of SS. The expression of EZH2 in B cells was positively correlated with the ESSDAI score, which is a measure of disease activity. With treatment of EZH2 inhibitor, SS-like signs alleviated and autoantibody production reduced in ESS mice. Similarly, in pSS patients, METTL3 expression was increased in the SGs and peripheral blood CD19+ B cells, also showing a positively correlated with the ESSDAI score. With knockdown of METTL3, the expression of EZH2 reduced. Mechanistically, EZH2 inhibited B-cell apoptosis and promoted B-cell proliferation by catalyzing H3K27me3 modification at the CDKN1A locus. Furthermore, METTL3 bound to EZH2 mRNA and increased m6A modification on EZH2 mRNA, enhancing its stability and promoting EZH2 expression. CONCLUSIONS The upregulation of EZH2 mediated by METTL3 is implicated in the B-cell autoimmune response in pSS. Inhibition of EZH2 presents a promising therapeutic strategy for pSS treatment.
Collapse
Affiliation(s)
- Yiying Yang
- Department of Rheumatology, Xiangya Hospital, Central South University, Changsha, Hunan, China; Department of Pathophysiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, China; Sepsis Translational Medicine Key Lab of Hunan Province, Changsha, Hunan, China; Postdoctoral Research Station of Biology, School of Basic Medicine Science, Central South University, Changsha, Hunan, China
| | - Muyuan Li
- Department of Rheumatology, Xiangya Hospital, Central South University, Changsha, Hunan, China; Department of Pathophysiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, China; Sepsis Translational Medicine Key Lab of Hunan Province, Changsha, Hunan, China; Provincial Clinical Research Center for Rheumatic and Immunologic Diseases, Xiangya Hospital, Central South University, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Liqing Ding
- Department of Rheumatology, Xiangya Hospital, Central South University, Changsha, Hunan, China; Provincial Clinical Research Center for Rheumatic and Immunologic Diseases, Xiangya Hospital, Central South University, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ying Zhang
- Department of Pathophysiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, China; Sepsis Translational Medicine Key Lab of Hunan Province, Changsha, Hunan, China
| | - Ke Liu
- Department of Pathophysiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, China; Sepsis Translational Medicine Key Lab of Hunan Province, Changsha, Hunan, China
| | - Meidong Liu
- Department of Pathophysiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, China; Sepsis Translational Medicine Key Lab of Hunan Province, Changsha, Hunan, China
| | - Yisha Li
- Department of Rheumatology, Xiangya Hospital, Central South University, Changsha, Hunan, China; Provincial Clinical Research Center for Rheumatic and Immunologic Diseases, Xiangya Hospital, Central South University, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Hui Luo
- Department of Rheumatology, Xiangya Hospital, Central South University, Changsha, Hunan, China; Provincial Clinical Research Center for Rheumatic and Immunologic Diseases, Xiangya Hospital, Central South University, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xiaoxia Zuo
- Department of Rheumatology, Xiangya Hospital, Central South University, Changsha, Hunan, China; Provincial Clinical Research Center for Rheumatic and Immunologic Diseases, Xiangya Hospital, Central South University, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Huali Zhang
- Department of Rheumatology, Xiangya Hospital, Central South University, Changsha, Hunan, China; Department of Pathophysiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, China; Sepsis Translational Medicine Key Lab of Hunan Province, Changsha, Hunan, China.
| | - Muyao Guo
- Department of Rheumatology, Xiangya Hospital, Central South University, Changsha, Hunan, China; Provincial Clinical Research Center for Rheumatic and Immunologic Diseases, Xiangya Hospital, Central South University, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
33
|
Liu Z, Lin X, Zhang D, Guo D, Tang W, Yu X, Zhang F, Zhang S, Xue R, Shen X, Dong L. Increased PRP19 in Hepatocyte Impedes B Cell Function to Promote Hepatocarcinogenesis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2407517. [PMID: 39422063 PMCID: PMC11633487 DOI: 10.1002/advs.202407517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 09/25/2024] [Indexed: 10/19/2024]
Abstract
Tumor immune microenvironment is strongly associated with the malignancy behavior of hepatocellular carcinoma (HCC). However, the immune function and regulatory mechanisms of B cells in HCC remain unclear. The expression differences between B cell high- and low-infiltration HCC samples are explored to identify the key regulator. Pre-mRNA processing factor 19 (PRP19) expression is increased in B cell low-infiltrated tissues and negatively correlated with the B cell marker, CD20. Inhibition of PRP19 expression promoted B cell infiltration in tumor tissue and impeded HCC growth. Mechanically, the co-immunoprecipitation (Co-IP) assay revealed that PRP19 interacts with DEAD-box helicase 5 (DDX5), leading to ubiquitination and degradation of the DDX5 protein. The attenuated DDX5 impairs CXCL12 mRNA stability to suppress B cell recruitment and plasma cell differentiation via CXCL12/CXCR4 axis. Moreover, the adoptive transfer of CXCR4+ B cells combined with CXCL12 treatment in mice models effectively inhibits HCC development by reshaping the immune response. The expression of PRP19, DDX5, and infiltrating B cells are recognized as clinical prognosis indicators for HCC patients. Overall, this study provides valuable insights into the clinical benefits of HCC immunotherapy by targeting PRP19 and modulating tumor-infiltrating B cell immune function.
Collapse
Affiliation(s)
- Zhiyong Liu
- Department of Gastroenterology and HepatologyShanghai Institute of Liver DiseaseZhongshan HospitalFudan UniversityShanghai200030China
| | - Xiahui Lin
- Department of Gastroenterology and HepatologyShanghai Institute of Liver DiseaseZhongshan HospitalFudan UniversityShanghai200030China
| | - Danying Zhang
- Department of Gastroenterology and HepatologyShanghai Institute of Liver DiseaseZhongshan HospitalFudan UniversityShanghai200030China
| | - Dezhen Guo
- Department of Liver SurgeryZhongshan HospitalFudan UniversityShanghai200030China
| | - Wenqing Tang
- Department of Gastroenterology and HepatologyShanghai Institute of Liver DiseaseZhongshan HospitalFudan UniversityShanghai200030China
| | - Xiangnan Yu
- Department of Gastroenterology and HepatologyShanghai Institute of Liver DiseaseZhongshan HospitalFudan UniversityShanghai200030China
| | - Feng Zhang
- Department of Gastroenterology and HepatologyShanghai Institute of Liver DiseaseZhongshan HospitalFudan UniversityShanghai200030China
| | - Si Zhang
- NHC Key Laboratory of Glycoconjugate ResearchDepartment of Biochemistry and Molecular BiologySchool of Basic Medical SciencesFudan UniversityShanghai200030China
| | - Ruyi Xue
- Department of Gastroenterology and HepatologyShanghai Institute of Liver DiseaseZhongshan HospitalFudan UniversityShanghai200030China
| | - Xizhong Shen
- Department of Gastroenterology and HepatologyShanghai Institute of Liver DiseaseZhongshan HospitalFudan UniversityShanghai200030China
| | - Ling Dong
- Department of Gastroenterology and HepatologyShanghai Institute of Liver DiseaseZhongshan HospitalFudan UniversityShanghai200030China
| |
Collapse
|
34
|
Zhang Z, Zhao W, Lv C, Wu Z, Liu W, Chang X, Yu Y, Xiao Z, He Y, Zhang H. Unraveling impact and potential mechanisms of baseline pain on efficacy of immunotherapy in lung cancer patients: a retrospective and bioinformatic analysis. Front Immunol 2024; 15:1456150. [PMID: 39654896 PMCID: PMC11625792 DOI: 10.3389/fimmu.2024.1456150] [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: 06/28/2024] [Accepted: 10/30/2024] [Indexed: 12/12/2024] Open
Abstract
Objective Pain is a prevalent discomfort symptom associated with cancer, yet the correlations and potential mechanisms between pain and the efficacy of cancer immunotherapy remain uncertain. Methods Non-small cell lung cancer (NSCLC) patients who received immune checkpoint inhibitors (ICIs) in the inpatient department of Guangdong Provincial Hospital of Chinese Medicine from January 1, 2018, to December 31, 2021, were retrospectively enrolled. Through cox regression analysis, prognostic factors and independent prognostic factors affecting the efficacy of ICIs were identified, and a nomogram model was constructed. Hub cancer-related pain genes (CRPGs) were identified through bioinformatic analysis. Finally, the expression levels of hub CRPGs were detected using an enzyme-linked immunosorbent assay (ELISA). Results Before PSM, a total of 222 patients were enrolled in this study. Univariate and multivariate cox analysis indicated that bone metastasis and NRS scores were independent prognostic factors for the efficacy of ICIs. After PSM, a total of 94 people were enrolled in this study. Univariate cox analysis and multivariate cox analysis indicated that age, platelets, Dnlr, liver metastasis, bone metastasis, and NRS scores were independent prognostic factors for the efficacy of ICIs. A nomogram was constructed based on 6 independent prognostic factors with AUC values of 0.80 for 1-year, 0.73 for 2-year, and 0.80 for 3-year survival. ELISA assay results indicated that the level of CXCL12 significantly decreased compared to baseline after pain was relieved. Conclusion Baseline pain is an independent prognostic factor affecting the efficacy of ICIs in lung cancer, potentially through CXCL12-mediated inflammation promotion and immunosuppression.
Collapse
Affiliation(s)
- Zexin Zhang
- The Second Clinical School of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Wenjie Zhao
- The Second Clinical School of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Chang Lv
- The Second Clinical School of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zexia Wu
- The Second Clinical School of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Wenhao Liu
- Clinical Medical College of Acupuncture Moxibustion and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xuesong Chang
- Deparment of Oncology, Guangdong Province Hospital of Chinese Medicine, Guangzhou, China
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yaya Yu
- Deparment of Oncology, Guangdong Province Hospital of Chinese Medicine, Guangzhou, China
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhenzhen Xiao
- Deparment of Oncology, Guangdong Province Hospital of Chinese Medicine, Guangzhou, China
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yihan He
- Deparment of Oncology, Guangdong Province Hospital of Chinese Medicine, Guangzhou, China
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Haibo Zhang
- Deparment of Oncology, Guangdong Province Hospital of Chinese Medicine, Guangzhou, China
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangzhou, China
- Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou University of Chinese Medicine, Guangzhou, China
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
35
|
Chen S, Liu J, He G, Tang N, Zeng Y. Research Hotspots and Trends in Global Cancer immunometabolism:A Bibliometric Analysis from 2000 to 2023. J Multidiscip Healthc 2024; 17:5117-5137. [PMID: 39553266 PMCID: PMC11568773 DOI: 10.2147/jmdh.s495330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Accepted: 10/30/2024] [Indexed: 11/19/2024] Open
Abstract
Background Cancer poses a major global health challenge, and immunotherapy, known as the third revolution in cancer treatment, has brought new hope to patients. The emerging field of immunometabolism has further enhanced the safety and efficacy of immunotherapy. Over the past two decades, this field has rapidly evolved in oncology, leading to numerous significant findings. This review systematically examines the literature on immunometabolism in cancer, visualizing research trends and identifying future directions. Methods A comprehensive literature search was conducted in the Web of Science, PubMed, and Scopus databases, covering publications from January 2000 to December 2023. We employed tools like Citespace, VOSviewer, and RStudio for visual analysis of publication trends, regional contributions, institutions, authors, journals, and keywords. Results A total of 3320 articles were published by 8090 authors across 1738 institutions, involving 71 countries. Leading contributors were China (n=469), the United States (n=361), and Germany (n=82). Harvard University was the most influential institution, while Frontiers in Immunology had the highest number of publications. The top research areas included glucose, lipid, and amino acid metabolism, the tumor microenvironment, and immune cell regulation. Conclusion International collaboration and interdisciplinary efforts are advancing the field of cancer immunometabolism. Future research will likely focus on the interplay between metabolism and immunity, metabolic markers, immune cell reprogramming, and tumor-immune metabolic competition.
Collapse
Affiliation(s)
- Shupeng Chen
- School of Clinical Medicine, Jiangxi University of Chinese Medicine, Nanchang, 330004, People’s Republic of China
| | - Jie Liu
- School of Clinical Medicine, Jiangxi University of Chinese Medicine, Nanchang, 330004, People’s Republic of China
| | - Guilian He
- School of Clinical Medicine, Jiangxi University of Chinese Medicine, Nanchang, 330004, People’s Republic of China
| | - Nana Tang
- Hematology Department, Affiliated Hospital of Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi, 330006, People’s Republic of China
| | - Yingjian Zeng
- Hematology Department, Affiliated Hospital of Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi, 330006, People’s Republic of China
| |
Collapse
|
36
|
Cang X, Li N, Qi J, Chen H, Xing H, Qiu J, Tian Y, Huang S, Deng P, Gao F, Chaulagain RP, Ullah U, Wang C, Liu L, Jin S. Identification of immune-associated genes for the diagnosis of ulcerative colitis-associated carcinogenesis via integrated bioinformatics analysis. Front Oncol 2024; 14:1475189. [PMID: 39582536 PMCID: PMC11581968 DOI: 10.3389/fonc.2024.1475189] [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: 08/03/2024] [Accepted: 10/23/2024] [Indexed: 11/26/2024] Open
Abstract
Background UC patients suffer more from colorectal cancer (CRC) than the general population, which increases with disease duration. Early colonoscopy is difficult because ulcerative colitis-associated colorectal cancer (UCAC) lesions are flat and multifocal. Our study aimed to identify promising UCAC biomarkers that are complementary endoscopy strategies in the early stages. Methods The datasets may be accessed from the Gene Expression Omnibus and The Cancer Genome Atlas databases. The co-expressed modules of UC and CRC were determined via weighted co-expression network analysis (WGCNA). The biological mechanisms of the shared genes were exported for analysis using the Gene Ontology and Kyoto Encyclopedia of Genes and Genomes. To identify protein interactions and hub genes, a protein-protein interaction network and CytoHubba analysis were conducted. To evaluate gene expression, external datasets and experimental validation of human colon tissues were utilized. The diagnostic value of core genes was examined through receiver operating characteristic (ROC) curves. Immune infiltration analysis was employed to investigate the associations between immune cell populations and hub genes. Results Three crucial modules were identified from the WGCNA of UC and CRC tissues, and 33 coexpressed genes that were predominantly enriched in the NF-κB pathway were identified. Two biomarkers (CXCL1 and BCL6) were identified via Cytoscape and validated in external datasets and human colon tissues. CRC patients expressed CXCL1 at the highest level, whereas UC and CRC patients showed higher levels than the controls. The UC cohort expressed BCL6 at the highest level, whereas the UC and CRC cohorts expressed it more highly than the controls. The hub genes exhibited significant diagnostic potential (ROC curve > 0.7). The immune infiltration results revealed a correlation among the hub genes and macrophages, neutrophils and B cells. Conclusions The findings of our research suggest that BCL6 and CXCL1 could serve as effective biomarkers for UCAC surveillance. Additionally, they demonstrated a robust correlation with immune cell populations within the CRC tumour microenvironment (TME). Our findings provide a valuable insight about diagnosis and therapy of UCAC.
Collapse
Affiliation(s)
- Xueyu Cang
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Ning Li
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jihan Qi
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Hongliang Chen
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Hui Xing
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jiawei Qiu
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yingying Tian
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Shiling Huang
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Pengchao Deng
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Feiyang Gao
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Ram Prasad Chaulagain
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Ubaid Ullah
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Chunjing Wang
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Lina Liu
- Department of Endoscopic Center, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Shizhu Jin
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
37
|
Conejo-Garcia JR, Lopez-Bailon LU, Anadon CM. Unraveling spontaneous humoral immune responses against human cancer: a road to novel immunotherapies. J Leukoc Biol 2024; 116:919-926. [PMID: 39190797 DOI: 10.1093/jleuko/qiae179] [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: 05/08/2024] [Revised: 07/01/2024] [Accepted: 08/23/2024] [Indexed: 08/29/2024] Open
Abstract
In immuno-oncology, the focus has traditionally been on αβ T cells, and immune checkpoint inhibitors that primarily target PD-1 or CTLA4 in these lymphocytes have revolutionized the management of multiple human malignancies. However, recent research highlights the crucial role of B cells and the antibodies they produce in antagonizing malignant progression, offering new avenues for immunotherapy. Our group has demonstrated that dimeric Immunoglobulin A can penetrate tumor cells, neutralize oncogenic drivers in endosomes, and expel them from the cytosol. This mechanistic insight suggests that engineered antibodies targeting this pathway may effectively reach previously inaccessible targets. Investigating antibody production within intratumoral germinal centers and understanding the impact of different immunoglobulins on malignant progression could furnish new tools for the therapeutic arsenal, including the development of tumor-penetrating antibodies. This review aims to elucidate the nature of humoral adaptive immune responses in human cancer and explore how they could herald a new era of immunotherapeutic modalities. By expanding the scope of antitumor immunotherapies, these approaches have the potential to benefit a broader range of cancer patients, particularly through the utilization of tumor cell-penetrating antibodies.
Collapse
Affiliation(s)
- Jose R Conejo-Garcia
- Department of Integrative Immunobiology, Duke School of Medicine, Durham, NC 27710, United States
- Duke Cancer Institute, Duke School of Medicine, Durham, NC 27710, United States
| | - Luis U Lopez-Bailon
- Department of Integrative Immunobiology, Duke School of Medicine, Durham, NC 27710, United States
- Duke Cancer Institute, Duke School of Medicine, Durham, NC 27710, United States
| | - Carmen M Anadon
- Department of Integrative Immunobiology, Duke School of Medicine, Durham, NC 27710, United States
- Duke Cancer Institute, Duke School of Medicine, Durham, NC 27710, United States
| |
Collapse
|
38
|
Yang H, Zhang Z, Li J, Wang K, Zhu W, Zeng Y. The Dual Role of B Cells in the Tumor Microenvironment: Implications for Cancer Immunology and Therapy. Int J Mol Sci 2024; 25:11825. [PMID: 39519376 PMCID: PMC11546796 DOI: 10.3390/ijms252111825] [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: 09/08/2024] [Revised: 10/21/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024] Open
Abstract
The tumor microenvironment (TME) is a complex and heterogeneous tissue composed of various cell types, including tumor cells, stromal cells, and immune cells, as well as non-cellular elements. Given their pivotal role in humoral immunity, B cells have emerged as promising targets for anti-tumor therapies. The dual nature of B cells, exhibiting both tumor-suppressive and tumor-promoting functions, has garnered significant attention. Understanding the distinct effects of various B cell subsets on different tumors could pave the way for novel targeted tumor therapies. This review provides a comprehensive overview of the heterogeneous B cell subsets and their multifaceted roles in tumorigenesis, as well as the therapeutic potential of targeting B cells in cancer treatment. To develop more effective cancer immunotherapies, it is essential to decipher the heterogeneity of B cells and their roles in shaping the TME.
Collapse
Affiliation(s)
| | | | | | | | | | - Yingyue Zeng
- School of Life Science, Liaoning University, Shenyang 110036, China; (H.Y.); (Z.Z.); (J.L.); (K.W.); (W.Z.)
| |
Collapse
|
39
|
Xiao F, Zhou D, Cao M, Wu H, Zheng C, Rui K, Lu L. The emerging roles of B cells in cancer development. Cell Mol Immunol 2024; 21:1180-1182. [PMID: 39187635 PMCID: PMC11527892 DOI: 10.1038/s41423-024-01211-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 08/14/2024] [Indexed: 08/28/2024] Open
Affiliation(s)
- Fan Xiao
- Department of Pathology and Shenzhen Institute of Research and Innovation, The University of Hong Kong, Hong Kong, China
| | - Dongmei Zhou
- Department of Rheumatology and Immunology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Meng Cao
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Haijing Wu
- Department of Dermatology, The Second Xiangya Hospital of Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha, 410011, Hunan, China
| | - Chunxing Zheng
- Centre for Oncology and Immunology, Hong Kong Science Park, Hong Kong, China
| | - Ke Rui
- Department of Laboratory Medicine, Institute of Medical Immunology of Jiangsu University, Affiliated Hospital of Jiangsu University, Zhenjiang, China.
| | - Liwei Lu
- Department of Pathology and Shenzhen Institute of Research and Innovation, The University of Hong Kong, Hong Kong, China.
- Centre for Oncology and Immunology, Hong Kong Science Park, Hong Kong, China.
| |
Collapse
|
40
|
Zhou Z, Dong D, Yuan Y, Luo J, Liu XD, Chen LY, Wang G, Yin Y. Single cell atlas reveals multilayered metabolic heterogeneity across tumour types. EBioMedicine 2024; 109:105389. [PMID: 39393173 DOI: 10.1016/j.ebiom.2024.105389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 09/23/2024] [Accepted: 09/24/2024] [Indexed: 10/13/2024] Open
Abstract
BACKGROUND Metabolic reprogramming plays a pivotal role in cancer progression, contributing to substantial intratumour heterogeneity and influencing tumour behaviour. However, a systematic characterization of metabolic heterogeneity across multiple cancer types at the single-cell level remains limited. METHODS We integrated 296 tumour and normal samples spanning six common cancer types to construct a single-cell compendium of metabolic gene expression profiles and identify cell type-specific metabolic properties and reprogramming patterns. A computational approach based on non-negative matrix factorization (NMF) was utilised to identify metabolic meta-programs (MMPs) showing intratumour heterogeneity. In-vitro cell experiments were conducted to confirm the associations between MMPs and chemotherapy resistance, as well as the function of key metabolic regulators. Survival analyses were performed to assess clinical relevance of cellular metabolic properties. FINDINGS Our analysis revealed shared glycolysis upregulation and divergent regulation of citric acid cycle across different cell types. In malignant cells, we identified a colorectal cancer-specific MMP associated with resistance to the cuproptosis inducer elesclomol, validated through in-vitro cell experiments. Furthermore, our findings enabled the stratification of patients into distinct prognostic subtypes based on metabolic properties of specific cell types, such as myeloid cells. INTERPRETATION This study presents a nuanced understanding of multilayered metabolic heterogeneity, offering valuable insights into potential personalized therapies targeting tumour metabolism. FUNDING National Key Research and Development Program of China (2021YFA1300601). National Natural Science Foundation of China (key grants 82030081 and 81874235). The Shenzhen High-level Hospital Construction Fund and Shenzhen Basic Research Key Project (JCYJ20220818102811024). The Lam Chung Nin Foundation for Systems Biomedicine.
Collapse
Affiliation(s)
- Zhe Zhou
- Institute of Systems Biomedicine, Department of Pathology, School of Basic Medical Sciences, Peking University Health Science Centre and School of Life Sciences, Peking University, Beijing 100191, China
| | - Di Dong
- Institute of Systems Biomedicine, Department of Pathology, School of Basic Medical Sciences, Peking University Health Science Centre and School of Life Sciences, Peking University, Beijing 100191, China
| | - Yuyao Yuan
- Institute of Systems Biomedicine, Department of Pathology, School of Basic Medical Sciences, Peking University Health Science Centre and School of Life Sciences, Peking University, Beijing 100191, China
| | - Juan Luo
- Institute of Precision Medicine, Peking University Shenzhen Hospital, Shenzhen 518036, China
| | - Xiao-Ding Liu
- Research Centre for Molecular Pathology, Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100032, China
| | - Long-Yun Chen
- Research Centre for Molecular Pathology, Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100032, China
| | - Guangxi Wang
- Institute of Systems Biomedicine, Department of Pathology, School of Basic Medical Sciences, Peking University Health Science Centre and School of Life Sciences, Peking University, Beijing 100191, China
| | - Yuxin Yin
- Institute of Systems Biomedicine, Department of Pathology, School of Basic Medical Sciences, Peking University Health Science Centre and School of Life Sciences, Peking University, Beijing 100191, China.
| |
Collapse
|
41
|
Cui Y, Wang H, Wang Y. Plasma metabolites as mediators in the relationship between inflammation-related proteins and benign prostatic hyperplasia: insights from mendelian randomization. Sci Rep 2024; 14:26152. [PMID: 39478098 PMCID: PMC11525667 DOI: 10.1038/s41598-024-77515-2] [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] [Accepted: 10/23/2024] [Indexed: 11/02/2024] Open
Abstract
Benign prostatic hyperplasia (BPH) is a condition commonly observed in aging males. Inflammatory and metabolic factors are pivotal in the development and progression of BPH. The degree to which the effects of 91 inflammation-related proteins on BPH are mediated by 1400 plasma metabolites remains ambiguous. Our research analyzed the impact of these traits utilizing genetic evidence.Two-sample Mendelian randomization (MR) and multivariable MR (MVMR) were utilized in our study to infer the genetic causal effect of inflammation-related proteins on BPH, with metabolites serving as mediators. Increased levels of IL-2 were linked to a heightened incidence of BPH (β = 0.071, OR:1.074, 95% CI [1.002-1.152], p = 0.045), whereas lower concentrations of N6,N6-dimethyllysine were associated with decreased risk (β1=-0.127, p = 0.02; β2=-0.039, p = 0.008). The mediation effect was 0.005 (95% CI [0.0004, 0.012], OR: 1.005, 95% CI [1.000, 1.012]), accounting for 7.04% of the total effect. subsequently, we examined the phenotypic co-localization of the two pairings independently, revealing that the posterior probability of rs145516501 associated with IL-2 and BPH was 80.7%, whereas the posterior likelihood of rs4917820 linked to N6,N6-dimethyllysine levels and BPH was 95.9%. The research indicated that N6,N6-dimethyllysine levels seem to influence the causative relationship between IL-2 and BPH. These results elucidate the complex interplay between inflammation-related proteins and metabolism in the context of BPH, offering novel diagnostic and therapeutic avenues and enhancing our comprehension of the disease's etiology for prospective research.
Collapse
Affiliation(s)
- Yan Cui
- Department of Ultrasound, First Affiliated Hospital of Dalian Medical University, Dalian, 116100, Liaoning, China
| | - Hui Wang
- Department of Ultrasound, First Affiliated Hospital of Dalian Medical University, Dalian, 116100, Liaoning, China.
| | - Yuting Wang
- Harbin Medical University, Harbin, 150000, Heilongjiang, China
| |
Collapse
|
42
|
Rouanne M, Chen N, Mariuzza DL, Li F, de Los Santos-Alexis K, Savage TM, Vincent RL, Mendelsohn CL, Danino T, Arpaia N. Tumor-specific antibodies elicited by engineered bacteria promote bladder cancer immunotherapy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.24.620122. [PMID: 39554157 PMCID: PMC11565727 DOI: 10.1101/2024.10.24.620122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
The intratumoral microbiome has recently emerged as a new hallmark of cancer, with implications for response or resistance to therapy. While bacteria can either promote or inhibit cancer growth, intratumoral bacteria can also be engineered using synthetic biology to remodel the tumor microenvironment. Here, we engineered the probiotic bacterium E. coli Nissle 1917 (EcN) to express the human chemokine CXCL13, a critical component of germinal center (GC) formation. The GC reaction is a fundamental aspect of adaptive immunity by which antibody affinity develops in secondary lymphoid organs for defense against pathogens. Using orthotopic models of bladder cancer, engineered CXCL13-expressing EcN colonized bladder tumors and elicited GC responses in bladder tumor-draining lymph nodes after intravesical delivery. Furthermore, when combined with PD-1 blockade, engineered EcN amplified the antitumor antibody response and promoted long-term survival and protective immunity upon tumor rechallenge. Thus, we demonstrate that synthetically engineered CXCL13-expressing EcN can enhance the efficacy of PD-1 checkpoint blockade immunotherapy by amplifying tumor-specific humoral immunity.
Collapse
|
43
|
Wang G, Zhang E, Chen A, Meng D. Single-cell RNA-seq analysis revealed the stemness of a specific cluster of B cells in acute lymphoblastic leukemia progression. PeerJ 2024; 12:e18296. [PMID: 39465162 PMCID: PMC11505884 DOI: 10.7717/peerj.18296] [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: 07/05/2024] [Accepted: 09/22/2024] [Indexed: 10/29/2024] Open
Abstract
Background Childhood acute lymphoblastic leukemia (ALL) is a common pediatric cancer. The heterogeneous characterization of B cells in ALL progression poses new challenges to researchers. We used single-cell sequencing to explore the critical role of B cells in regulating the ALL immune microenvironment. Method We collected the single cell (sc) RNA-seq data of ALL and health sample from the gene expression omnibus (GEO) database, the "Seurat" and "harmony" R package was used for quality control and scRNA-seq analysis, in which the CellMarker2.0 database was used for cell type annotation. Subsequently, the FindAllMarkers function was used to identify the differentially expressed genes (DEGs) among various cell types and the DAVID database was applied for the biological process of DEGs. Then, the "inferCNV" package was used for copy number variation, regulons and cell communication were performed by SCENIC tool and CellChat package. The role of the target gene in regulating ALL progression was assessed using RT-qPCR, Transwell and scratch healing assays. Results We identified nine mainly cell clusters after scRNA-seq analysis, in which the B cells had higher infiltration proportion in the ALL samples and were sub-clustered into five cell sub-groups. The B cells 1 is closely associated with cell proliferation and stemness (TNFAIP3 and KDM5B), and the significant CNV of amplification occurred on chr6 and chr21 that supported stemness of B cells1. RXRB is a key transcription factor mediated the proliferation of B cells 1, which in turn suppressed hematopoietic stem cells (HSCs) proliferation and promoted cytotoxic NK/T cells activation through diverse cell communication ways. One of the key regulators of B cells is MYC, which promotes the migration and invasive ability of cell line leukemia cell lines. Conclusion This study reveals the stemness characteristics of B cells and their critical role in ALL progression, a finding that provides new potential directions for the development of targeted therapies against ALL.
Collapse
Affiliation(s)
- Guifang Wang
- Department of Pediatric Medicine, The Second Affiliated Hospital of Shandong First Medical University, Tai’an, Shandong, China
| | - Ensheng Zhang
- Department of Pediatric Hematology, Shandong Maternal and Child Health Hospital, Jinan, Shandong, China
| | - An Chen
- Department of Otolaryngology, The Second Affiliated Hospital of Shandong First Medical University, Tai’an, Shandong, China
| | - Dachuan Meng
- Department of Pediatric Hematology, Shandong Maternal and Child Health Hospital, Jinan, Shandong, China
| |
Collapse
|
44
|
Lin L, Zou J, Pei S, Huang W, Zhang Y, Zhao Z, Ding Y, Xiao C. Germinal center B-cell subgroups in the tumor microenvironment cannot be overlooked: Their involvement in prognosis, immunotherapy response, and treatment resistance in head and neck squamous carcinoma. Heliyon 2024; 10:e37726. [PMID: 39391510 PMCID: PMC11466559 DOI: 10.1016/j.heliyon.2024.e37726] [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: 03/05/2024] [Revised: 09/09/2024] [Accepted: 09/09/2024] [Indexed: 10/12/2024] Open
Abstract
Background More than 60 % of patients with head and neck squamous carcinoma (HNSCC) are diagnosed at advanced stages and miss radical treatment. This has prompted the need to find new biomarkers to achieve early diagnosis and predict early recurrence and metastasis of tumors. Methods Single-cell RNA sequencing (scRNA-seq) data from HNSCC tissues and peripheral blood samples were obtained through the Gene Expression Omnibus (GEO) database (GSE164690) to characterize the B-cell subgroups, differentiation trajectories, and intercellular communication networks in HNSCC and to construct a prognostic model of the associated risks. In addition, this study analyzed the differences in clinical features, immune cell infiltration, functional enrichment, tumor mutational burden (TMB), and drug sensitivity between the high- and low-risk groups. Results Using scRNA-seq of HNSCC, we classified B and plasma cells into a total of four subgroups: naive B cells (NBs), germinal center B cells (GCBs), memory B cells (MBs), and plasma cells (PCs). Pseudotemporal trajectory analysis revealed that NBs and GCBs were at the early stage of B cell differentiation, while MBs and PCs were at the end. Cellular communication revealed that GCBs acted on tumor cells through the CD99 and SEMA4 signaling pathways. The independent prognostic value, immune cell infiltration, TMB and drug sensitivity assays were validated for the MEF2B+ GCB score groups. Conclusions We identified GCBs as B cell-specific prognostic biomarkers for the first time. The MEF2B+ GCB score fills the research gap in the genetic prognostic prediction model of HNSCC and is expected to provide a theoretical basis for finding new therapeutic targets for HNSCC.
Collapse
Affiliation(s)
- Li Lin
- Department of Stomatology, the First Affiliated Hospital of Soochow University, 188 Shi Zi Rd, Suzhou, 215006, China
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, School of Medicine, Shanghai Jiao Tong University, 639 Zhi Zao Ju Rd, Shanghai, 200011, China
| | - Jiani Zou
- China Eastern Airlines, Comprehensive Management Department, Aviation Health Department, China
| | - Shengbin Pei
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wenyi Huang
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, School of Medicine, Shanghai Jiao Tong University, 639 Zhi Zao Ju Rd, Shanghai, 200011, China
| | - Yichi Zhang
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, School of Medicine, Shanghai Jiao Tong University, 639 Zhi Zao Ju Rd, Shanghai, 200011, China
| | - Zhijie Zhao
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, School of Medicine, Shanghai Jiao Tong University, 639 Zhi Zao Ju Rd, Shanghai, 200011, China
| | - Yantao Ding
- Institute of Dermatology and Department of Dermatology, The First Affiliated Hospital, Anhui Medical University, No. 81 Meishan Road, Hefei, Anhui, 230032, China
- China bKey Laboratory of Dermatology, Anhui Medical University, Ministry of Education, Hefei, Anhui, 230032, China
| | - Can Xiao
- Department of Stomatology, the First Affiliated Hospital of Soochow University, 188 Shi Zi Rd, Suzhou, 215006, China
| |
Collapse
|
45
|
Gong D, Arbesfeld-Qiu JM, Perrault E, Bae JW, Hwang WL. Spatial oncology: Translating contextual biology to the clinic. Cancer Cell 2024; 42:1653-1675. [PMID: 39366372 PMCID: PMC12051486 DOI: 10.1016/j.ccell.2024.09.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 08/01/2024] [Accepted: 09/06/2024] [Indexed: 10/06/2024]
Abstract
Microscopic examination of cells in their tissue context has been the driving force behind diagnostic histopathology over the past two centuries. Recently, the rise of advanced molecular biomarkers identified through single cell profiling has increased our understanding of cellular heterogeneity in cancer but have yet to significantly impact clinical care. Spatial technologies integrating molecular profiling with microenvironmental features are poised to bridge this translational gap by providing critical in situ context for understanding cellular interactions and organization. Here, we review how spatial tools have been used to study tumor ecosystems and their clinical applications. We detail findings in cell-cell interactions, microenvironment composition, and tissue remodeling for immune evasion and therapeutic resistance. Additionally, we highlight the emerging role of multi-omic spatial profiling for characterizing clinically relevant features including perineural invasion, tertiary lymphoid structures, and the tumor-stroma interface. Finally, we explore strategies for clinical integration and their augmentation of therapeutic and diagnostic approaches.
Collapse
Affiliation(s)
- Dennis Gong
- Center for Systems Biology, Department of Radiation Oncology, Center for Cancer Research, Massachusetts General Hospital, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA; Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Jeanna M Arbesfeld-Qiu
- Center for Systems Biology, Department of Radiation Oncology, Center for Cancer Research, Massachusetts General Hospital, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA; Harvard University, Graduate School of Arts and Sciences, Cambridge, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Ella Perrault
- Center for Systems Biology, Department of Radiation Oncology, Center for Cancer Research, Massachusetts General Hospital, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA; Harvard University, Graduate School of Arts and Sciences, Cambridge, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Jung Woo Bae
- Center for Systems Biology, Department of Radiation Oncology, Center for Cancer Research, Massachusetts General Hospital, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - William L Hwang
- Center for Systems Biology, Department of Radiation Oncology, Center for Cancer Research, Massachusetts General Hospital, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA; Harvard University, Graduate School of Arts and Sciences, Cambridge, MA, USA; Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
46
|
Zhao B, Gu Y, Shi D, Chen X, Li Y. Elucidating the molecular markers and biological pathways associated with extrahepatic cholangiocarcinoma: a transcriptome sequencing study. Front Oncol 2024; 14:1417374. [PMID: 39355132 PMCID: PMC11442168 DOI: 10.3389/fonc.2024.1417374] [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: 04/14/2024] [Accepted: 08/26/2024] [Indexed: 10/03/2024] Open
Abstract
Background Cholangiocarcinoma is a malignancy with high aggressiveness, and extrahepatic cholangiocarcinoma (ECCA) represents the predominant subtype. However, the molecular architecture and underlying pathogenic mechanisms of ECCA remain poorly understood. The objective of this study is to elucidate the molecular markers and biological pathways associated with ECCA. Methods In order to identify the factors influencing ECCA, we conducted transcriptome sequencing on a cohort of 8 surgically resected ECCA specimens. To validate our findings, we integrated data from The Cancer Genome Atlas and Gene Expression Omnibus (GEO) databases using batch integration analysis. Finally, we confirmed our results using clinical samples. Results The findings of this study reveal that through the analysis of sequencing data, we have successfully identified the genes that are differentially expressed and have a significant role in the development of ECCA. Utilizing the Weighted Gene Co-expression Network Analysis approach, we have integrated these identified gene modules with the GEO dataset, leading to the identification of four key genes (PTGDS, ITIH2, LSAMP, HBB) that are strongly associated with the progression-free survival of ECCA. We screened a key gene LSAMP from four genes using immunohistochemistry. The gene primarily participate in crucial biological processes such as the ECCA cell cycle and DNA replication. The qRT-PCR reaction and Western Blot conducted on the tissues provided confirmation of the expression levels of the gene, which exhibited consistency with the outcomes of our analysis. Conclusions Our study has successfully identified potential biomarkers LSAMP for ECCA, which can serve as valuable tools for early detection and targeted therapeutic interventions in clinical settings.
Collapse
Affiliation(s)
- Bin Zhao
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, China
| | - Yanmei Gu
- Department of the Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Daixiu Shi
- Department of Nursing, Lanzhou University Second Hospital, Lanzhou, China
| | - Xiaokang Chen
- Department of the Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Yumin Li
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, China
| |
Collapse
|
47
|
Peng X, He Z, Yuan D, Liu Z, Rong P. Lactic acid: The culprit behind the immunosuppressive microenvironment in hepatocellular carcinoma. Biochim Biophys Acta Rev Cancer 2024; 1879:189164. [PMID: 39096976 DOI: 10.1016/j.bbcan.2024.189164] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 07/25/2024] [Accepted: 07/28/2024] [Indexed: 08/05/2024]
Abstract
As a solid tumor with high glycolytic activity, hepatocellular carcinoma (HCC) produces excess lactic acid and increases extracellular acidity, thus forming a unique immunosuppressive microenvironment. L-lactate dehydrogenase (LDH) and monocarboxylate transporters (MCTs) play a very important role in glycolysis. LDH is the key enzyme for lactic acid (LA) production, and MCT is responsible for the cellular import and export of LA. The synergistic effect of the two promotes the formation of an extracellular acidic microenvironment. In the acidic microenvironment of HCC, LA can not only promote the proliferation, survival, transport and angiogenesis of tumor cells but also have a strong impact on immune cells, ultimately leading to an inhibitory immune microenvironment. This article reviews the role of LA in HCC, especially its effect on immune cells, summarizes the progress of LDH and MCT-related drugs, and highlights the potential of immunotherapy targeting lactate combined with HCC.
Collapse
Affiliation(s)
- Xiaopei Peng
- Department of Radiology, the Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China; Molecular Imaging Research Center, Central South University, Changsha, Hunan 410013, China
| | - Zhenhu He
- Department of Radiology, the Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China; Molecular Imaging Research Center, Central South University, Changsha, Hunan 410013, China
| | - Dandan Yuan
- Department of Radiology, the Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China; Molecular Imaging Research Center, Central South University, Changsha, Hunan 410013, China
| | - Zhenguo Liu
- Department of Infectious Disease, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China
| | - Pengfei Rong
- Department of Radiology, the Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China; Molecular Imaging Research Center, Central South University, Changsha, Hunan 410013, China.
| |
Collapse
|
48
|
Cai X, Yang J, Guo Y, Yu Y, Zheng C, Dai X. Re-analysis of single cell and spatial transcriptomics data reveals B cell landscape in gastric cancer microenvironment and its potential crosstalk with tumor cells for clinical prognosis. J Transl Med 2024; 22:807. [PMID: 39215354 PMCID: PMC11365245 DOI: 10.1186/s12967-024-05606-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2024] [Accepted: 08/14/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND At present, immunotherapy has become a powerful treatment for advanced gastric cancer (AGC), but not all patients can benefit from it. According to the latest research, the impact of B cell subpopulations on the immune microenvironment of gastric cancer (GC) is unknown. Exploring whether the interaction between B cells and tumor cells in GC affects the effectiveness of immunotherapy has attracted our interest. METHODS This study involved the re-analysis of single-cell RNA (scRNA) and spatial transcriptomics (ST) data from publicly available datasets. The focus was on investigating the subpopulations and differentiation trajectories of B cells in the gastric cancer (GC) tumor immune microenvironment (TIME). Spatial transcriptomics (ST) and multiple immunofluorescence (mIF) revealed a clear co-localization pattern between B cells and tumor cells. Multiple immunotherapy datasets were collected to identify unique immunotherapy biomarkers. The unique immunotherapeutic potential of targeting CCL28 was validated through a mouse gastric cancer model. In addition, flow cytometry revealed changes in the tumor immune microenvironment targeting CCL28. RESULTS The re-analysis of ST data from multiple cancer types revealed a co-localization pattern between B cells and tumor cells. A significant number of IgA plasma cells were identified in the GC TIME. Five different tumor-infiltrating B cell subpopulations and two unique B cell differentiation trajectories were characterized, along with seven GC-related states. By analyzing the communication between GC cells and B cells, it was further discovered that tumor cells can influence and recruit plasma cells through CCL28-CCR10 signaling. Additionally, there was a crosstalk between GC cells and B cells. Finally, we identified the LAMA/CD44 signaling axis as a potential prognostic marker for immunotherapy through a large amount of immunotherapy data. We also validated through various animal tumor models that targeting CCL28 can significantly promote CD8+T cell infiltration and function in the TME by regulating B cell and plasma cell functions, and has the ability to synergize immunotherapy. CONCLUSION The co-localization and crosstalk between GC cells and B cells significantly affect the efficacy of immunotherapy, and inhibiting the CCL28-CCR10 signal axis is a potential immunotherapy target for GC. Meanwhile, LAMA/CD44 pair may be a potential adverse indicator for immunotherapy and tumor prognosis.
Collapse
Affiliation(s)
- Xing Cai
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China
| | - Jinru Yang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, People's Republic of China
| | - Yusheng Guo
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China
- Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, People's Republic of China
| | - Yanchao Yu
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China
| | - Chuansheng Zheng
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China.
- Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, People's Republic of China.
| | - Xiaofang Dai
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China.
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China.
| |
Collapse
|
49
|
Hollern D. Memory B cell fitness and anergy has significant links to cancer lethality. Cell 2024; 187:4551-4553. [PMID: 39178833 DOI: 10.1016/j.cell.2024.07.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 07/19/2024] [Accepted: 07/22/2024] [Indexed: 08/26/2024]
Abstract
Two recent studies reveal that the extent of fitness or anergy in tumor-associated memory B cells is vital to anti-tumor immune response, cancer patient survival, and response to immune therapy. The impact of these seminal findings demonstrates the untapped potential for using B cells to combat the lethality of cancer.
Collapse
Affiliation(s)
- Daniel Hollern
- Nomis Center for Immunobiology and Microbial Pathogenesis, Salk Cancer Center, Salk Institute for Biological Studies, La Jolla, CA, USA; School of Biological Sciences and Department of Radiation Medicine and Applied Sciences, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
50
|
Yang Y, Chen X, Pan J, Ning H, Zhang Y, Bo Y, Ren X, Li J, Qin S, Wang D, Chen MM, Zhang Z. Pan-cancer single-cell dissection reveals phenotypically distinct B cell subtypes. Cell 2024; 187:4790-4811.e22. [PMID: 39047727 DOI: 10.1016/j.cell.2024.06.038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 04/25/2024] [Accepted: 06/26/2024] [Indexed: 07/27/2024]
Abstract
Characterizing the compositional and phenotypic characteristics of tumor-infiltrating B cells (TIBs) is important for advancing our understanding of their role in cancer development. Here, we establish a comprehensive resource of human B cells by integrating single-cell RNA sequencing data of B cells from 649 patients across 19 major cancer types. We demonstrate substantial heterogeneity in their total abundance and subtype composition and observe immunoglobulin G (IgG)-skewness of antibody-secreting cell isotypes. Moreover, we identify stress-response memory B cells and tumor-associated atypical B cells (TAABs), two tumor-enriched subpopulations with prognostic potential, shared in a pan-cancer manner. In particular, TAABs, characterized by a high clonal expansion level and proliferative capacity as well as by close interactions with activated CD4 T cells in tumors, are predictive of immunotherapy response. Our integrative resource depicts distinct clinically relevant TIB subsets, laying a foundation for further exploration of functional commonality and diversity of B cells in cancer.
Collapse
Affiliation(s)
- Yu Yang
- Biomedical Pioneering Innovation Center (BIOPIC), Academy for Advanced Interdisciplinary Studies, and School of Life Sciences, Peking University, Beijing 100871, China
| | - Xueyan Chen
- Biomedical Pioneering Innovation Center (BIOPIC), Academy for Advanced Interdisciplinary Studies, and School of Life Sciences, Peking University, Beijing 100871, China
| | - Jieying Pan
- Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen 518132, China
| | - Huiheng Ning
- Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen 518132, China
| | - Yaojun Zhang
- State Key Laboratory of Oncology in South China, Department of Liver Surgery, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Yufei Bo
- Biomedical Pioneering Innovation Center (BIOPIC), Academy for Advanced Interdisciplinary Studies, and School of Life Sciences, Peking University, Beijing 100871, China
| | - Xianwen Ren
- Biomedical Pioneering Innovation Center (BIOPIC), Academy for Advanced Interdisciplinary Studies, and School of Life Sciences, Peking University, Beijing 100871, China
| | - Jiesheng Li
- Biomedical Pioneering Innovation Center (BIOPIC), Academy for Advanced Interdisciplinary Studies, and School of Life Sciences, Peking University, Beijing 100871, China
| | - Shishang Qin
- Biomedical Pioneering Innovation Center (BIOPIC), Academy for Advanced Interdisciplinary Studies, and School of Life Sciences, Peking University, Beijing 100871, China
| | - Dongfang Wang
- Biomedical Pioneering Innovation Center (BIOPIC), Academy for Advanced Interdisciplinary Studies, and School of Life Sciences, Peking University, Beijing 100871, China.
| | - Min-Min Chen
- Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen 518132, China.
| | - Zemin Zhang
- Biomedical Pioneering Innovation Center (BIOPIC), Academy for Advanced Interdisciplinary Studies, and School of Life Sciences, Peking University, Beijing 100871, China.
| |
Collapse
|