1
|
Fu Y, Yang Q, Xu N, Zhang X. MiRNA affects the advancement of breast cancer by modulating the immune system's response. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167759. [PMID: 40037267 DOI: 10.1016/j.bbadis.2025.167759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 02/05/2025] [Accepted: 02/26/2025] [Indexed: 03/06/2025]
Abstract
Breast cancer (BC), which is the most common tumor in women, has greatly endangered women's lives and health. Currently, patients with BC receive comprehensive treatments, including surgery, chemotherapy, radiotherapy, endocrine therapy, and targeted therapy. According to the latest research, the development of BC is closely related to the inflammatory immune response, and the immunogenicity of BC has steadily been recognized. As such, immunotherapy is one of the promising and anticipated forms of treatment for BC. The potential values of miRNA in the diagnosis and prognosis of BC have been established, and aberrant expression of associated miRNA can either facilitate or inhibit progression of BC. In the tumor immune microenvironment (TME), miRNAs are considered to be an essential molecular mechanism by which tumor cells interact with immunocytes and immunologic factors. Aberrant expression of miRNAs results in reprogramming of tumor cells actively, which may suppress the generation and activation of immunocytes and immunologic factors, avoid tumor cells apoptosis, and ultimately result in uncontrolled proliferation and deterioration. Therefore, through activating and regulating the immunocytes related to tumors and associated immunologic factors, miRNA can contribute to the advancement of BC. In this review, we assessed the function of miRNA and associated immune system components in regulating the advancement of BC, as well as the potential and viability of using miRNA in immunotherapy for BC.
Collapse
Affiliation(s)
- Yeqin Fu
- Zhejiang cancer hospital, Hangzhou, Zhejiang 310022, China; Postgraduate Training Base Alliance of Wenzhou Medical University (Zhejiang Cancer Hospital), Hangzhou, Zhejiang 310022, China
| | - Qiuhui Yang
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), 310006, China
| | - Ning Xu
- Zhejiang cancer hospital, Hangzhou, Zhejiang 310022, China; School of Medicine, Shaoxing University, Shaoxing, Zhejiang 312000, China
| | - Xiping Zhang
- Zhejiang cancer hospital, Hangzhou, Zhejiang 310022, China.
| |
Collapse
|
2
|
Wang K, Lu Y, Cao Y, Feng P, Wu Q, Xiao P, Ding Y. Establishment and validation of an immune-related nomogram for the prognosis of pancreatic adenocarcinoma. Sci Rep 2025; 15:13431. [PMID: 40251364 PMCID: PMC12008212 DOI: 10.1038/s41598-025-98503-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Accepted: 04/11/2025] [Indexed: 04/20/2025] Open
Abstract
Pancreatic adenocarcinoma (PDAC) is a highly aggressive neoplasm characterized by limited therapeutic options, particularly in the realm of immunotherapy. This study aims to improve prognosis prediction to guide therapeutic decision-making, and to identify novel targets for immunotherapy of PDAC. We conducted Cox and LASSO regression analyses to develop immune-related gene signature and corresponding nomogram, and the robustness of these signatures was demonstrated using multiple approaches. Additionally, CIBERSORT, ESTIMATE, and xCell algorithms were utilized to assess immune cell infiltration, with experimental validation performed though qPCR. An immune-related gene signature consisting of 18 genes, and the prognostic nomogram was established with superior performance compared to the conventional staging system. Key parameters incorporated into the nomogram included the gene signature, tumor stage, and postoperative treatment. Patients identified as high-risk exhibited an anti-inflammatory tumor microenvironment, characterized by an increase in M2-like tumor-associated macrophages and heightened tumor purity. Notably, the expression of interleukin 6 receptor (IL6R) in PDAC was predominantly derived from macrophages and was significantly associated with patient survival outcomes. Furthermore, attenuated IL-6/IL-6R signaling was found to promote M2-like macrophage differentiation. This study successfully established an immune-related gene signature and a robust nomogram for predicting clinical outcomes in patients with PDAC. Furthermore, we identified IL6R as a promising target for future immunotherapeutic strategies.
Collapse
Affiliation(s)
- Kan Wang
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China
| | - Yunkun Lu
- Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China
| | - Yanfei Cao
- Department of Gastroenterology, The Third Affiliated Hospital of Zhejiang University of Traditional Chinese Medicine, Hangzhou, 310000, China
| | - Ping Feng
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China
| | - Qiu Wu
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China
| | - Peng Xiao
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China
| | - Yimin Ding
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China.
| |
Collapse
|
3
|
Sun YG, Yu ZK, Chen X, Zhang SY, Wu WJ, Liu K, Cheng L. circHIPK2 promotes malignant progression of laryngeal squamous cell carcinoma through the miR-889-3p/MCTS1/IL-6 axis. Transl Oncol 2025; 56:102390. [PMID: 40222337 PMCID: PMC12018564 DOI: 10.1016/j.tranon.2025.102390] [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: 08/19/2024] [Revised: 03/31/2025] [Accepted: 04/05/2025] [Indexed: 04/15/2025] Open
Abstract
Laryngeal squamous cell carcinoma (LSCC) is a common malignant tumor of the head and neck with a poor prognosis. The role of circRNAs in LSCC remains largely unknown. In this study, quantitative real-time PCR (qRT-PCR), Sanger sequencing and fluorescence in situ hybridization were undertaken to detect the expression, localization, and clinical significance of circHIPK2 in LSCC tissues and TU686 and TU212 cells. The functions of circHIPK2 in LSCC were explored through proliferation analysis, EdU staining, colony formation assay, wound healing assay, and Transwell assay. The regulatory mechanisms underpinning circHIPK2, miR-889-3p, and MCTS1 were investigated using luciferase assay, Western blotting, and qRT-PCR. We found that LSCC tissues and cells demonstrated high expression of circHIPK2 that was closely associated with the malignant progression and poor prognosis of LSCC. Knockdown of circHIPK2 inhibited the proliferation and migration of LSCC cells in vitro. Mechanistic studies showed that circHIPK2 competitively bound to miR-889-3p, elevated MCTS1 level, promoted IL-6 secretion, and ultimately accelerated the malignant progression of LSCC. In conclusion, an axis involving circHIPK2, miR-889-3p, MCTS1 and IL-6 regulates the malignant progression of LSCC. circHIPK2 expression may serve as a novel diagnostic and prognostic biomarker for LSCC.
Collapse
Affiliation(s)
- Yang-Guang Sun
- Department of Otorhinolaryngology, The First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
| | - Zhen-Kun Yu
- Nanjing Medical Key Laboratory of Laryngopharynx-Head & Neck Oncology, Department of Otolaryngology-Head and Neck Surgery, BenQ Medical Center, The Affiliated BenQ Hospital of Nanjing Medical University, Nanjing 210019, China
| | - Xi Chen
- Department of Otorhinolaryngology, The First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
| | - Si-Yao Zhang
- Nanjing Medical Key Laboratory of Laryngopharynx-Head & Neck Oncology, Department of Otolaryngology-Head and Neck Surgery, BenQ Medical Center, The Affiliated BenQ Hospital of Nanjing Medical University, Nanjing 210019, China
| | - Wan-Juan Wu
- Nanjing Medical Key Laboratory of Laryngopharynx-Head & Neck Oncology, Department of Otolaryngology-Head and Neck Surgery, BenQ Medical Center, The Affiliated BenQ Hospital of Nanjing Medical University, Nanjing 210019, China
| | - Kai Liu
- Nanjing Medical Key Laboratory of Laryngopharynx-Head & Neck Oncology, Department of Otolaryngology-Head and Neck Surgery, BenQ Medical Center, The Affiliated BenQ Hospital of Nanjing Medical University, Nanjing 210019, China
| | - Lei Cheng
- Department of Otorhinolaryngology, The First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China; Department of Allergology, The First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China.
| |
Collapse
|
4
|
Zhou J, Cheng A, Guo J, Liu Y, Li X, Chen M, Hu D, Wu J. Targeting PRDX2 to inhibit tumor growth and metastasis in triple-negative breast cancer: the role of FN1 and the PI3K/AKT/SP1 pathway. J Transl Med 2025; 23:434. [PMID: 40217291 PMCID: PMC11992733 DOI: 10.1186/s12967-025-06441-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Accepted: 03/28/2025] [Indexed: 04/14/2025] Open
Abstract
BACKGROUND Triple-negative breast cancer (TNBC) is characterized by high invasiveness, high potential for metastasis, easy recurrence, and poor prognosis. There is an urgent need to develop new clinical treatments. METHODS This study utilized TNBC tissue microarrays to detect Peroxiredoxin 2 (PRDX2) expression levels and analyzed the correlation between PRDX2 and tumor invasion as well as invasion-related gene expression through the TCGA database. A stable PRDX2-knockdown triple-negative breast cancer cell line was established using lentiviral technology. The effects of PRDX2 on triple-negative breast cancer cell migration, invasion, and epithelial-mesenchymal transition (EMT) were investigated via wound healing assays, Transwell assays, qPCR, and Western blotting. RNA sequencing (RNA-seq), Western blotting, and dual luciferase reporter assays were performed to confirm that PRDX2 regulates FN1 expression through SP1. Furthermore, subcutaneous tumor xenograft models in nude mice were constructed to assess the effects of PRDX2 knockdown and the PRDX2 inhibitor Conoidin A on tumor growth in vivo. RESULTS Tissue microarray detection and correlative analysis revealed that PRDX2 is significantly upregulated in triple-negative breast cancer (TNBC) tumor tissues and positively correlated with genes associated with cell migration and invasion. Functional experiments demonstrated that in vitro knockdown of PRDX2 suppresses migration, invasion, and epithelial-mesenchymal transition (EMT) in TNBC cells. Furthermore, in vivo knockdown of PRDX2 or treatment with the PRDX2 inhibitor Conoidin A effectively reduced tumor burden. Mechanistic investigations utilizing RNA sequencing (RNA-seq) identified FN1 as a critical gene promoting TNBC cell migration and invasion. PRDX2 facilitates TNBC progression by activating the PI3K/AKT signaling pathway, which enhances SP1 binding to the FN1 gene promoter. This regulatory cascade ultimately drives tumor advancement in TNBC. CONCLUSIONS This study elucidates the role of the PRDX2/SP1/FN1 axis in TNBC migration and invasion, and highlights PRDX2 as a promising therapeutic target for triple-negative breast cancer.
Collapse
Affiliation(s)
- Jiawei Zhou
- School of Medicine, Anhui University of Science and Technology, Huainan, Anhui, 232000, China
- School of Medicine, The First Affiliated Hospital of Anhui University of Science and Technology Huainan First People's Hospital, Huainan, Anhui, 232000, China
- Department of Laboratory Medicine, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui, 232001, China
| | - Anqi Cheng
- School of Medicine, Anhui University of Science and Technology, Huainan, Anhui, 232000, China
- School of Medicine, The First Affiliated Hospital of Anhui University of Science and Technology Huainan First People's Hospital, Huainan, Anhui, 232000, China
| | - Jianqiang Guo
- School of Medicine, Anhui University of Science and Technology, Huainan, Anhui, 232000, China
- School of Medicine, The First Affiliated Hospital of Anhui University of Science and Technology Huainan First People's Hospital, Huainan, Anhui, 232000, China
| | - Yafeng Liu
- School of Medicine, Anhui University of Science and Technology, Huainan, Anhui, 232000, China
- School of Medicine, The First Affiliated Hospital of Anhui University of Science and Technology Huainan First People's Hospital, Huainan, Anhui, 232000, China
| | - Xuan Li
- School of Medicine, Anhui University of Science and Technology, Huainan, Anhui, 232000, China
- School of Medicine, The First Affiliated Hospital of Anhui University of Science and Technology Huainan First People's Hospital, Huainan, Anhui, 232000, China
| | - Maoqian Chen
- School of Medicine, Anhui University of Science and Technology, Huainan, Anhui, 232000, China
- School of Medicine, The First Affiliated Hospital of Anhui University of Science and Technology Huainan First People's Hospital, Huainan, Anhui, 232000, China
| | - Dong Hu
- School of Medicine, Anhui University of Science and Technology, Huainan, Anhui, 232000, China.
- School of Medicine, The First Affiliated Hospital of Anhui University of Science and Technology Huainan First People's Hospital, Huainan, Anhui, 232000, China.
- Anhui Occupational Health and Safety Engineering Laboratory, Huainan, Anhui, 232000, China.
- Key Laboratory of Industrial Dust Deep Reduction and Occupational Health and Safety of Anhui Higher Education Institutes, Huainan, Anhui, 232000, China.
- Key Laboratory of Industrial Dust Prevention and Control & Occupational Safety and Health of the Ministry of Education, Anhui University of Science and Technology, Huainan, Anhui, 232000, China.
- Department of Laboratory Medicine, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui, 232001, China.
- , 168 Taifeng Street, Huainan, 232001, China.
| | - Jing Wu
- School of Medicine, The First Affiliated Hospital of Anhui University of Science and Technology Huainan First People's Hospital, Huainan, Anhui, 232000, China.
- Anhui Occupational Health and Safety Engineering Laboratory, Huainan, Anhui, 232000, China.
- Key Laboratory of Industrial Dust Deep Reduction and Occupational Health and Safety of Anhui Higher Education Institutes, Huainan, Anhui, 232000, China.
- Key Laboratory of Industrial Dust Prevention and Control & Occupational Safety and Health of the Ministry of Education, Anhui University of Science and Technology, Huainan, Anhui, 232000, China.
- Department of Laboratory Medicine, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui, 232001, China.
| |
Collapse
|
5
|
Richard V, Lee K, Kerin MJ. MicroRNAs as Endocrine Modulators of Breast Cancer. Int J Mol Sci 2025; 26:3449. [PMID: 40244378 PMCID: PMC11989600 DOI: 10.3390/ijms26073449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2025] [Revised: 04/03/2025] [Accepted: 04/04/2025] [Indexed: 04/18/2025] Open
Abstract
Breast cancer is an aggressive disease of multiple subtypes with varying phenotypic, hormonal, and clinicopathological features, offering enhanced resistance to conventional therapeutic regimens. There is an unmet need for reliable molecular biomarkers capable of detecting the malignant transformation from the early stages of the disease to enhance diagnosis and treatment outcomes. A subset of small non-coding nucleic acid molecules, micro ribonucleic acids (microRNAs/miRNAs), have emerged as promising biomarkers due to their role in gene regulation and cancer pathogenesis. This review discusses, in detail, the different origins and hormone-like regulatory functionalities of miRNAs localized in tumor tissue and in the circulation, as well as their inherent stability and turnover that determines the utility of miRNAs as biomarkers for disease detection, monitoring, prognosis, and therapeutic targets.
Collapse
Affiliation(s)
- Vinitha Richard
- Discipline of Surgery, Lambe Institute for Translational Research, University of Galway, H91 V4AY Galway, Ireland
| | - Kevin Lee
- School of Medicine, University of Galway, H91 V4AY Galway, Ireland;
| | - Michael Joseph Kerin
- Discipline of Surgery, Lambe Institute for Translational Research, University of Galway, H91 V4AY Galway, Ireland
- School of Medicine, University of Galway, H91 V4AY Galway, Ireland;
| |
Collapse
|
6
|
Dai S, Li B, Wu Q, Han S, Zhao Q, Wang Y, Zhang Y, Gao Y. Pan-cancer analysis reveals BAF complexes as immune-related biomarkers and validation in triple-negative breast cancer. Life Sci 2025; 372:123607. [PMID: 40194763 DOI: 10.1016/j.lfs.2025.123607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 03/11/2025] [Accepted: 04/01/2025] [Indexed: 04/09/2025]
Abstract
AIMS BAF complexes (BAFs), ATP-dependent regulators of chromatin structure, play a significant role in cancer progression. This pan-cancer study aimed to decode the potential of specific BAFs in the pathology, immunity, and therapy of targeted cancers. MATERIALS AND METHODS Data were retrieved from The Cancer Genome Atlas, Gene Expression Omnibus, and IMvigor210 databases and were analyzed for expression patterns, prognostic value, mutational signatures, biological pathways, tumor immune microenvironment (TIME) remodeling, and therapeutic resistance of BAFs. Experimental validation was also conducted. KEY FINDINGS BAFs exhibit abnormal expression in various human cancers. The BAFs model and nomogram (based on multiple variables) were developed as prognostic tools. BAFs regulate the TIME and influence the response to anti-PD-L1 therapy, particularly through ACTL6A, as observed in RNA sequencing and single-cell RNA sequencing datasets (high-resolution gene expression data at the single-cell level). ACTLA6 is a major adverse gene in the prognostic model. Patients with high ACTL6A expression showed significantly worse overall survival (hazard ratio = 1.32, 95 % CI: 1.26-1.39, p < 0.001). ACTL6A expression escalates with breast cancer (BRCA) malignancy, particularly in triple-negative BRCA (TNBC), and correlates with immune checkpoint expression while playing a crucial role in promoting cancer metastasis in TNBC. SIGNIFICANCE Our findings first emphasize the significance of a novel BAFs model for patient prognosis and corroborate the considerable role of BAFs as immune-related biomarkers in pan-cancer progression. ACTL6A has a dual role as an immune-related biomarker and potential therapeutic target in TNBC, deepening our comprehension of its function as an oncogene.
Collapse
Affiliation(s)
- Shuying Dai
- Zhejiang Key Laboratory of Traditional Chinese Medicine for the Prevention and Treatment of Senile Chronic Diseases, Department of Geriatrics, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Zhejiang 310006, China
| | - Bei Li
- Zhejiang Key Laboratory of Traditional Chinese Medicine for the Prevention and Treatment of Senile Chronic Diseases, Department of Geriatrics, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Zhejiang 310006, China
| | - Qingqian Wu
- Zhejiang Key Laboratory of Traditional Chinese Medicine for the Prevention and Treatment of Senile Chronic Diseases, Department of Geriatrics, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Zhejiang 310006, China
| | - Shuang Han
- Zhejiang Key Laboratory of Traditional Chinese Medicine for the Prevention and Treatment of Senile Chronic Diseases, Department of Geriatrics, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Zhejiang 310006, China; School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China.
| | - Qingwen Zhao
- Zhejiang Key Laboratory of Traditional Chinese Medicine for the Prevention and Treatment of Senile Chronic Diseases, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Zhejiang 310006, China.
| | - Yule Wang
- Zhejiang Key Laboratory of Traditional Chinese Medicine for the Prevention and Treatment of Senile Chronic Diseases, Department of Geriatrics, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Zhejiang 310006, China
| | - Yingjuan Zhang
- Zhejiang Key Laboratory of Traditional Chinese Medicine for the Prevention and Treatment of Senile Chronic Diseases, Department of Geriatrics, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Zhejiang 310006, China
| | - Yue Gao
- Zhejiang Key Laboratory of Traditional Chinese Medicine for the Prevention and Treatment of Senile Chronic Diseases, Department of Geriatrics, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Zhejiang 310006, China.
| |
Collapse
|
7
|
TIAN Y, BU H, WANG T, YANG D, ZHANG W, LIU T, ZHANG L, HUO Z. Efficacy of electro-acupuncture at "Weizhong" (BL40) on macrophage polarization in rats with injured lumbar multifidus. J TRADIT CHIN MED 2025; 45:335-347. [PMID: 40151120 PMCID: PMC11955755 DOI: 10.19852/j.cnki.jtcm.20220419.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Accepted: 03/17/2022] [Indexed: 03/29/2025]
Abstract
OBJECTIVE To investigate the anti-inflammatory effect of electroacupuncture in rats with bupivacaine-induced lumbar multifidus injury and its underlying regulatory mechanism on macrophage polarization. METHODS A total of seventy-two Sprague-Dawley male rats were randomly divided into control, model, and electroacupuncture groups. Forty-eight rats categorized in model groups were injected 0.5% bupivacaine (BPVC) into the lumbar multifidus at the L4-L5 segment. Rats in the electroacupuncture groups received the intervention for 1, 2, 3 and 5 d, respectively. The degree of macrophage infiltration and change of M1/M2 polarization were observed based on hematoxylin and eosin staining, immunohistochemistry and immunofluorescence to evaluate the anti-inflammatory effect of electroacupuncture. Meanwhile, exosomal miRNA-sequencing and bioinformatics analysis predicted the pathways and biological processes related to inflammatory response and macrophage polarization regulated by electroacupuncture intervention. RESULTS BPVC injection induced the infiltration of local macrophages at the L4-L5 segment of lumbar multifidus. Comparison of mean IOD values with 2 d and 5 d post injury revealed the highest expression of CD68+ macrophages on day 3 post injury by immunohistochemistry. (P < 0.001, P < 0.001, respectively). Compared with the model group, the cell counts of iNOs+ CD68+ M1-macrophages were lower in the electroacupuncture group, while the positive percent of CD163+ CD206+ M2-macrophages was higher in the electroacupuncture group, on day 3 after BPVC injection (P < 0.001, P < 0.001, respectively). Moreover, the results of sequencing and bioinformatic analysis suggested that exosomal miRNAs were involved in the EA regulating macrophage polarization. CONCLUSIONS Electroacupuncture can promote macrophage polarization to reduce inflammation following lumbar multifidus muscular injury.
Collapse
Affiliation(s)
- Yuan TIAN
- 1 Department of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing 100029, China
- 2 Department of Acupuncture-Moxibustion and Tuina, the Second Affiliated Hospital of Baotou Medical College, Inner Mongolia Autonomous Region, Baotou 014030, China
| | - He BU
- 2 Department of Acupuncture-Moxibustion and Tuina, the Second Affiliated Hospital of Baotou Medical College, Inner Mongolia Autonomous Region, Baotou 014030, China
| | - Tieshan WANG
- 3 Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | | | - Wei ZHANG
- 7 Department of Pathology, the First Affiliated Hospital of Baotou Medical College, Inner Mongolia University of Science and Technology, Inner Mongolia 014010, China
| | - Tong LIU
- 5 the Fifth College of Clinical Medicine, Guangzhou University of Traditional Chinese Medicine, Guangzhou 510006, China
- 6 Department of Acupuncture and Rehabilitation, Guangdong Second Hospital of Traditional Chinese Medicine, Guangzhou 510095, China
| | - Li ZHANG
- 1 Department of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Zejun HUO
- 8 Department of Chinese Medicine, Peking University 3rd Hospital, Beijing 100191, China
| |
Collapse
|
8
|
Jiang R, Yang L, Liu X, Xu Y, Han L, Chen Y, Gao G, Wang M, Su T, Li H, Fang L, Sun N, Du H, Zheng J, Wang G. Genetically engineered macrophages reverse the immunosuppressive tumor microenvironment and improve immunotherapeutic efficacy in TNBC. Mol Ther 2025:S1525-0016(25)00198-4. [PMID: 40119517 DOI: 10.1016/j.ymthe.2025.03.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 01/21/2025] [Accepted: 03/17/2025] [Indexed: 03/24/2025] Open
Abstract
The main challenges in current immunotherapy for triple-negative breast cancer (TNBC) lie in the immunosuppressive tumor microenvironment (TME). Considering tumor-associated macrophages (TAMs) are the most abundant immune cells in the TME, resetting TAMs is a promising strategy for ameliorating the immunosuppressive TME. Here, we developed genetically engineered macrophages (GEMs) with gene-carrying adenoviruses, to maintain the M1-like phenotype and directly deliver the immune regulators interleukin-12 and CXCL9 into local tumors, thereby reversing the immunosuppressive TME. In tumor-bearing mice, GEMs demonstrated targeted enrichment in tumors and successfully reprogramed TAMs to M1-like macrophages. Moreover, GEMs significantly enhanced the accumulation, proliferation, and activation of CD8+ T cells, mature dendritic cells, and natural killer cells within tumors, while diminishing M2-like macrophages, immunosuppressive myeloid-derived suppressor cells, and regulatory T cells. This treatment efficiently suppressed tumor growth. In addition, combination therapy with GEMs and anti-programmed cell death protein 1 further improved interferon-γ+CD8+ T cell percentages and tumor inhibition efficacy in an orthotopic murine TNBC model. Therefore, this study provides a novel strategy for reversing the immunosuppressive TME and improving immunotherapeutic efficacy through live macrophage-mediated gene delivery.
Collapse
Affiliation(s)
- Ranran Jiang
- Department of Oncology, Nanjing Medical University, 101 Longmian Avenue, Nanjing, Jiangsu 211166, China; Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Department of Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China
| | - Liechi Yang
- Department of Neurosurgery, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China
| | - Xin Liu
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Department of Urology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China
| | - Yujun Xu
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China
| | - Lulu Han
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China
| | - Yuxin Chen
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China
| | - Ge Gao
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China
| | - Meng Wang
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China
| | - Tong Su
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China
| | - Huizhong Li
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China
| | - Lin Fang
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China
| | - Nan Sun
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China
| | - Hongwei Du
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China
| | - Junnian Zheng
- Department of Oncology, Nanjing Medical University, 101 Longmian Avenue, Nanjing, Jiangsu 211166, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China.
| | - Gang Wang
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China.
| |
Collapse
|
9
|
Ozasa T, Nakajima M, Tsunedomi R, Goto S, Adachi K, Takahashi H, Tamada K, Nagano H. Novel immune drug combination induces tumour microenvironment remodelling and reduces the dosage of anti-PD-1 antibody. Sci Rep 2025; 15:8956. [PMID: 40089538 PMCID: PMC11910518 DOI: 10.1038/s41598-025-87344-6] [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: 08/21/2024] [Accepted: 01/17/2025] [Indexed: 03/17/2025] Open
Abstract
Immune checkpoint inhibitors (ICIs) are effective in clinical settings; however, they present immune-related adverse effects and financial burden. Although dose reduction of ICIs may mitigate these limitations, it could compromise therapeutic efficacy. Using two adjuvants (poly(I:C) and LAG-3-Ig) combined with three neoantigen peptides (Comb), we examined whether Comb could enhance the efficacy of reduced dose of αPD-1 monoclonal antibody (RD-αPD-1 mAb), which has limited efficacy. In a murine colorectal cancer model using an MC38 cell line, Comb addition to RD-αPD-1 mAb enhanced treatment efficacy. Analysis of the tumour microenvironment (TME) in mice treated with Comb using flow cytometry and single-cell RNA sequencing revealed decreased macrophages with highly expressing immunosuppressive genes and increased plasmacytoid dendritic cells with highly expressing antigen-presenting genes. A potent infiltration of CD8+ tumour-infiltrating lymphocytes (TILs) with an effector profile was only observed in RD-αPD-1 mAb with Comb. Additionally, single-cell T cell receptor repertoire analysis underscored an oligoclonal expansion of CD8+ TILs following treatment with RD-αPD-1 mAb with Comb. This novel immune drug combination may be a promising strategy for reducing αPD-1 mAb dosage while preserving antitumour efficacy through modulating the TME.
Collapse
Affiliation(s)
- Takahiro Ozasa
- Department of Gastroenterological, Breast and Endocrine Surgery, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi, 755-8505, Japan
| | - Masao Nakajima
- Department of Gastroenterological, Breast and Endocrine Surgery, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi, 755-8505, Japan
| | - Ryouichi Tsunedomi
- Department of Gastroenterological, Breast and Endocrine Surgery, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi, 755-8505, Japan
- Research Institute for Cell Design Medical Science, Yamaguchi University, Yamaguchi, Japan
| | - Shunsuke Goto
- Department of Urology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Keishi Adachi
- Department of Immunology, Yamaguchi University Graduate School of Medicine, Yamaguchi, Japan
| | - Hidenori Takahashi
- Department of Gastroenterological, Breast and Endocrine Surgery, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi, 755-8505, Japan
| | - Koji Tamada
- Research Institute for Cell Design Medical Science, Yamaguchi University, Yamaguchi, Japan
- Department of Immunology, Yamaguchi University Graduate School of Medicine, Yamaguchi, Japan
| | - Hiroaki Nagano
- Department of Gastroenterological, Breast and Endocrine Surgery, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi, 755-8505, Japan.
- Research Institute for Cell Design Medical Science, Yamaguchi University, Yamaguchi, Japan.
| |
Collapse
|
10
|
Jaiswal A, Negi M, Choi EH, Kaushik NK, Kaushik N. Upstream-binding protein-1 promotes breast tumorigenesis by inducing NRG2-mediated metastasis, plasticity, and macrophage polarization. Int J Biol Macromol 2025; 307:141915. [PMID: 40064277 DOI: 10.1016/j.ijbiomac.2025.141915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 03/07/2025] [Accepted: 03/07/2025] [Indexed: 03/18/2025]
Abstract
Upstream binding protein 1 (UBP1) is a transcription factor (TF) of the CP2/grainyhead family involved in various biological processes, including cancer cell proliferation, differentiation, and embryonic development. While other mammalian grainyhead-like TFs have been linked to different cancers, including breast cancer (BC), the role of UBP1 in BC remains unexplored. In this study, we provide a preliminary investigation into the novel functions of UBP1 in BC. Using online database screening, we first demonstrated that elevated UBP1 levels in breast carcinoma are associated with poor prognosis and adverse clinical outcomes. We further showed that UBP1 promotes epithelial-mesenchymal transition (EMT) and stemness in BC cells while regulating key signaling pathways, including the PI3K-Akt. Additionally, UBP1 modulates tumor metastasis by influencing tumor-associated macrophage (TAM) polarization, promoting an immunosuppressive macrophage phenotype, and driving tumor progression. Our findings highlight UBP1's pivotal role in BC progression through multiple mechanisms, including EMT induction, stemness maintenance, and macrophage polarization via activation of the NRG2/Akt axis. Moreover, higher UBP1 expression correlates with lower overall and recurrence-free survival, underscoring its potential as a prognostic marker and therapeutic target for aggressive BC.
Collapse
Affiliation(s)
- Apurva Jaiswal
- Plasma Bioscience Research Center/Department of Electrical and Biological Physics, Kwangwoon University, Seoul 01897, Republic of Korea
| | - Manorma Negi
- Plasma Bioscience Research Center/Department of Electrical and Biological Physics, Kwangwoon University, Seoul 01897, Republic of Korea
| | - Eun Ha Choi
- Plasma Bioscience Research Center/Department of Electrical and Biological Physics, Kwangwoon University, Seoul 01897, Republic of Korea.
| | - Nagendra Kumar Kaushik
- Plasma Bioscience Research Center/Department of Electrical and Biological Physics, Kwangwoon University, Seoul 01897, Republic of Korea.
| | - Neha Kaushik
- Department of Biotechnology, College of Engineering, The University of Suwon, Hwaseong 18323, Republic of Korea.
| |
Collapse
|
11
|
Dong X, Wang X, Zheng X, Jiang H, Liu L, Ma N, Wang S. Targeted nanoparticle delivery system for tumor-associated macrophage reprogramming to enhance TNBC therapy. Cell Biol Toxicol 2025; 41:58. [PMID: 40056273 DOI: 10.1007/s10565-025-10001-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Accepted: 02/12/2025] [Indexed: 03/10/2025]
Abstract
Triple-negative breast cancer (TNBC) poses as a daunting and intricate manifestation of breast cancer, highlighted by few treatment options and a poor outlook. The crucial element in fostering tumor growth and immune resistance is the polarization of tumor-associated macrophages (TAMs) into the M2 state within the tumor microenvironment (TME). To address this, we developed M2 targeting peptide-chitosan-curcumin nanoparticles (M2pep-Cs-Cur NPs), a targeted delivery system utilizing chitosan (Cs) as a carrier, curcumin (Cur) as a therapeutic agent, and targeting peptides for specificity. These NPs effectively inhibited TNBC cell proliferation (~ 70%) and invasion (~ 70%), while increasing the responsiveness of tumors to anti-PD-L1 treatment (~ 50% survival enhancement) in vitro and in vivo. Bioinformatics analysis suggested that Cur modulates TAM polarization by influencing key genes such as COX-2, offering insights into its underlying mechanisms. This study highlights the potential of M2pep-Cs-Cur NPs to reverse M2 polarization in TAMs, providing a promising targeted therapeutic strategy to overcome immunotherapy resistance and improve TNBC outcomes.
Collapse
Affiliation(s)
- Xiaoshen Dong
- Department of Surgical Oncology, Breast Surgery, General Surgery, The First Hospital of China Medical University, 155 North Nanjing St, Shenyang, 110001, China
| | - Xiaoou Wang
- Department of Geriatric Cardiovascular, The First Hospital of China Medical University, 155 North Nanjing St, Shenyang, 110001, China
| | - Xinyu Zheng
- Department of Surgical Oncology, Breast Surgery, General Surgery, The First Hospital of China Medical University, 155 North Nanjing St, Shenyang, 110001, China
- Lab 1, Cancer Institute, the First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Haiyang Jiang
- Department of Surgical Oncology, Breast Surgery, General Surgery, The First Hospital of China Medical University, 155 North Nanjing St, Shenyang, 110001, China
| | - Lu Liu
- Department of Surgical Oncology, Breast Surgery, General Surgery, The First Hospital of China Medical University, 155 North Nanjing St, Shenyang, 110001, China
| | - Ningye Ma
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, 36 Sanhao Street, Heping District, Shenyang, Liaoning Province, China.
| | - Shuo Wang
- Department of Surgical Oncology, Breast Surgery, General Surgery, The First Hospital of China Medical University, 155 North Nanjing St, Shenyang, 110001, China.
| |
Collapse
|
12
|
Jiang T, Zhang J, Zhao S, Zhang M, Wei Y, Liu X, Zhang S, Fan W, Liu Y, Lv Y, Zhang G. MCT4: a key player influencing gastric cancer metastasis and participating in the regulation of the metastatic immune microenvironment. J Transl Med 2025; 23:276. [PMID: 40045374 PMCID: PMC11884109 DOI: 10.1186/s12967-025-06279-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 02/20/2025] [Indexed: 03/09/2025] Open
Abstract
BACKGROUND MCT4 is a lactate transporter associated with glycolysis, which has been found to be associated with various tumorigenesis and development processes. Gastric cancer is a malignant disease with high incidence and mortality. The role of MCT4 in the occurrence and development of gastric cancer has not been clarified. METHODS In this study, we comprehensively utilized single-cell sequencing and external transcriptome sequencing databases to deeply analyze the mechanism of the impact of MCT4 on gastric cancer and its microenvironment. We verified the function of MCT4 in gastric cancer through in vitro cell line experiments and in vivo experiments using gastric cancer liver metastasis and subcutaneous tumor models. Meanwhile, we collected tumor and normal tissue samples from clinical gastric cancer patients and employed immunohistochemistry and multiplex immunofluorescence techniques to detect the expression and localization of relevant indicators, thereby validating the results of computer simulation analysis and providing a basis for revealing the internal relationship between MCT4 and gastric cancer. RESULTS The expression of MCT4 is upregulated in gastric cancer patients, and the upregulation is more significant than that in patients with gastric cancer metastasis. MCT4 can mediate the proliferation and migration of gastric cancer cells in vitro. MCT4 can mediate the metastasis of gastric cancer cells in vivo. Multi-omics analysis showed that the expression of MCT4 was related to the composition of the immune microenvironment, and it could mediate the emergence of the inhibitory immune microenvironment. The results of immunofluorescence and immunohistochemistry proved the robustness of the multi-omics analysis. CONCLUSION Our study found that MCT4 plays an important role in the occurrence and development of gastric cancer, which may mediate the occurrence of gastric cancer metastasis and shape the immunosuppressive tumor microenvironment.
Collapse
Affiliation(s)
- Tao Jiang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
- Key Laboratory of Blood-Stasis-Toxin Syndrome of Zhejiang Province, Hangzhou, China
| | - Jingcheng Zhang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
- Key Laboratory of Blood-Stasis-Toxin Syndrome of Zhejiang Province, Hangzhou, China
| | - Sicheng Zhao
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
- Key Laboratory of Blood-Stasis-Toxin Syndrome of Zhejiang Province, Hangzhou, China
| | - Mingsi Zhang
- School of Sport, Loughborough University, Loughborough, LE, UK
| | - Yunhai Wei
- Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, No.1558, Sanhuan North Road, Wuxing District, Huzhou, 313000, Zhejiang Province, China
| | - Xiaojuan Liu
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
- Key Laboratory of Blood-Stasis-Toxin Syndrome of Zhejiang Province, Hangzhou, China
| | - Shuo Zhang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
- Key Laboratory of Blood-Stasis-Toxin Syndrome of Zhejiang Province, Hangzhou, China
| | - Wei Fan
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
- Key Laboratory of Blood-Stasis-Toxin Syndrome of Zhejiang Province, Hangzhou, China
| | - Yueying Liu
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
- Key Laboratory of Blood-Stasis-Toxin Syndrome of Zhejiang Province, Hangzhou, China
| | - Yuanlin Lv
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China.
| | - Guangji Zhang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China.
- Key Laboratory of Blood-Stasis-Toxin Syndrome of Zhejiang Province, Hangzhou, China.
| |
Collapse
|
13
|
Li J, Yu S, Rao M, Cheng B. Tumor-derived extracellular vesicles: key drivers of immunomodulation in breast cancer. Front Immunol 2025; 16:1548535. [PMID: 40103824 PMCID: PMC11914124 DOI: 10.3389/fimmu.2025.1548535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Accepted: 02/20/2025] [Indexed: 03/20/2025] Open
Abstract
Breast cancer (BC) remains a significant global health challenge characterized by its heterogeneity and treatment complexities. Extracellular vesicles (EVs) are small membranous particles released by cells, facilitating intercellular communication by transporting bioactive molecules such as proteins, lipids, and nucleic acids. Tumor-derived EVs have emerged as pivotal regulators in the tumor microenvironment (TME) and drivers of BC progression. These EVs carry diverse cargoes of bioactive molecules, influencing critical processes such as immune modulation, angiogenesis, and metastasis. By altering the behaviors of immune cells including macrophages, dendritic cells, and T cells, tumor-derived EVs contribute to immune evasion and tumor growth. Furthermore, Tumor-derived EVs play a role in mediating drug resistance, impacting the effectiveness of therapeutic interventions. Understanding the multifaceted roles of BC tumor-derived EVs is essential for the development of innovative therapeutic strategies. Targeting pathways mediated by EVs holds promise for enhancing the efficacy of cancer treatments and improving patient outcomes. This comprehensive review provides insights into the intricate interactions of tumor-derived EVs in immune modulation and BC progression, highlighting potential therapeutic targets and avenues for novel cancer therapies.
Collapse
Affiliation(s)
- Jieming Li
- Traditional Chinese Medicine (Zhong Jing) School, Henan University of Chinese Medicine, Zhengzhou, China
- Department of Polysaccharides and Drugs, Henan Key Laboratory of Chinese Medicine, Zhengzhou, China
| | - Shuo Yu
- Department of Biliary-Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Min Rao
- Nursing Department, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bomin Cheng
- Chinese Medicine Health Management Center, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, China
| |
Collapse
|
14
|
Zhang X, Cong L, Yu R, Yu Q, Hou X, Zhou Y. MicroRNA‑96 promotes the proliferation and migration of breast cancer cells by inhibiting Smad7 expression. Oncol Lett 2025; 29:151. [PMID: 39898288 PMCID: PMC11783994 DOI: 10.3892/ol.2025.14897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 10/26/2023] [Indexed: 02/04/2025] Open
Abstract
The present study aimed to investigate the effects of microRNA (miR)-96 on the proliferation and migration of breast cancer cells, and indicated that miR-96 may have a promoting role in breast cancer by inhibiting Smad7. Reverse transcription-quantitative (RT-q)PCR was used to detect the expression levels of miR-96 and Smad7 in breast cancer tissues and adjacent tissues. Western blotting and immunohistochemistry were conducted to determine the expression levels of SMAD7 in breast cancer and adjacent tissues. A dual luciferase assay was performed to verify the targeted binding between miR-96 and Smad7. Furthermore, the different expression patterns of miR-96 and Smad7 were compared in various breast cancer cell lines using RT-qPCR and western blotting. Among these cell lines, MDA-MB-231, which exhibited the highest expression of miR-96, was chosen for subsequent functional verification. The expression levels of miR-96 were significantly higher in breast cancer tissues compared with those in adjacent tissues. By contrast, the expression levels of Smad7 were significantly lower in breast cancer tissues compared with those in adjacent tissues. The dual luciferase assay revealed a targeted binding effect between miR-96 and Smad7. Notably, transfection with miR-96-5p mimics and short hairpin RNA-Smad7 markedly promoted the proliferation, adhesion, invasion and migration of breast cancer cells. Conversely, transfection with a miR-96-5p inhibitor and Smad7 overexpression plasmid exhibited the opposite trend. In conclusion, the expression levels of miR-96 were significantly elevated in breast cancer tissues compared with those in adjacent tissues. Overexpression of miR-96 was shown to promote the migration of breast cancer cells by downregulating the expression of Smad7. These findings indicated that miR-96 may serve as a prognostic marker for breast cancer.
Collapse
Affiliation(s)
- Xiumei Zhang
- Department of Pathology, The People's Hospital of Xinghua City, Xinghua, Jiangsu 225700, P.R. China
| | - Lin Cong
- Department of Pathology, The People's Hospital of Xinghua City, Xinghua, Jiangsu 225700, P.R. China
| | - Rong Yu
- Department of Gastrointestinal Surgery, The People's Hospital of Xinghua City, Xinghua, Jiangsu 225700, P.R. China
| | - Qianwen Yu
- Department of Pathology, The People's Hospital of Xinghua City, Xinghua, Jiangsu 225700, P.R. China
| | - Xian Hou
- Department of Radiology, The People's Hospital of Xinghua City, Xinghua, Jiangsu 225700, P.R. China
| | - Yonghua Zhou
- Department of Breast Surgery, The People's Hospital of Xinghua City, Xinghua, Jiangsu 225700, P.R. China
| |
Collapse
|
15
|
Miura R, Ono A, Nakahara H, Shirane Y, Yamaoka K, Fujii Y, Uchikawa S, Fujino H, Murakami E, Kawaoka T, Miki D, Tsuge M, Kishi T, Ohishi W, Sakamoto N, Arihiro K, Hayes CN, Oka S. Serum IL-6 concentration is a useful biomarker to predict the efficacy of atezolizumab plus bevacizumab in patients with hepatocellular carcinoma. J Gastroenterol 2025; 60:328-339. [PMID: 39652104 PMCID: PMC11880141 DOI: 10.1007/s00535-024-02185-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 11/18/2024] [Indexed: 03/05/2025]
Abstract
BACKGROUND This study aims to identify biomarkers for treatment response of atezolizumab plus bevacizumab (Atezo+Bev) in patients with hepatocellular carcinoma (HCC). METHODS 96 patients who received Atezo+Bev or lenvatinib as a first-line systemic therapy were enrolled as the training group after propensity score matching (PSM), and 42 patients treated with Atezo+Bev were enrolled as the validation group. 17 serum cytokines were measured by Luminex multiplex assay at the start of treatment. For further assessment of the association between cytokine levels and the tumor microenvironment (TME), immunohistochemistry (IHC) was performed on pre-treatment liver biopsy specimens. RESULTS In the derivation set, multivariate analysis identified elevated IL-6 as an independent risk factor in the Atezo+Bev group (HR 5.80: p<0.01), but not in the lenvatinib group; in a subset analysis of patients with low IL-6, PFS was longer in the Atezo+Bev training group than in the lenvatinib group (p = 0.02). A validation study also showed a significantly longer prognosis in the low IL-6 group for both PFS (p = 0.0001) and OS (p = 0.03). Serum IL-6 had a positive correlation with tumor IL-6 expression (ρ = 0.56, p < 0.0001) and an inverse correlation with the CD8/CD163-positive cell count ratio (ρ = -0.4, p < 0.01). CONCLUSION Serum IL-6 levels are thought to be involved in the suppression of tumor immunity and are useful in predicting the therapeutic effect of Atezo+Bev treatment.
Collapse
Affiliation(s)
- Ryoichi Miura
- Department of Gastroenterology, Graduate School of Biomedical and Health Sciences, Hiroshima University Hospital, Hiroshima, 734-8551, Japan
| | - Atsushi Ono
- Department of Gastroenterology, Graduate School of Biomedical and Health Sciences, Hiroshima University Hospital, Hiroshima, 734-8551, Japan.
| | - Hikaru Nakahara
- Department of Gastroenterology, Graduate School of Biomedical and Health Sciences, Hiroshima University Hospital, Hiroshima, 734-8551, Japan
| | - Yuki Shirane
- Department of Gastroenterology, Graduate School of Biomedical and Health Sciences, Hiroshima University Hospital, Hiroshima, 734-8551, Japan
| | - Kenji Yamaoka
- Department of Gastroenterology, Graduate School of Biomedical and Health Sciences, Hiroshima University Hospital, Hiroshima, 734-8551, Japan
| | - Yasutoshi Fujii
- Department of Clinical Oncology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Shinsuke Uchikawa
- Department of Gastroenterology, Graduate School of Biomedical and Health Sciences, Hiroshima University Hospital, Hiroshima, 734-8551, Japan
| | - Hatsue Fujino
- Department of Gastroenterology, Graduate School of Biomedical and Health Sciences, Hiroshima University Hospital, Hiroshima, 734-8551, Japan
| | - Eisuke Murakami
- Department of Gastroenterology, Graduate School of Biomedical and Health Sciences, Hiroshima University Hospital, Hiroshima, 734-8551, Japan
| | - Tomokazu Kawaoka
- Department of Gastroenterology, Graduate School of Biomedical and Health Sciences, Hiroshima University Hospital, Hiroshima, 734-8551, Japan
| | - Daiki Miki
- Department of Gastroenterology, Graduate School of Biomedical and Health Sciences, Hiroshima University Hospital, Hiroshima, 734-8551, Japan
| | - Masataka Tsuge
- Department of Gastroenterology, Graduate School of Biomedical and Health Sciences, Hiroshima University Hospital, Hiroshima, 734-8551, Japan
- Liver Center, Hiroshima University, Hiroshima, 734-8551, Japan
| | - Takeshi Kishi
- Biosample Research Center, Radiation Effects Research Foundation, Hiroshima, Japan
| | - Waka Ohishi
- Department of Clinical Studies, Radiation Effects Research Foundation, Hiroshima, Japan
| | - Naoya Sakamoto
- Department of Pathology and Clinical Laboratories, National Cancer Center-Hospital East, Kashiwa, Chiba, Japan
| | - Koji Arihiro
- Department of Anatomical Pathology, Hiroshima University Hospital, Hiroshima, Japan
| | - Clair Nelson Hayes
- Department of Gastroenterology, Graduate School of Biomedical and Health Sciences, Hiroshima University Hospital, Hiroshima, 734-8551, Japan
| | - Shiro Oka
- Department of Gastroenterology, Graduate School of Biomedical and Health Sciences, Hiroshima University Hospital, Hiroshima, 734-8551, Japan
| |
Collapse
|
16
|
Zheng B, Wang Y, Zhou B, Qian F, Liu D, Ye D, Zhou X, Fang L. Urolithin A inhibits breast cancer progression via activating TFEB-mediated mitophagy in tumor macrophages. J Adv Res 2025; 69:125-138. [PMID: 38615740 PMCID: PMC11954813 DOI: 10.1016/j.jare.2024.04.010] [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: 01/11/2024] [Revised: 03/01/2024] [Accepted: 04/10/2024] [Indexed: 04/16/2024] Open
Abstract
INTRODUCTION Urolithin A (UA) is a naturally occurring compound that is converted from ellagitannin-like precursors in pomegranates and nuts by intestinal flora. Previous studies have found that UA exerts tumor-suppressive effects through antitumor cell proliferation and promotion of memory T-cell expansion, but its role in tumor-associated macrophages remains unknown. OBJECTIVES Our study aims to reveal how UA affects tumor macrophages and tumor cells to inhibit breast cancer progression. METHODS Observe the effect of UA treatment on breast cancer progression though in vivo and in vitro experiments. Western blot and PCR assays were performed to discover that UA affects tumor macrophage autophagy and inflammation. Co-ip and Molecular docking were used to explore specific molecular mechanisms. RESULTS We observed that UA treatment could simultaneously inhibit harmful inflammatory factors, especially for InterleuKin-6 (IL-6) and tumor necrosis factor α (TNF-α), in both breast cancer cells and tumor-associated macrophages, thereby improving the tumor microenvironment and delaying tumor progression. Mechanistically, UA induced the key regulator of autophagy, transcription factor EB (TFEB), into the nucleus in a partially mTOR-dependent manner and inhibited the ubiquitination degradation of TFEB, which facilitated the clearance of damaged mitochondria via the mitophagy-lysosomal pathway in macrophages under tumor supernatant stress, and reduced the deleterious inflammatory factors induced by the release of nucleic acid from damaged mitochondria. Molecular docking and experimental studies suggest that UA block the recognition of TFEB by 1433 and induce TFEB nuclear localization. Notably, UA treatment demonstrated inhibitory effects on tumor progression in multiple breast cancer models. CONCLUSION Our study elucidated the anti-breast cancer effect of UA from the perspective of tumor-associated macrophages. Specifically, TFEB is a crucial downstream target in macrophages.
Collapse
Affiliation(s)
- Bowen Zheng
- Department of Breast and Thyroid Surgery, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072, China
| | - Yuying Wang
- Department of Breast and Thyroid Surgery, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072, China
| | - Baian Zhou
- Department of Breast and Thyroid Surgery, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072, China
| | - Fengyuan Qian
- Department of Breast and Thyroid Surgery, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072, China
| | - Diya Liu
- Department of Breast and Thyroid Surgery, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072, China
| | - Danrong Ye
- Department of Breast and Thyroid Surgery, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072, China; Department of Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325015, China
| | - Xiqian Zhou
- Department of Breast and Thyroid Surgery, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072, China
| | - Lin Fang
- Department of Breast and Thyroid Surgery, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072, China.
| |
Collapse
|
17
|
Jin H, Meng X, Feng J. Mechanisms of tumor-associated macrophages in breast cancer and treatment strategy. Front Immunol 2025; 16:1560393. [PMID: 40092996 PMCID: PMC11906463 DOI: 10.3389/fimmu.2025.1560393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Accepted: 02/13/2025] [Indexed: 03/19/2025] Open
Abstract
Breast cancer (BC) is the most common cancer in women and a leading cause of cancer-related mortality. Despite advances in screening and treatment, outcomes for advanced or recurrent BC remain poor, highlighting the need for new strategies. Recent research emphasizes the tumor microenvironment (TME), particularly tumor-associated macrophages (TAMs), as key drivers of tumor growth, metastasis, and resistance to therapy. The presence of M2-like TAMs in the TME promotes immune evasion and tumor progression across BC subtypes. This review summarizes TAMs classification, their role in BC, and emerging therapies targeting TAMs, including depletion, inhibition of recruitment, and reprogramming from pro-tumoral M2 to anti-tumoral M1 phenotypes. Targeting TAMs offers a promising strategy to improve BC treatment outcomes.
Collapse
Affiliation(s)
- Hong Jin
- Department of Ultrasound, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Xinyue Meng
- Department of Ultrasound, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Jianwei Feng
- Department of Ultrasound, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
18
|
Berkley K, Zalejski J, Sharma A. Targeting STAT3 for Cancer Therapy: Focusing on Y705, S727, or Dual Inhibition? Cancers (Basel) 2025; 17:755. [PMID: 40075607 PMCID: PMC11898704 DOI: 10.3390/cancers17050755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Revised: 02/14/2025] [Accepted: 02/20/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND/OBJECTIVES Signal Transducer and Activator of Transcription 3 (STAT3) is a transcription factor that is strongly implicated in various cancers. In its canonical signaling pathway, Janus kinases (JAKs) phosphorylate STAT3 at the Y705 residue in response to cytokines or growth factors, with pY705 serving as a key marker of STAT3 oncogenic activity. Elevated pY705 levels correlate with poor prognosis, and numerous small-molecule inhibitors have been developed to block this phosphorylation site. More recently, phosphorylation at the S727 residue (pS727) has emerged as a critical contributor to STAT3-mediated oncogenesis, particularly due to its role in mitochondrial translocation. Evidence suggests that pS727 may even surpass pY705 in driving oncogenic activity. These findings prompt an important question: Which residue should be prioritized for effective STAT3 inhibition in cancer therapy? METHODS This review compiles and critically analyzes the current literature on STAT3 inhibitors targeting pY705 and/or pS727, evaluating their therapeutic efficacy in vitro, in vivo, and in clinical trials. We assess the unique effects of targeting each residue on downstream signaling, toxicity, and clinical outcomes. RESULTS Our analysis indicates that inhibitors targeting both pY705 and pS727 achieve the greatest therapeutic effectiveness. However, pS727 targeting is associated with higher toxicity risks. CONCLUSIONS Comprehensive evaluation of STAT3 inhibitors underscores the importance of targeting pY705 for maximum therapeutic benefit. The analysis also shows that co-targeting pS727 may increase overall efficacy. However, pS727 inhibition should be approached with lower affinity to minimize toxicity and enhance the clinical feasibility of dual-targeting strategies.
Collapse
Affiliation(s)
| | | | - Ashutosh Sharma
- Department of Chemistry, University of Illinois Chicago, Chicago, IL 60607, USA; (K.B.); (J.Z.)
| |
Collapse
|
19
|
Sun M, Wang K, Lu F, Yu D, Liu S. Regulatory role and therapeutic prospect of lactate modification in cancer. Front Pharmacol 2025; 16:1508552. [PMID: 40034817 PMCID: PMC11872897 DOI: 10.3389/fphar.2025.1508552] [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: 10/09/2024] [Accepted: 01/20/2025] [Indexed: 03/05/2025] Open
Abstract
Post-translational modifications (PTMs) of proteins refer to the process of adding chemical groups, sugars, or other molecules to specific residues of target proteins following their biosynthesis by ribosomes. PTMs play a crucial role in processes such as signal transduction, epigenetics, and disease development. Lactylation is a newly discovered PTM that, due to its close association with lactate-the end product of glycolytic metabolism-provides a new perspective on the connection between cellular metabolic reprogramming and epigenetic regulation. Studies have demonstrated that lactylation plays a significant role in tumor progression and is associated with poor clinical prognosis. Abnormal histone lactylation can influence gene expression in both tumor cells and immune cells, thereby regulating tumor progression and immunosuppression. Lactylation of non-histone proteins can also modulate processes such as tumor proliferation and drug resistance. This review summarizes the latest research progress in the field of lactylation, highlighting its roles and mechanisms in tumorigenesis, tumor development, the tumor microenvironment, and immunosuppression. It also explores the potential application value of lactylation in tumor-targeted therapy and combined immunotherapy.
Collapse
Affiliation(s)
- Mengdi Sun
- Graduate School, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Kejing Wang
- Graduate School, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Fang Lu
- Institute of Traditional Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Donghua Yu
- Institute of Traditional Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Shumin Liu
- Institute of Traditional Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin, China
| |
Collapse
|
20
|
Abudourousuli A, Aierken Z, Mamuti H, Yimamu T, Da C. Apoptosis antagonizing transcription factor expression and its validation as a potential diagnostic and prognostic biomarker in oral squamous cell carcinoma. Front Oncol 2025; 15:1542730. [PMID: 39911629 PMCID: PMC11794051 DOI: 10.3389/fonc.2025.1542730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Accepted: 01/02/2025] [Indexed: 02/07/2025] Open
Abstract
Background Oral squamous cell carcinoma (OSCC) is characterized by a high degree of malignancy and poor prognosis. This study aimed to investigate the expression of apoptosis antagonizing transcription factor (AATF) in OSCC, examine its correlation with clinicopathological features, assess its prognostic implications, and explore its potential role in OSCC progression. Methods Expression profiles and clinical data of OSCC patients were obtained from The Cancer Genome Atlas (TCGA). Immunohistochemical analysis on tissue microarrays was performed to assess AATF expression in OSCC. Functional enrichment analyses were conducted to identify potential signaling pathways and biological functions associated with AATF. Logistic regression analyses were employed to evaluate the relationship between AATF expression and clinicopathological features. Immune cell infiltration was assessed using single-sample gene set enrichment analysis (ssGSEA). The prognostic value of AATF was determined using Kaplan-Meier and Cox regression analyses. A nomogram was developed to predict overall survival (OS) rates at one, three-, and five years post-cancer diagnosis. Validation of AATF expression was performed using quantitative real-time PCR (qRT-PCR). Results AATF was significantly overexpressed in OSCC, and high AATF expression correlated with adverse clinicopathological features, including histologic grade and lymph node metastasis. Functional enrichment analysis revealed several enriched pathways, including epidermis development, immunoglobulin complex, antigen binding and IL-17 signaling pathway. Notably, AATF overexpression was negatively correlated with the infiltration levels of mast cells, interdigitating dendritic cells and Th 17 cells. High AATF expression significantly predicted poorer overall survival (OS) and disease-specific survival (DSS). Multivariate Cox analysis confirmed AATF as an independent negative prognostic marker of OS. Validation via qRT-PCR confirmed the overexpression of AATF in OSCC tissues. Conclusion Elevated expression of AATF in OSCC correlates with adverse clinicopathological features and negatively impacts immune cell infiltration. High AATF levels serve as an independent marker of poor OS and DSS. These findings support AATF as a valuable prognostic biomarker and a potential therapeutic target in OSCC, warranting further investigation.
Collapse
Affiliation(s)
| | - Zumulaiti Aierken
- Department of Stomatology, The First People`s Hospital of Kashi Prefecture, Kashi, China
| | - Hasiyati Mamuti
- Department of Pathology, The First People`s Hospital of Kashi Prefecture, Kashi, China
| | - Tuxunayi Yimamu
- Department of Pathology, The First People`s Hospital of Kashi Prefecture, Kashi, China
| | - Chengli Da
- Department of Oral and Maxillofacial Surgery, The First People`s Hospital of Kashi Prefecture, Kashi, China
| |
Collapse
|
21
|
Chen Z, Zhao Y. The mechanism underlying metastasis in triple-negative breast cancer: focusing on the interplay between ferroptosis, epithelial-mesenchymal transition, and non-coding RNAs. Front Pharmacol 2025; 15:1437022. [PMID: 39881868 PMCID: PMC11774878 DOI: 10.3389/fphar.2024.1437022] [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: 05/23/2024] [Accepted: 12/27/2024] [Indexed: 01/31/2025] Open
Abstract
Triple-negative breast cancer (TNBC) is a type of breast cancer with lack the expression of estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2). It is the most aggressive breast cancer and the most difficult to treat due to its poor response to treatments and extremely invasive characteristics. The typical treatment for TNBC frequently results in relapse because of the lack of particular treatment choices. It is urgent to focus on identifying a workable and effective target for the treatment of TNBC. Cancer metastasis is significantly influenced by epithelial-mesenchymal transition (EMT). Ferroptosis is an iron-dependent cell death form, and changes its key factor to affect the proliferation and metastasis of TNBC. Several reports have established associations between EMT and ferroptosis in TNBC metastasis. Furthermore, non-coding RNA (ncRNA), which has been previously described, can also control cancer cell death and metastasis. Thus, in this review, we summarize the correlation and pathways among the ferroptosis, EMT, and ncRNAs in TNBC metastasis. Also, aim to find out a novel strategy for TNBC treatment through the ncRNA-ferroptosis-EMT axis.
Collapse
Affiliation(s)
- Ziyi Chen
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
| | - Yi Zhao
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
- Key Laboratory of Birth Regulation and Control Technology of National Health Commission of China, Maternal and Child Healthcare Hospital of Shandong Province Affiliated to Qingdao University, Jinan, Shandong, China
| |
Collapse
|
22
|
Panda VK, Mishra B, Mahapatra S, Swain B, Malhotra D, Saha S, Khanra S, Mishra P, Majhi S, Kumari K, Nath AN, Saha S, Jena S, Kundu GC. Molecular Insights on Signaling Cascades in Breast Cancer: A Comprehensive Review. Cancers (Basel) 2025; 17:234. [PMID: 39858015 PMCID: PMC11763662 DOI: 10.3390/cancers17020234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 12/27/2024] [Accepted: 01/01/2025] [Indexed: 01/27/2025] Open
Abstract
The complex signaling network within the breast tumor microenvironment is crucial for its growth, metastasis, angiogenesis, therapy escape, stem cell maintenance, and immunomodulation. An array of secretory factors and their receptors activate downstream signaling cascades regulating breast cancer progression and metastasis. Among various signaling pathways, the EGFR, ER, Notch, and Hedgehog signaling pathways have recently been identified as crucial in terms of breast cancer proliferation, survival, differentiation, maintenance of CSCs, and therapy failure. These receptors mediate various downstream signaling pathways such as MAPK, including MEK/ERK signaling pathways that promote common pro-oncogenic signaling, whereas dysregulation of PI3K/Akt, Wnt/β-catenin, and JAK/STAT activates key oncogenic events such as drug resistance, CSC enrichment, and metabolic reprogramming. Additionally, these cascades orchestrate an intricate interplay between stromal cells, immune cells, and tumor cells. Metabolic reprogramming and adaptations contribute to aggressive breast cancer and are unresponsive to therapy. Herein, recent insights into the novel signaling pathways operating within the breast TME that aid in their advancement are emphasized and current developments in practices targeting the breast TME to enhance treatment efficacy are reviewed.
Collapse
Affiliation(s)
- Venketesh K. Panda
- School of Biotechnology, KIIT Deemed to Be University, Bhubaneswar 751024, India; (V.K.P.); (B.M.); (S.M.); (B.S.); (D.M.); (S.S.); (S.K.); (P.M.); (S.M.); (K.K.); (A.N.N.); (S.S.); (S.J.)
- School of Applied Sciences, KIIT Deemed to Be University, Bhubaneswar 751024, India
| | - Barnalee Mishra
- School of Biotechnology, KIIT Deemed to Be University, Bhubaneswar 751024, India; (V.K.P.); (B.M.); (S.M.); (B.S.); (D.M.); (S.S.); (S.K.); (P.M.); (S.M.); (K.K.); (A.N.N.); (S.S.); (S.J.)
| | - Samikshya Mahapatra
- School of Biotechnology, KIIT Deemed to Be University, Bhubaneswar 751024, India; (V.K.P.); (B.M.); (S.M.); (B.S.); (D.M.); (S.S.); (S.K.); (P.M.); (S.M.); (K.K.); (A.N.N.); (S.S.); (S.J.)
| | - Biswajit Swain
- School of Biotechnology, KIIT Deemed to Be University, Bhubaneswar 751024, India; (V.K.P.); (B.M.); (S.M.); (B.S.); (D.M.); (S.S.); (S.K.); (P.M.); (S.M.); (K.K.); (A.N.N.); (S.S.); (S.J.)
| | - Diksha Malhotra
- School of Biotechnology, KIIT Deemed to Be University, Bhubaneswar 751024, India; (V.K.P.); (B.M.); (S.M.); (B.S.); (D.M.); (S.S.); (S.K.); (P.M.); (S.M.); (K.K.); (A.N.N.); (S.S.); (S.J.)
| | - Suryendu Saha
- School of Biotechnology, KIIT Deemed to Be University, Bhubaneswar 751024, India; (V.K.P.); (B.M.); (S.M.); (B.S.); (D.M.); (S.S.); (S.K.); (P.M.); (S.M.); (K.K.); (A.N.N.); (S.S.); (S.J.)
| | - Sinjan Khanra
- School of Biotechnology, KIIT Deemed to Be University, Bhubaneswar 751024, India; (V.K.P.); (B.M.); (S.M.); (B.S.); (D.M.); (S.S.); (S.K.); (P.M.); (S.M.); (K.K.); (A.N.N.); (S.S.); (S.J.)
| | - Priyanka Mishra
- School of Biotechnology, KIIT Deemed to Be University, Bhubaneswar 751024, India; (V.K.P.); (B.M.); (S.M.); (B.S.); (D.M.); (S.S.); (S.K.); (P.M.); (S.M.); (K.K.); (A.N.N.); (S.S.); (S.J.)
| | - Sambhunath Majhi
- School of Biotechnology, KIIT Deemed to Be University, Bhubaneswar 751024, India; (V.K.P.); (B.M.); (S.M.); (B.S.); (D.M.); (S.S.); (S.K.); (P.M.); (S.M.); (K.K.); (A.N.N.); (S.S.); (S.J.)
| | - Kavita Kumari
- School of Biotechnology, KIIT Deemed to Be University, Bhubaneswar 751024, India; (V.K.P.); (B.M.); (S.M.); (B.S.); (D.M.); (S.S.); (S.K.); (P.M.); (S.M.); (K.K.); (A.N.N.); (S.S.); (S.J.)
| | - Angitha N. Nath
- School of Biotechnology, KIIT Deemed to Be University, Bhubaneswar 751024, India; (V.K.P.); (B.M.); (S.M.); (B.S.); (D.M.); (S.S.); (S.K.); (P.M.); (S.M.); (K.K.); (A.N.N.); (S.S.); (S.J.)
| | - Swarnali Saha
- School of Biotechnology, KIIT Deemed to Be University, Bhubaneswar 751024, India; (V.K.P.); (B.M.); (S.M.); (B.S.); (D.M.); (S.S.); (S.K.); (P.M.); (S.M.); (K.K.); (A.N.N.); (S.S.); (S.J.)
| | - Sarmistha Jena
- School of Biotechnology, KIIT Deemed to Be University, Bhubaneswar 751024, India; (V.K.P.); (B.M.); (S.M.); (B.S.); (D.M.); (S.S.); (S.K.); (P.M.); (S.M.); (K.K.); (A.N.N.); (S.S.); (S.J.)
| | - Gopal C. Kundu
- School of Biotechnology, KIIT Deemed to Be University, Bhubaneswar 751024, India; (V.K.P.); (B.M.); (S.M.); (B.S.); (D.M.); (S.S.); (S.K.); (P.M.); (S.M.); (K.K.); (A.N.N.); (S.S.); (S.J.)
- School of Applied Sciences, KIIT Deemed to Be University, Bhubaneswar 751024, India
- Kalinga Institute of Medical Sciences (KIMS), KIIT Deemed to Be University, Bhubaneswar 751024, India
| |
Collapse
|
23
|
Du J, Shen M, Chen J, Yan H, Xu Z, Yang X, Yang B, Luo P, Ding K, Hu Y, He Q. The impact of solute carrier proteins on disrupting substance regulation in metabolic disorders: insights and clinical applications. Front Pharmacol 2025; 15:1510080. [PMID: 39850557 PMCID: PMC11754210 DOI: 10.3389/fphar.2024.1510080] [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: 10/12/2024] [Accepted: 12/20/2024] [Indexed: 01/25/2025] Open
Abstract
Carbohydrates, lipids, bile acids, various inorganic salt ions and organic acids are the main nutrients or indispensable components of the human body. Dysregulation in the processes of absorption, transport, metabolism, and excretion of these metabolites can lead to the onset of severe metabolic disorders, such as type 2 diabetes, non-alcoholic fatty liver disease, gout and hyperbilirubinemia. As the second largest membrane receptor supergroup, several major families in the solute carrier (SLC) supergroup have been found to play key roles in the transport of substances such as carbohydrates, lipids, urate, bile acids, monocarboxylates and zinc ions. Based on common metabolic dysregulation and related metabolic substances, we explored the relationship between several major families of SLC supergroup and metabolic diseases, providing examples of drugs targeting SLC proteins that have been approved or are currently in clinical/preclinical research as well as SLC-related diagnostic techniques that are in clinical use or under investigation. By highlighting these connections, we aim to provide insights that may contribute to the development of improved treatment strategies and targeted therapies for metabolic disorders.
Collapse
Affiliation(s)
- Jiangxia Du
- Center for Medical Research and Innovation in Digestive System Tumors, Ministry of Education, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Minhui Shen
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jiajia Chen
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Hao Yan
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Zhifei Xu
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xiaochun Yang
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Bo Yang
- Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
- School of Medicine, Hangzhou City University, Hangzhou, Zhejiang, China
| | - Peihua Luo
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
- Department of Pharmaceutical and Translational Toxicology, Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Hangzhou, Zhejiang, China
| | - Kefeng Ding
- Center for Medical Research and Innovation in Digestive System Tumors, Ministry of Education, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yuhuai Hu
- Yuhong Pharmaceutical Technology Co., Ltd., Hangzhou, Zhejiang, China
| | - Qiaojun He
- Center for Medical Research and Innovation in Digestive System Tumors, Ministry of Education, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
- School of Medicine, Hangzhou City University, Hangzhou, Zhejiang, China
- Department of Pharmaceutical and Translational Toxicology, Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|
24
|
Chen Y, Luo Y, Liu Y, Luo D, Liu A. Dual efficacy of tocilizumab in managing PD-1 inhibitors-induced myocardial inflammatory injury and suppressing tumor growth with PD-1 inhibitors: a preclinical study. Cancer Immunol Immunother 2025; 74:52. [PMID: 39752010 PMCID: PMC11699076 DOI: 10.1007/s00262-024-03899-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 11/16/2024] [Indexed: 01/04/2025]
Abstract
The combined use of tocilizumab (TCZ) and immune checkpoint inhibitors (ICIs) in cancer treatment is gaining attention, but preclinical studies are lacking. Our study aims to investigate the synergistic anti-tumor effect of TCZ combined with ICIs and its role in treating immune-related adverse events (irAEs). The clinical significance of high interleukin-6 (IL-6) expression in tumor patients was analyzed from the Cancer Genome Atlas (TCGA) database. The expression levels of IL-6 were compared before and during the onset of ICIs-associated myocarditis patients. ICIs-related myocardial inflammatory injury and therapeutic lung cancer models were constructed in C57BL/6 J mice using murine-derived programmed death-1 (PD-1) inhibitors alone or in combination with TCZ. Possible inflammatory mechanisms were proposed and validated. The anti-tumor effects and mechanisms of both drugs in combination were assessed. Patients with high IL-6 expression had a poor prognosis, and those with ICIs-associated myocarditis exhibited elevated IL-6 from baseline. In the PD-1 inhibitors-associated myocardial inflammatory injury mouse model, the levels of IL-6 in the blood and cardiac tissues were significantly elevated. TCZ ameliorated immune myocardial inflammatory injury by inhibiting the IL-6/janus kinase 2 (JAK2)/signal transducer and activator of the transcription 3 (STAT3) pathway. The group treated with PD-1 inhibitors combined with TCZ showed significantly slower tumor growth than that treated with PD-1 inhibitors alone. TCZ resisted tumor growth by inhibiting the IL-6-JAK2-STAT3 pathway. By targeting the IL-6-JAK2-STAT3 pathway, TCZ can alleviate PD-1 inhibitors-associated myocardial inflammatory injury mediated by M1-polarized macrophages and plays a synergistic anti-tumor role by inhibiting lung cancer cell proliferation.
Collapse
Affiliation(s)
- Yanxin Chen
- Department of Oncology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi Province, China
- Department of Radiotherapy, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410000, Hunan Province, China
- Jiangxi Key Laboratory of Clinical Translational Cancer Research, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi Province, China
- Radiation Induced Heart Damage Institute, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi Province, China
| | - Yuxi Luo
- Department of Oncology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi Province, China
- Jiangxi Key Laboratory of Clinical Translational Cancer Research, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi Province, China
- Radiation Induced Heart Damage Institute, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi Province, China
| | - Yunwei Liu
- Department of Oncology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi Province, China
- Jiangxi Key Laboratory of Clinical Translational Cancer Research, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi Province, China
- Radiation Induced Heart Damage Institute, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi Province, China
| | - Daya Luo
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Nanchang University, Nanchang, 330006, Jiangxi Province, China
| | - Anwen Liu
- Department of Oncology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi Province, China.
- Jiangxi Key Laboratory of Clinical Translational Cancer Research, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi Province, China.
- Radiation Induced Heart Damage Institute, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi Province, China.
| |
Collapse
|
25
|
Balkrishna A, Mittal R, Bishayee A, Kumar AP, Bishayee A. miRNA signatures affecting the survival outcome in distant metastasis of triple-negative breast cancer. Biochem Pharmacol 2025; 231:116683. [PMID: 39608504 DOI: 10.1016/j.bcp.2024.116683] [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: 08/03/2024] [Revised: 11/11/2024] [Accepted: 11/26/2024] [Indexed: 11/30/2024]
Abstract
Triple-negative breast cancer (TNBC) constitutes for 10-15% of all breast cancer cases. Tumor heterogeneity, high invasiveness, distant metastasis, lack of estrogen receptors, progesterone receptors, and human epidermal growth factor receptor 2 expression contribute to TNBC associated with poor overall survival outcomes amongst diseased individuals. The disparity in clinico-pathological and metastatic patterns to distant sites has substantially enhanced the incidences of tumor recurrence. Survival outcomes amongst metastatic TNBC patients are worse in comparison to non-metastatic TNBC counterparts. MicroRNAs (miRNAs) have emerged as significant drivers to function either as oncogene or tumor suppressors by exerting modulating effects on the expression of target genes in the TNBC tumor microenvironment. The pleiotropic nature of miRNAs expands their preclinical and clinical utility in combating both metastatic and non-metastatic TNBC cases and thereby improves their survival outcomes. The present review article aims to highlight the varying survival outcomes in metastatic and non-metastatic TNBC cases. The present review article emphasizes the therapeutic and prognostic potential of miRNAs in TNBC to improve survival outcomes by retarding distant metastasis to lung, bone, brain, and lymph nodes.
Collapse
Affiliation(s)
- Acharya Balkrishna
- Patanjali Herbal Research Department, Patanjali Research Institute, Haridwar 249 405, India
| | - Rashmi Mittal
- Patanjali Herbal Research Department, Patanjali Research Institute, Haridwar 249 405, India.
| | | | - Alan Prem Kumar
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore; Center for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119077, Singapore
| | - Anupam Bishayee
- Department of Pharmacology, College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, Bradenton, FL 34211, USA.
| |
Collapse
|
26
|
Duan X, Yu X, Gan J. Extracellular vesicle-packaged miR-4253 secreted by cancer-associated fibroblasts facilitates cell proliferation in gastric cancer by inducing macrophage M2 polarization. Cancer Biol Ther 2024; 25:2424490. [PMID: 39505708 PMCID: PMC11542604 DOI: 10.1080/15384047.2024.2424490] [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: 06/17/2024] [Revised: 10/28/2024] [Accepted: 10/29/2024] [Indexed: 11/08/2024] Open
Abstract
Cancer-associated fibroblasts (CAFs) can interact with macrophages in the tumor microenvironment by secreting extracellular vesicles (EVs), thereby affecting tumor progression. However, the mechanisms of CAF-secreted EVs in gastric cancer (GC) remain not well understood. Here, we investigated the effect of CAF-EVs on macrophage polarization in GC and the underlying mechanisms. Macrophage polarization was evaluated using flow cytometry and quantitative real-time polymerase chain reaction. GC cell proliferation was determined using cell counting kit-8, EdU, and colony formation assays. The molecular mechanism was explored using microarray analysis, dual-luciferase reporter assay, and RNA pull-down analysis. The results showed that CAFs secreted EVs that inhibit macrophage M1 polarization and promote M2 polarization. Moreover, miR-4253 expression was increased in CAF-EVs, and inhibition of miR-4253 reversed the macrophage polarization induced by EVs. IL6R was identified as the target of miR-4253. Additionally, macrophages treated with EVs that encapsulated miR-4253 promote GC cell proliferation. In conclusion, CAF-secreted EVs packaging miR-4253 facilitate macrophage polarization from M1 to M2 phenotype by targeting IL6R, thereby accelerating GC cell proliferation. The findings suggest that EV-encapsulated miR-4253 may be a promising therapeutic target of GC.
Collapse
Affiliation(s)
- Xinxing Duan
- General Surgery Center, Jiujiang City Key Laboratory of Cell Therapy, Jiujiang, China
| | - Xiong Yu
- General Surgery Center, Jiujiang City Key Laboratory of Cell Therapy, Jiujiang, China
| | - Jin Gan
- General Surgery Center, Jiujiang City Key Laboratory of Cell Therapy, Jiujiang, China
| |
Collapse
|
27
|
Chen Y, Guo Y, Li S, Xu J, Zhao C, Wang J, Yang J, Ning W, Qu Y, Zhang M, Wang S, Zhang H. Tumor-derived IL-6 promotes chordoma invasion by stimulating tumor-associated macrophages M2 polarization and TNFα secretion. Int Immunopharmacol 2024; 143:113315. [PMID: 39393273 DOI: 10.1016/j.intimp.2024.113315] [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: 01/29/2024] [Revised: 09/04/2024] [Accepted: 10/01/2024] [Indexed: 10/13/2024]
Abstract
AIMS Chordoma is a rare and aggressive bone tumor with high-recurrence and lack of effective treatment methods. Tumor associated macrophages (TAMs) are abundant in tumor microenvironment (TME) and polarize toward M2 in chordoma. It has been observed that the high proportion of M2 cells is associated with chordoma rapid progression. However, the mechanism of TAMs polarization and promotion to tumor progression in chordoma is still unclear. The is an urgent need for further research. MATERIALS AND METHODS Flow cytometry and immunohistochemical staining was used to detect the degree of macrophages infiltration in chordoma. A co-culture model of chordoma cells and macrophages was established in vitro to investigate the effects of their interaction on cell function, cytokine secretion, and RNA transcriptome expression. KEY FINDINGS In this study, we found M2 macrophage was predominantly abundant immune cell population in chordoma, and its proportion was associated with the degree of bone destruction. We demonstrated that interleukin 6 (IL-6) derived from chordoma cells could induce TAMs polarization by activating STAT3 phosphorylation, and TAMs could enhance chordoma cells migration and invasion through TNFα/NF-κB pathway. The interaction of chordoma cells and TAMs could promote the bone destruction-related factor Cathepsin B (CTSB) and inhibitory immune checkpoints expression. We also confirmed blocking IL-6/STAT3 pathway could significantly attenuate the M2 polarization of TAMs and decrease the secretion of TNFα. SIGNIFICANCE This study illustrates the dynamics between chordoma cells and TAMs in promoting chordoma invasion and suggests that IL-6/STAT3 pathway is a potential therapeutic target to reduce TAM-induced chordoma invasion.
Collapse
Affiliation(s)
- Yujia Chen
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing 100093, China
| | - Yuduo Guo
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing 100093, China
| | - Shenglun Li
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing 100093, China
| | - Jiacheng Xu
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing 100093, China
| | - Chao Zhao
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing 100093, China
| | - Jun Wang
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing 100093, China
| | - Jingjing Yang
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing 100093, China
| | - Weihai Ning
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing 100093, China
| | - Yanming Qu
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing 100093, China
| | - Mingshan Zhang
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing 100093, China
| | - Shengdian Wang
- CAS Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, 100101 Beijing, China.
| | - Hongwei Zhang
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing 100093, China.
| |
Collapse
|
28
|
Wang X, Lin L, Zhang X, Zhang M, Sun Z, Yang Y, Zhang X, Yuan Y, Zhang Y, Chen H, Wen T. Single-cell Atlas reveals core function of CPVL/MSR1 expressing macrophages in the prognosis of triple-negative breast cancer. Front Immunol 2024; 15:1501009. [PMID: 39776914 PMCID: PMC11703973 DOI: 10.3389/fimmu.2024.1501009] [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: 09/24/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025] Open
Abstract
Background Triple-negative breast cancer (TNBC) is the most aggressive subtype of breast cancer, with the worst prognosis among all subtypes. The impact of distinct cell subpopulations within the tumor microenvironment (TME) on TNBC patient prognosis has yet to be clarified. Methods Utilizing single-cell RNA sequencing (scRNA-seq) integrated with bulk RNA sequencing (bulk RNA-seq), we applied Cox regression models to compute hazard ratios, and cross-validated prognostic scoring using a GLMNET-based Cox model. Cell communication analysis was used to elucidate the potential mechanisms of CPVL and MSR1. Ultimately, RNA interference-mediated gene knockdown was utilized to validate the impact of specific genes on the polarization of tumor-associated macrophages (TAMs). Results Our findings revealed that the function of immune cells is more pivotal in prognosis, with TAMs showing the strongest correlation with TNBC patient outcomes, compared with other immune cells. Additionally, we identified CPVL and MSR1 as critical prognostic genes within TAMs, with CPVL expression positively correlated with favorable outcomes and MSR1 expression associated with poorer prognosis. Mechanistically, CPVL may contribute to favorable prognosis by inhibiting the SPP1-CD44 ligand-receptor and promoting CXCL9-CXCR3, C3-C3AR1 ligand-receptor, through which TAMs interact with other cells such as monocytes, neutrophils, and T cells. Moreover, cytokines including IL-18, IFNγR1, CCL20, and CCL2, along with complement-related gene like TREM2 and complement component CFD, may participate in the process of CPVL or MSR1 regulating macrophage polarization. Furthermore, RT-PCR experiments confirmed that CPVL is positively associated with M1-like TAM polarization, while MSR1 is linked to M2-like TAM polarization. Finally, the prognostic significance of these two genes is also validated in HER2-positive breast cancer subtypes. Conclusions CPVL and MSR1 are potential biomarkers for macrophage-mediated TNBC prognosis, suggesting the therapeutic potential of macrophage targeting in TNBC.
Collapse
Affiliation(s)
- Xinan Wang
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, Liaoning, China
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, The First Hospital of China Medical University, Shenyang, Liaoning, China
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, Liaoning, China
- Clinical Cancer Treatment and Research Center of Shenyang, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Li Lin
- Department of Pulmonary and Critical Care Medicine, Institute of Respiratory Disease, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Xue Zhang
- Department of Gynecology, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Minghui Zhang
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, Liaoning, China
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, The First Hospital of China Medical University, Shenyang, Liaoning, China
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, Liaoning, China
- Clinical Cancer Treatment and Research Center of Shenyang, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Zhuo Sun
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, Liaoning, China
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, The First Hospital of China Medical University, Shenyang, Liaoning, China
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, Liaoning, China
- Clinical Cancer Treatment and Research Center of Shenyang, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yichen Yang
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, Liaoning, China
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, The First Hospital of China Medical University, Shenyang, Liaoning, China
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, Liaoning, China
- Clinical Cancer Treatment and Research Center of Shenyang, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Xiuna Zhang
- Department of Medical Oncology, Second People’s Hospital of Huludao, Huludao, Liaoning, China
| | - Yonghui Yuan
- Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning, China
| | - Yong Zhang
- Department of Pathology, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning, China
| | - Hao Chen
- Department of Breast Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Ti Wen
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, Liaoning, China
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, The First Hospital of China Medical University, Shenyang, Liaoning, China
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, Liaoning, China
- Clinical Cancer Treatment and Research Center of Shenyang, The First Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
29
|
Liu P, Gao X, Yu Z, Liu Y, Liu Y, Lin J, Cao Y, Zhai S, Li J, Huang Y, Zou S, Wen C, Fu D, Lin J, Shen B. H19 promotes polarization and alternative splicing in tumor-associated macrophages, facilitating pancreatic cancer progression. Cancer Lett 2024; 611:217389. [PMID: 39653238 DOI: 10.1016/j.canlet.2024.217389] [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: 05/19/2024] [Revised: 10/09/2024] [Accepted: 12/06/2024] [Indexed: 12/16/2024]
Abstract
Tumor-associated macrophages (TAMs) play a crucial physiological role in the pancreatic tumor microenvironment. However, the role of long non-coding RNAs (lncRNAs) in TAMs within pancreatic tumors remains unclear. By lncRNA sequencing between TAMs and resident macrophages from normal tissues in pancreatic cancer, it is found that H19 is highly expressed in TAMs and is correlated with the prognosis and stages of pancreatic cancer. Constructing a co-culture model of THP-1 derived TAMs and pancreatic cancer cells, H19 promotes the polarization of TAMs towards the M2 phenotype and the secretion of IL-6, IL-10, and TGF-β, both in vivo and in vitro, indirectly enhancing pancreatic cancer proliferation and metastasis. Mechanistically, H19 competitively binds to the mRNA of YTHDC1 with MiR-107, and also interacts with the YTHDC1 protein, regulating the stability of SRSF1 and thereby affecting the alternative splicing of IL-6 and IL-10. Utilizing organoids and the patient-derived xenograft (PDX) model, it is found that ruxolitinib may represent a promising treatment option for PDAC patients with high H19 expression.
Collapse
Affiliation(s)
- Pengyi Liu
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China; Research Institute of Pancreatic Diseases, Shanghai Key Laboratory of Translational Research for Pancreatic Neoplasms, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China; State Key Laboratory of Oncogenes and Related Genes, Institute of Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200025, China.
| | - Xia Gao
- Department of Pathology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhengwei Yu
- Department of Cardiothoracic Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Yang Liu
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China; Research Institute of Pancreatic Diseases, Shanghai Key Laboratory of Translational Research for Pancreatic Neoplasms, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China; State Key Laboratory of Oncogenes and Related Genes, Institute of Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200025, China
| | - Yihao Liu
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China; Research Institute of Pancreatic Diseases, Shanghai Key Laboratory of Translational Research for Pancreatic Neoplasms, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China; State Key Laboratory of Oncogenes and Related Genes, Institute of Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200025, China
| | - Jiayu Lin
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China; Research Institute of Pancreatic Diseases, Shanghai Key Laboratory of Translational Research for Pancreatic Neoplasms, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China; State Key Laboratory of Oncogenes and Related Genes, Institute of Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200025, China
| | - Yizhi Cao
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China; Research Institute of Pancreatic Diseases, Shanghai Key Laboratory of Translational Research for Pancreatic Neoplasms, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China; State Key Laboratory of Oncogenes and Related Genes, Institute of Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200025, China
| | - Shuyu Zhai
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China; Research Institute of Pancreatic Diseases, Shanghai Key Laboratory of Translational Research for Pancreatic Neoplasms, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China; State Key Laboratory of Oncogenes and Related Genes, Institute of Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200025, China
| | - Jingwei Li
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China; Research Institute of Pancreatic Diseases, Shanghai Key Laboratory of Translational Research for Pancreatic Neoplasms, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China; State Key Laboratory of Oncogenes and Related Genes, Institute of Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200025, China
| | - Yishu Huang
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China; Research Institute of Pancreatic Diseases, Shanghai Key Laboratory of Translational Research for Pancreatic Neoplasms, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China; State Key Laboratory of Oncogenes and Related Genes, Institute of Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200025, China
| | - Siyi Zou
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China; Research Institute of Pancreatic Diseases, Shanghai Key Laboratory of Translational Research for Pancreatic Neoplasms, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China; State Key Laboratory of Oncogenes and Related Genes, Institute of Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200025, China
| | - Chenlei Wen
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China; Research Institute of Pancreatic Diseases, Shanghai Key Laboratory of Translational Research for Pancreatic Neoplasms, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China; State Key Laboratory of Oncogenes and Related Genes, Institute of Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200025, China
| | - Da Fu
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China; Research Institute of Pancreatic Diseases, Shanghai Key Laboratory of Translational Research for Pancreatic Neoplasms, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China; State Key Laboratory of Oncogenes and Related Genes, Institute of Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200025, China.
| | - Jiewei Lin
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China.
| | - Baiyong Shen
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China; Research Institute of Pancreatic Diseases, Shanghai Key Laboratory of Translational Research for Pancreatic Neoplasms, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China; State Key Laboratory of Oncogenes and Related Genes, Institute of Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200025, China.
| |
Collapse
|
30
|
Huang F, Wang F, Hu Q, Li Y, Jiang D. PTGR1-mediated immune evasion mechanisms in late-stage triple-negative breast cancer: mechanisms of M2 macrophage infiltration and CD8 + T cell suppression. Apoptosis 2024; 29:2002-2024. [PMID: 39068625 DOI: 10.1007/s10495-024-01991-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/03/2024] [Indexed: 07/30/2024]
Abstract
Triple-negative breast cancer (TNBC) is a heterogeneous disease characterized by metabolic dysregulation. Tumor cell immune escape plays an indispensable role in the development of TNBC tumors. Furthermore, in the abstract, we explicitly mention the techniques used and enhance the clarity and impact of our findings. "Based on bioinformatics analysis results, we utilized CRISPR/Cas9 technology to knockout the target gene and established a mouse model of breast cancer. Through experiments such as CCK8, scratch assay, and Transwell assay, we further investigated the impact of target gene knockout on the malignant behavior of tumor cells. Subsequently, we conducted immunohistochemistry and Western Blot experiments to study the expression of macrophage polarization and infiltration-related markers and evaluate the effect of the target gene on macrophage polarization. Next, through co-culture experiments, we simulated the tumor microenvironment and used immunohistochemistry staining to observe and analyze the distribution and activation status of M2 macrophages and CD8+ T cells in the co-culture system. We validated in vivo experiments the molecular mechanism by which the target gene regulates immune cell impact on TNBC progression.
Collapse
Affiliation(s)
- Fang Huang
- Department of Medical Oncology, The Fourth Hospital of Hebei Medical University, East Campus, No.169 Tianshan Street, Shijiazhuang, 050000, Hebei Province, P. R. China
| | - Fuhe Wang
- Department of General surgery, Hebei Yiling Hospital, Shijiazhuang, 050000, P. R. China
| | - Qilu Hu
- Department of Radiotherapy, Heze Traditional Chinese Medicine Hospital, Heze, 274008, P. R. China
| | - Ying Li
- Department of Medical Oncology, The Fourth Hospital of Hebei Medical University, East Campus, No.169 Tianshan Street, Shijiazhuang, 050000, Hebei Province, P. R. China
| | - Da Jiang
- Department of Medical Oncology, The Fourth Hospital of Hebei Medical University, East Campus, No.169 Tianshan Street, Shijiazhuang, 050000, Hebei Province, P. R. China.
| |
Collapse
|
31
|
Xu J, Xu X, Zhang H, Wu J, Pan R, Zhang B. Tumor-associated inflammation: The role and research progress in tumor therapy. J Drug Deliv Sci Technol 2024; 102:106376. [DOI: 10.1016/j.jddst.2024.106376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
32
|
Feng M, Cui H, Li S, Li L, Zhou C, Chen L, Cao Y, Gao Y, Li D. Ubiquitin-Activating Enzyme E1 (UBA1) as a Prognostic Biomarker and Therapeutic Target in Breast Cancer: Insights into Immune Infiltration and Functional Implications. Int J Mol Sci 2024; 25:12696. [PMID: 39684409 DOI: 10.3390/ijms252312696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Revised: 11/13/2024] [Accepted: 11/16/2024] [Indexed: 12/18/2024] Open
Abstract
Ubiquitin-Activating Enzyme E1 (UBA1), an E1 enzyme involved in the activation of ubiquitin enzymes, has been involved in the onset and progression of different cancers in humans. Nevertheless, the precise contribution of UBA1 in breast cancer (BC) is still poorly characterized. In this study, a thorough investigation was carried out to elucidate the significance of UBA1 and validate its functionality in BC. Through the analysis of mRNA sequencing data of BC patients, the mRNA expression of UBA1 was observed to be notably enhanced in cancer tissues relative to controls, and high UBA1 expression was linked to worse overall survival (OS), disease-specific survival (DSS), and progress-free survival (PFS). Moreover, UBA1 exhibited potential as an independent prognostic and diagnostic biomarker for individuals with BC. Additionally, functional enrichment analysis revealed the involvement of UBA1 in inflammation-linked pathways, like the TNF-α signaling pathway, the IL-6 signaling pathway, and various immune-related biological processes. Notably, single-sample gene set enrichment analysis (ssGSEA) aided in the identification of a negative link between UBA1 expression and the levels of infiltrating mast cells, Th1 cells, iDC cells, B cells, DC cells, Tem cells, Cytotoxic cells, T cells, CD8T cells, and pDC cells. Finally, this study demonstrated that silencing UBA1 significantly impeded the growth and development of BC cell lines. These findings highlight UBA1 as a potential prognostic biomarker linked to immune infiltration in BC, thereby depicting its potential as a new therapeutic target for individuals with BC.
Collapse
Affiliation(s)
- Mingtao Feng
- Department of Neurosurgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Huanhuan Cui
- Department of Neurosurgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Sen Li
- Department of Neurosurgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Liangdong Li
- Department of Neurosurgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Changshuai Zhou
- Department of Neurosurgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Lei Chen
- Department of Neurosurgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Yiqun Cao
- Department of Neurosurgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Yang Gao
- Department of Neurosurgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Deheng Li
- Department of Neurosurgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| |
Collapse
|
33
|
Friedman-DeLuca M, Karagiannis GS, Condeelis JS, Oktay MH, Entenberg D. Macrophages in tumor cell migration and metastasis. Front Immunol 2024; 15:1494462. [PMID: 39555068 PMCID: PMC11563815 DOI: 10.3389/fimmu.2024.1494462] [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: 09/10/2024] [Accepted: 10/15/2024] [Indexed: 11/19/2024] Open
Abstract
Tumor-associated macrophages (TAMs) are a phenotypically diverse, highly plastic population of cells in the tumor microenvironment (TME) that have long been known to promote cancer progression. In this review, we summarize TAM ontogeny and polarization, and then explore how TAMs enhance tumor cell migration through the TME, thus facilitating metastasis. We also discuss how chemotherapy and host factors including diet, obesity, and race, impact TAM phenotype and cancer progression. In brief, TAMs induce epithelial-mesenchymal transition (EMT) in tumor cells, giving them a migratory phenotype. They promote extracellular matrix (ECM) remodeling, allowing tumor cells to migrate more easily. TAMs also provide chemotactic signals that promote tumor cell directional migration towards blood vessels, and then participate in the signaling cascade at the blood vessel that allows tumor cells to intravasate and disseminate throughout the body. Furthermore, while chemotherapy can repolarize TAMs to induce an anti-tumor response, these cytotoxic drugs can also lead to macrophage-mediated tumor relapse and metastasis. Patient response to chemotherapy may be dependent on patient-specific factors such as diet, obesity, and race, as these factors have been shown to alter macrophage phenotype and affect cancer-related outcomes. More research on how chemotherapy and patient-specific factors impact TAMs and cancer progression is needed to refine treatment strategies for cancer patients.
Collapse
Affiliation(s)
- Madeline Friedman-DeLuca
- Integrated Imaging Program for Cancer Research, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States
- Department of Pathology, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States
- Montefiore Einstein Comprehensive Cancer Center, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States
- Cancer Dormancy Institute, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States
| | - George S. Karagiannis
- Integrated Imaging Program for Cancer Research, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States
- Montefiore Einstein Comprehensive Cancer Center, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States
- Cancer Dormancy Institute, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States
- Department of Microbiology and Immunology, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States
- Marilyn and Stanley M. Katz Institute for Immunotherapy of Cancer and Inflammatory Disorders, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States
| | - John S. Condeelis
- Integrated Imaging Program for Cancer Research, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States
- Montefiore Einstein Comprehensive Cancer Center, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States
- Cancer Dormancy Institute, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States
- Department of Surgery, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States
- Department of Cell Biology, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States
| | - Maja H. Oktay
- Integrated Imaging Program for Cancer Research, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States
- Department of Pathology, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States
- Montefiore Einstein Comprehensive Cancer Center, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States
- Cancer Dormancy Institute, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States
- Department of Surgery, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States
| | - David Entenberg
- Integrated Imaging Program for Cancer Research, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States
- Department of Pathology, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States
- Montefiore Einstein Comprehensive Cancer Center, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States
- Cancer Dormancy Institute, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States
| |
Collapse
|
34
|
Fu R, Chen Y, Zhao J, Xie X. The signature of SARS-CoV-2-related genes predicts the immune therapeutic response and prognosis in breast cancer. BMC Med Genomics 2024; 17:260. [PMID: 39482662 PMCID: PMC11526603 DOI: 10.1186/s12920-024-02032-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 10/15/2024] [Indexed: 11/03/2024] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is an exceptionally contagious single-stranded RNA virus with a strong positive contagion. The COVID-19 pandemic refers to the swift worldwide dissemination of SARS-CoV-2 infection, which began in late 2019. The COVID-19 epidemic has disrupted many cancer treatments. A few reports indicate that the prevalence of SARS-CoV-2 has disrupted the treatment of breast cancer patients (BCs). However, the role of SARS-CoV-2 in the occurrence and prognosis of BC has not been elucidated. Here, we applied bioinformatics to construct a prognostic signature of SARS-CoV-2-related genes (SCRGs). Specifically, weighted gene co-expression network analysis (WGCNA) was utilized to extract co-expressed genes of differentially expressed genes (DEGs) in breast cancer and SCRGs. Then, we utilized the least absolute shrinkage and selection operator (LASSO) algorithm and univariate regression analysis to screen out three hub genes (DCTPP1, CLIP4 and ANO6) and constructed a risk score model. We further analyzed tumor immune invasion, HLA-related genes, immune checkpoint inhibitors (ICIs), and sensitivity to anticancer drugs in different SARS-CoV-2 related risk subgroups. In addition, we have developed a nomination map to expand clinical applicability. The results of our study indicate that BCs with a high-risk score are linked to negative outcomes, lower immune scores, and reduced responsiveness to anticancer medications. This suggests that the SARS-CoV-2 related signature could be used to guide prognosis assessment and treatment decisions for BCs.
Collapse
Affiliation(s)
- Ruizhi Fu
- Department of General Surgery, The First Affiliated Hospital of Shantou University Medical College, Shantou, 515000, China
| | - Yequn Chen
- Department of Cardiology, The First Affiliated Hospital of Shantou University Medical College, Shantou, 515000, China
| | - Jiajing Zhao
- Department of General Surgery, The First Affiliated Hospital of Shantou University Medical College, Shantou, 515000, China.
| | - Xiaojun Xie
- Department of General Surgery, The First Affiliated Hospital of Shantou University Medical College, Shantou, 515000, China.
| |
Collapse
|
35
|
Qian H, Maghsoudloo M, Kaboli PJ, Babaeizad A, Cui Y, Fu J, Wang Q, Imani S. Decoding the Promise and Challenges of miRNA-Based Cancer Therapies: An Essential Update on miR-21, miR-34, and miR-155. Int J Med Sci 2024; 21:2781-2798. [PMID: 39512697 PMCID: PMC11539376 DOI: 10.7150/ijms.102123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 10/14/2024] [Indexed: 11/15/2024] Open
Abstract
MicroRNAs (miRNAs)-based therapies hold great promise for cancer treatment, challenges such as expression variability, off-target effects, and limited clinical effectiveness have led to the withdrawal of many clinical trials. This review investigates the setbacks in miRNA-based therapies by examining miR-21, miR-34, and miR-155, highlighting their functional complexity, off-target effects, and the challenges in delivering these therapies effectively. Moreover, It highlights recent advances in delivery methods, combination therapies, and personalized treatment approaches to overcome these challenges. This review highlights the intricate molecular networks involving miRNAs, particularly their interactions with other non-coding RNAs, such as long non-coding RNAs (lncRNAs) and circular RNAs (circRNAs), emphasizing the pivotal role of miRNAs in cancer biology and therapeutic strategies. By addressing these hurdles, this review aims to steer future research toward harnessing the potential of miRNA therapies to target cancer pathways effectively, enhance anti-tumor responses, and ultimately improve patient outcomes in precision cancer therapy.
Collapse
Affiliation(s)
- Hongbo Qian
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, Zhejiang, China
| | - Mazaher Maghsoudloo
- Key Laboratory of Epigenetics and Oncology, The Research Center for Preclinical Medicine, Southwest Medical University, Luzhou 646000, Sichuan, China
| | - Parham Jabbarzadeh Kaboli
- Department of Biochemistry, Faculty of Medicine, Medical University of Warsaw, Warsaw 02-091, Poland
| | - Ali Babaeizad
- Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Yulan Cui
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, Zhejiang, China
| | - Junjiang Fu
- Key Laboratory of Epigenetics and Oncology, The Research Center for Preclinical Medicine, Southwest Medical University, Luzhou 646000, Sichuan, China
| | - Qingjing Wang
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, Zhejiang, China
| | - Saber Imani
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, Zhejiang, China
| |
Collapse
|
36
|
Liu H, Wang L. MicroRNA-34a negatively regulates Netrin1 and mediates MEK/ERK pathway to regulate chemosensitivity of gastric cancer cells. Discov Oncol 2024; 15:563. [PMID: 39404782 PMCID: PMC11480279 DOI: 10.1007/s12672-024-01451-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 10/11/2024] [Indexed: 10/19/2024] Open
Abstract
OBJECTIVE To explore the mechanism of action of MicroRNAs-34a (miR-34a) and Eurite growth guiding factor 1 (Netrin1) in cisplatin resistance in gastric cancer (GC), providing new clues for overcoming tumor resistance and optimizing anti-tumor therapy for GC. METHODS The Cancer Genome Atlas (TCGA), Differentially Expressed MicroRNAs (miRNAs) in human cancers (dbDEMC), and Starbase online databases were used to analyze the correlation between miR-34a and Netrin-1 and prognosis in GC, and to predict and verify the targeted binding of miR-34a to Netrin-1. The experimental methods including Cell transfection, real-time polymerase chain reaction (RT-PCR), Cell-Counting-Kit-8 (CCK8) assay, flow cytometry, wound scratch assay, transwell assay, and western blotting were used to investigate the effects of miR-34a and Netrin1 on chemotherapy resistance and biological characteristics in cisplatin-resistant GC cells (HGC27/DDP), and to analyze the molecular mechanism of cisplatin resistance. RESULTS miR-34a expression was downregulated in gastric cancer clinical samples and cisplatin-resistant cells, while Netrin1 was upregulated, and was related to overall survival (OS). Upregulation of miR-34a can significantly reduce the IC50 value of cisplatin(0.65 vs 1.6 ng/mL) and Multidrug Resistance 1 (MDR-1) protein level, inhibit the proliferation activity, reduce the expression levels of proliferating cell nuclear antigen (PCNA) and ki-67 protein, and induce the increase of apoptosis rate and the enhancement of cycle arrest. Upregulation of miR-34a can also significantly reduce the expression level of Matrix metalloproteinase 9 (MMP9) protein, promote the expression of E-cadherin protein, reduce the wound healing rate and invasion number to inhibit migration and invasion ability in drug-resistant gastric cancer cells. Moreover, overexpression of Netrin1 on the basis of upregulation of miR-34a can weaken the above changes caused by upregulation of miR-34a. In addition, upregulation of miR-34a can significantly inhibit the Mitogen-activated protein kinase kinase (MEK) / Extracellular regulated protein kinases (ERK) pathway, while overexpression of Netrin1 can activate the MEK/ERK pathway, and inhibition of MEK/ERK pathway can effectively counteract the protein expression of Netrin1, and reverse changes in the expression of cisplatin IC50 and MDR-1 proteins caused by co-upregulation of miR-34a/Netrin1 in HGC27/DDP, as well as changes in proliferation, apoptosis, migration and invasion. In addition, upregulation of miR-34a can significantly inhibit the MEK/ERK pathway, while overexpression of Netrin1 can activate the MEK/ERK pathway. If the MEK/ERK pathway was inhibited, it can effectively counteract the protein overexpression of Netrin1, and reverse the changes in the expression of cisplatin IC50 and MDR-1 proteins in HGC27/DDP induced by co-upregulation of miR-34a / Netrin1, as well as changes in proliferation, apoptosis, migration and invasion. CONCLUSION miR-34a targets and negatively regulates Netrin1 to mediate the proliferation, apoptosis, apoptosis, migration, and invasion of drug-resistant gastric cancer cells via the MEK/ERK pathway, and change the chemosensitivity in GC cells. miR-34a/Netrin1/MEK/ERK axis may serve as a novel therapeutic target for chemoresistance in GC, it is of great significance for overcoming drug resistance and developing new therapeutic strategies for GC.
Collapse
Affiliation(s)
- Haiping Liu
- Key Laboratory of Microecology-Immune Regulatory Network and Related Diseases, School of Basic Medicine, Jiamusi University, 348 Dexiang Street, Xiangyang District, Jiamusi City, 154000, Heilongjiang Province, People's Republic of China
| | - Limin Wang
- Key Laboratory of Microecology-Immune Regulatory Network and Related Diseases, School of Basic Medicine, Jiamusi University, 348 Dexiang Street, Xiangyang District, Jiamusi City, 154000, Heilongjiang Province, People's Republic of China.
| |
Collapse
|
37
|
Lan W, Yang L, Tan X. Crosstalk between ferroptosis and macrophages: potential value for targeted treatment in diseases. Mol Cell Biochem 2024; 479:2523-2543. [PMID: 37880443 DOI: 10.1007/s11010-023-04871-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 10/05/2023] [Indexed: 10/27/2023]
Abstract
Ferroptosis is a newly identified form of programmed cell death that is connected to iron-dependent lipid peroxidization. It involves a variety of physiological processes involving iron metabolism, lipid metabolism, oxidative stress, and biosynthesis of nicotinamide adenine dinucleotide phosphate, glutathione, and coenzyme Q10. So far, it has been discovered to contribute to the pathological process of many diseases, such as myocardial infarction, acute kidney injury, atherosclerosis, and so on. Macrophages are innate immune system cells that regulate metabolism, phagocytize pathogens and dead cells, mediate inflammatory reactions, promote tissue repair, etc. Emerging evidence shows strong associations between macrophages and ferroptosis, which can provide us with a deeper comprehension of the pathological process of diseases and new targets for the treatments. In this review, we summarized the crosstalk between macrophages and ferroptosis and anatomized the application of this association in disease treatments, both non-neoplastic and neoplastic diseases. In addition, we have also addressed problems that remain to be investigated, in the hope of inspiring novel therapeutic strategies for diseases.
Collapse
Affiliation(s)
- Wanxin Lan
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Operative Dentistry and Endodontics West China Hospital of Stomatology, Sichuan University, 14# 3rd Section, Renmin South Road, Chengdu, 610041, Sichuan, China
| | - Lei Yang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Operative Dentistry and Endodontics West China Hospital of Stomatology, Sichuan University, 14# 3rd Section, Renmin South Road, Chengdu, 610041, Sichuan, China
| | - Xuelian Tan
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Operative Dentistry and Endodontics West China Hospital of Stomatology, Sichuan University, 14# 3rd Section, Renmin South Road, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
38
|
Wang L, Tang D. Akkermania muciniphila: a rising star in tumor immunology. Clin Transl Oncol 2024; 26:2418-2430. [PMID: 38653927 DOI: 10.1007/s12094-024-03493-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 04/01/2024] [Indexed: 04/25/2024]
Abstract
Tumor is accompanied by complex and dynamic microenvironment development, and the interaction of all its components influences disease progression and response to treatment. Once the tumor microenvironment has been eradicated, various mechanisms can induce the tumors. Microorganisms can maintain the homeostasis of the tumor microenvironment through immune regulation, thereby inhibiting tumor development. Akkermania muciniphila (A. muciniphila), an anaerobic bacterium, can induce tumor immunity, regulate the gastrointestinal microenvironment through metabolites, outer membrane proteins, and some cytokines, and enhance the curative effect through combined immunization. Therefore, a comprehensive understanding of the complex interaction between A. muciniphila and human immunity will facilitate the development of immunotherapeutic strategies in the future and enable patients to obtain a more stable clinical response. This article reviews the most recent developments in the tumor immunity of A. muciniphila.
Collapse
Affiliation(s)
- Leihan Wang
- Clinical Medical College, Yangzhou University, Yangzhou, People's Republic of China
| | - Dong Tang
- Department of General Surgery, Institute of General Surgery, Clinical Medical College, Northern Jiangsu People's Hospital, Yangzhou University, Yangzhou, 225001, People's Republic of China.
| |
Collapse
|
39
|
Maleki R, Ghith A, Heydarlou H, Grzeskowiak LE, Ingman WV. The Role of Breastmilk in Macrophage-Tumour Cell Interactions in Postpartum Breast Cancer. FRONT BIOSCI-LANDMRK 2024; 29:328. [PMID: 39344339 DOI: 10.31083/j.fbl2909328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 08/22/2024] [Accepted: 09/03/2024] [Indexed: 10/01/2024]
Abstract
BACKGROUND Lactation is associated with long-term reduced risk of breast cancer. However, there is a transient increased risk of breast cancer in the 5 to 10 years postpartum and this is associated with a high incidence of metastasis and mortality. Breastmilk is a physiological fluid secreted by the mammary glands intimately connected with breast cells and the microenvironment that may affect postpartum breast cancer development and progression. This study aims to investigate the effect of breastmilk on interactions between breast cancer cells and macrophages in vitro. METHODS Human breastmilk from healthy donors (n = 7) was pooled and incubated with breast cancer (MCF-7 and MDA-MB-231) and macrophage (RAW264.7) cell lines to assess cell proliferation, viability, migration, and expression of key genes associated with epithelial-mesenchymal transition (EMT) and macrophage phenotype. Indirect co-culture studies assessed the effect of breastmilk on interactions between breast cancer cells and macrophages. RESULTS Breastmilk increased the proliferation and viability of breast cancer cells, reduced EMT markers, and reduced cell migration in MDA-MB-231 cells. Breastmilk decreased mRNA expression of interleukin 1B (IL1B) and interleukin 10 (IL10) in macrophages. Reduced EMT marker expression was observed in breast cancer cells co-cultured with macrophages pre-treated with breastmilk. Macrophages co-cultured with breast cancer cells pre-treated with breastmilk exhibited increased expression of a pro-inflammatory cytokine tumor necrosis factor A (TNFA) and pro-inflammatory nitric oxide synthase 2 (NOS2), and reduced expression of cytokines IL10 and transforming growth factor B1 (TGFB1) which are associated with the alternatively-activated macrophage phenotype. CONCLUSIONS Breastmilk has the potential to promote breast cancer proliferation, however, it can also reduce breast cancer progression through inhibition of breast cancer cell migration and regulation of macrophage polarisation. These findings suggest that breastmilk has potential to shape the tumour microenvironment in postpartum breast cancer.
Collapse
Affiliation(s)
- Reza Maleki
- Discipline of Surgical Specialties, Adelaide Medical School, University of Adelaide, The Queen Elizabeth Hospital, Woodville South, SA 5011, Australia
- Robinson Research Institute, University of Adelaide, Adelaide, SA 5006, Australia
| | - Amna Ghith
- Discipline of Surgical Specialties, Adelaide Medical School, University of Adelaide, The Queen Elizabeth Hospital, Woodville South, SA 5011, Australia
- Robinson Research Institute, University of Adelaide, Adelaide, SA 5006, Australia
| | - Hanieh Heydarlou
- Discipline of Surgical Specialties, Adelaide Medical School, University of Adelaide, The Queen Elizabeth Hospital, Woodville South, SA 5011, Australia
- Robinson Research Institute, University of Adelaide, Adelaide, SA 5006, Australia
| | - Luke E Grzeskowiak
- College of Medicine and Public Health, Flinders Health and Medical Research Institute, Flinders University, Bedford Park, SA 5042, Australia
- SAHMRI Women and Kids, South Australian Health and Medical Research Institute, Women's and Children's Hospital, North Adelaide, SA 5006, Australia
| | - Wendy V Ingman
- Discipline of Surgical Specialties, Adelaide Medical School, University of Adelaide, The Queen Elizabeth Hospital, Woodville South, SA 5011, Australia
- Robinson Research Institute, University of Adelaide, Adelaide, SA 5006, Australia
| |
Collapse
|
40
|
Li C, Yu X, Han X, Lian C, Wang Z, Shao S, Shao F, Wang H, Ma S, Liu J. Innate immune cells in tumor microenvironment: A new frontier in cancer immunotherapy. iScience 2024; 27:110750. [PMID: 39280627 PMCID: PMC11399700 DOI: 10.1016/j.isci.2024.110750] [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] [Indexed: 09/18/2024] Open
Abstract
Innate immune cells, crucial in resisting infections and initiating adaptive immunity, play diverse and significant roles in tumor development. These cells, including macrophages, granulocytes, dendritic cells (DCs), innate lymphoid cells, and innate-like T cells, are pivotal in the tumor microenvironment (TME). Innate immune cells are crucial components of the TME, based on which various immunotherapy strategies have been explored. Immunotherapy strategies, such as novel immune checkpoint inhibitors, STING/CD40 agonists, macrophage-based surface backpack anchoring, ex vivo polarization approaches, DC-based tumor vaccines, and CAR-engineered innate immune cells, aim to enhance their anti-tumor potential and counteract cancer-induced immunosuppression. The proximity of innate immune cells to tumor cells in the TME also makes them excellent drug carriers. In this review, we will first provide a systematic overview of innate immune cells within the TME and then discuss innate cell-based therapeutic strategies. Furthermore, the research obstacles and perspectives within the field will also be addressed.
Collapse
Affiliation(s)
- Changhui Li
- Department of Respiratory and Critical Care Medicine, the Second Affiliated Hospital, and Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310029, China
- Edinburgh Medical School: Biomedical Sciences, College of Medicine and Veterinary Medicine, The University of Edinburgh, Edinburgh, UK
| | - Xinyu Yu
- Department of Respiratory and Critical Care Medicine, the Second Affiliated Hospital, and Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310029, China
- Edinburgh Medical School: Biomedical Sciences, College of Medicine and Veterinary Medicine, The University of Edinburgh, Edinburgh, UK
| | - Xinyan Han
- Department of Respiratory and Critical Care Medicine, the Second Affiliated Hospital, and Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310029, China
- Edinburgh Medical School: Biomedical Sciences, College of Medicine and Veterinary Medicine, The University of Edinburgh, Edinburgh, UK
| | - Chen Lian
- Department of Respiratory and Critical Care Medicine, the Second Affiliated Hospital, and Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310029, China
| | - Zijin Wang
- Department of Respiratory and Critical Care Medicine, the Second Affiliated Hospital, and Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310029, China
- Edinburgh Medical School: Biomedical Sciences, College of Medicine and Veterinary Medicine, The University of Edinburgh, Edinburgh, UK
| | - Shiqun Shao
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
| | - Fangwei Shao
- National Key Laboratory of Biobased Transportation Fuel Technology, ZJU-UIUC Institute, Zhejiang University, Hangzhou 310027, China
| | - Hua Wang
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Shenglin Ma
- Department of Thoracic Oncology, Hangzhou Cancer Hospital, Hangzhou 310002, China
| | - Jian Liu
- Department of Respiratory and Critical Care Medicine, the Second Affiliated Hospital, and Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310029, China
- Edinburgh Medical School: Biomedical Sciences, College of Medicine and Veterinary Medicine, The University of Edinburgh, Edinburgh, UK
- Biomedical and Heath Translational Research Center of Zhejiang Province, Haining, China
- Cancer Center, Zhejiang University, Hangzhou 310058, China
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
41
|
Tosi G, Paoli A, Zuccolotto G, Turco E, Simonato M, Tosoni D, Tucci F, Lugato P, Giomo M, Elvassore N, Rosato A, Cogo P, Pece S, Santoro MM. Cancer cell stiffening via CoQ 10 and UBIAD1 regulates ECM signaling and ferroptosis in breast cancer. Nat Commun 2024; 15:8214. [PMID: 39294175 PMCID: PMC11410950 DOI: 10.1038/s41467-024-52523-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 09/11/2024] [Indexed: 09/20/2024] Open
Abstract
CoQ10 (Coenzyme Q10) is an essential fat-soluble metabolite that plays a key role in cellular metabolism. A less-known function of CoQ10 is whether it may act as a plasma membrane-stabilizing agent and whether this property can affect cancer development and progression. Here, we show that CoQ10 and its biosynthetic enzyme UBIAD1 play a critical role in plasmamembrane mechanical properties that are of interest for breast cancer (BC) progression and treatment. CoQ10 and UBIAD1 increase membrane fluidity leading to increased cell stiffness in BC. Furthermore, CoQ10 and UBIAD1 states impair ECM (extracellular matrix)-mediated oncogenic signaling and reduce ferroptosis resistance in BC settings. Analyses on human patients and mouse models reveal that UBIAD1 loss is associated with BC development and progression and UBIAD1 expression in BC limits CTCs (circulating tumor cells) survival and lung metastasis formation. Overall, this study reveals that CoQ10 and UBIAD1 can be further investigated to develop therapeutic interventions to treat BC patients with poor prognosis.
Collapse
Affiliation(s)
- Giovanni Tosi
- Laboratory of Angiogenesis and Cancer Metabolism, Department of Biology, University of Padova, Padova, Italy
| | - Alessandro Paoli
- Laboratory of Angiogenesis and Cancer Metabolism, Department of Biology, University of Padova, Padova, Italy
| | - Gaia Zuccolotto
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology IOV - IRCCS, Padova, Italy
| | - Emilia Turco
- Molecular Biotechnology Center, University of Turin, Torino, Italy
| | - Manuela Simonato
- Pediatric Research Institute "Città della Speranza", Padova, Italy
| | | | | | - Pietro Lugato
- Laboratory of Angiogenesis and Cancer Metabolism, Department of Biology, University of Padova, Padova, Italy
| | - Monica Giomo
- Department of Industrial Engineering, University of Padova, Padova, Italy
| | - Nicola Elvassore
- Department of Industrial Engineering, University of Padova, Padova, Italy
- Veneto Institute of Molecular Medicine, Padova, Italy
| | - Antonio Rosato
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology IOV - IRCCS, Padova, Italy
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
| | - Paola Cogo
- Pediatric Research Institute "Città della Speranza", Padova, Italy
- Division of Pediatrics, Department of Medicine, Udine University, Udine, Italy
| | - Salvatore Pece
- IEO, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Haemato-Oncology, University of Milan, Milano, Italy
| | - Massimo M Santoro
- Laboratory of Angiogenesis and Cancer Metabolism, Department of Biology, University of Padova, Padova, Italy.
- Veneto Institute of Molecular Medicine, Padova, Italy.
| |
Collapse
|
42
|
Li DD, Zhou T, Gao J, Wu GL, Yang GR. Circadian rhythms and breast cancer: from molecular level to therapeutic advancements. J Cancer Res Clin Oncol 2024; 150:419. [PMID: 39266868 PMCID: PMC11393214 DOI: 10.1007/s00432-024-05917-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: 06/25/2024] [Accepted: 08/05/2024] [Indexed: 09/14/2024]
Abstract
BACKGROUND AND OBJECTIVES Circadian rhythms, the endogenous biological clocks that govern physiological processes, have emerged as pivotal regulators in the development and progression of breast cancer. This comprehensive review delves into the intricate interplay between circadian disruption and breast tumorigenesis from multifaceted perspectives, encompassing biological rhythms, circadian gene regulation, tumor microenvironment dynamics, and genetic polymorphisms. METHODS AND RESULTS Epidemiological evidence underscores the profound impact of external factors, such as night shift work, jet lag, dietary patterns, and exercise routines, on breast cancer risk and progression through the perturbation of circadian homeostasis. The review elucidates the distinct roles of key circadian genes, including CLOCK, BMAL1, PER, and CRY, in breast cancer biology, highlighting their therapeutic potential as molecular targets. Additionally, it investigates how circadian rhythm dysregulation shapes the tumor microenvironment, fostering epithelial-mesenchymal transition, chronic inflammation, and immunosuppression, thereby promoting tumor progression and metastasis. Furthermore, the review sheds light on the association between circadian gene polymorphisms and breast cancer susceptibility, paving the way for personalized risk assessment and tailored treatment strategies. CONCLUSIONS Importantly, it explores innovative therapeutic modalities that harness circadian rhythms, including chronotherapy, melatonin administration, and traditional Chinese medicine interventions. Overall, this comprehensive review emphasizes the critical role of circadian rhythms in the pathogenesis of breast cancer and highlights the promising prospects for the development of circadian rhythm-based interventions to enhance treatment efficacy and improve patient outcomes.
Collapse
Affiliation(s)
- Dou-Dou Li
- School of Clinical Medicine, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Teng Zhou
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jing Gao
- School of Clinical Medicine, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Guan-Lin Wu
- School of Clinical Medicine, Shanghai University of Medicine and Health Sciences, Shanghai, China.
| | - Guang-Rui Yang
- School of Clinical Medicine, Shanghai University of Medicine and Health Sciences, Shanghai, China.
| |
Collapse
|
43
|
Deswal B, Bagchi U, Santra MK, Garg M, Kapoor S. Inhibition of STAT3 by 2-Methoxyestradiol suppresses M2 polarization and protumoral functions of macrophages in breast cancer. BMC Cancer 2024; 24:1129. [PMID: 39256694 PMCID: PMC11389501 DOI: 10.1186/s12885-024-12871-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: 08/05/2023] [Accepted: 08/28/2024] [Indexed: 09/12/2024] Open
Abstract
BACKGROUND Breast cancer metastasis remains the leading cause of cancer-related deaths in women worldwide. Infiltration of tumor-associated macrophages (TAMs) in the tumor stroma is known to be correlated with reduced overall survival. The inhibitors of TAMs are sought after for reprogramming the tumor microenvironment. Signal transducer and activator of transcription 3 (STAT3) is well known to contribute in pro-tumoral properties of TAMs. 2-Methoxyestradiol (2ME2), a potent anticancer and antiangiogenic agent, has been in clinical trials for treatment of breast cancer. Here, we investigated the potential of 2ME2 in modulating the pro-tumoral effects of TAMs in breast cancer. METHODS THP-1-derived macrophages were polarized to macrophages with or without 2ME2. The effect of 2ME2 on macrophage surface markers and anti-inflammatory genes was determined by Western blotting, flow cytometry, immunofluorescence, qRT‒PCR. The concentration of cytokines secreted by cells was monitored by ELISA. The effect of M2 macrophages on malignant properties of breast cancer cells was determined using colony formation, wound healing, transwell, and gelatin zymography assays. An orthotopic model of breast cancer was used to determine the effect of 2ME2 on macrophage polarization and metastasis in vivo. RESULTS First, our study found that polarization of monocytes to alternatively activated M2 macrophages is associated with the reorganization of the microtubule cytoskeleton. At lower concentrations, 2ME2 treatment depolymerized microtubules and reduced the expression of CD206 and CD163, suggesting that it inhibits the polarization of macrophages to M2 phenotype. However, the M1 polarization was not significantly affected at these concentrations. Importantly, 2ME2 inhibited the expression of several anti-inflammatory cytokines and growth factors, including CCL18, TGF-β, IL-10, FNT, arginase, CXCL12, MMP9, and VEGF-A, and hindered the metastasis-promoting effects of M2 macrophages. Concurrently, 2ME2 treatment reduced the expression of CD163 in tumors and inhibited lung metastasis in the orthotopic breast cancer model. Mechanistically, 2ME2 treatment reduced the phosphorylation and nuclear translocation of STAT3, an effect which was abrogated by colivelin. CONCLUSIONS Our study presents novel findings on mechanism of 2ME2 from the perspective of its effects on the polarization of the TAMs via the STAT3 signaling in breast cancer. Altogether, the data supports further clinical investigation of 2ME2 and its derivatives as therapeutic agents to modulate the tumor microenvironment and immune response in breast carcinoma.
Collapse
Affiliation(s)
- Bhawna Deswal
- Amity Institute of Molecular Medicine and Stem Cell Research, Amity University Uttar Pradesh, Noida, Uttar Pradesh, 201313, India
| | - Urmi Bagchi
- Amity Institute of Molecular Medicine and Stem Cell Research, Amity University Uttar Pradesh, Noida, Uttar Pradesh, 201313, India
| | - Manas Kumar Santra
- National Centre for Cell Science Complex, Savitribai Phule Pune University, Campus Ganeshkhind Road, Pune, Maharashtra, 411007, India
| | - Manoj Garg
- Amity Institute of Molecular Medicine and Stem Cell Research, Amity University Uttar Pradesh, Noida, Uttar Pradesh, 201313, India.
| | - Sonia Kapoor
- Amity Institute of Molecular Medicine and Stem Cell Research, Amity University Uttar Pradesh, Noida, Uttar Pradesh, 201313, India.
| |
Collapse
|
44
|
Stefanes NM, Cunha-Silva ME, de Oliveira Silva L, Walter LO, Santos-Silva MC, Gartia MR. Circulating biomarkers for diagnosis and response to therapies in cancer patients. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2024; 391:1-41. [PMID: 39939074 PMCID: PMC11969414 DOI: 10.1016/bs.ircmb.2024.08.007] [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] [Indexed: 01/03/2025]
Abstract
Cancer presents a significant challenge to global health, driving worldwide concerted efforts to advance early detection, predict therapeutic response, and identify novel targeted therapies. Liquid biopsies emerge as promising avenues for discerning cancer biomarkers, offering less invasive approaches compared to conventional methods. Utilizing increasingly robust technologies, diverse bodily fluids can unveil genetic variants, epigenetic modifications, transcriptional alterations, and metabolomic signatures associated with cancer, thereby furnishing valuable insights for clinical management. This chapter intends to review the sources of cancer-related biomarkers found in circulation, prevalent techniques utilized for their identification, and the potential implications of different biomarker types on the management of cancer. Certain biomarkers currently used in clinical practice will be addressed, as well as potential biomarkers still in the study phase, and the inherent challenges in their practical implementation.
Collapse
Affiliation(s)
- Natália Marcéli Stefanes
- Department of Mechanical and Industrial Engineering, Louisiana State University, Baton Rouge, LA, United States
| | - Maria Eduarda Cunha-Silva
- Post-Graduation Program in Pharmacy, Health Science Center, Federal University of Santa Catarina, Florianópolis, SC, Brazil
| | - Lisandra de Oliveira Silva
- Post-Graduation Program in Pharmacy, Health Science Center, Federal University of Santa Catarina, Florianópolis, SC, Brazil
| | - Laura Otto Walter
- Post-Graduation Program in Pharmacy, Health Science Center, Federal University of Santa Catarina, Florianópolis, SC, Brazil
| | - Maria Cláudia Santos-Silva
- Post-Graduation Program in Pharmacy, Health Science Center, Federal University of Santa Catarina, Florianópolis, SC, Brazil
| | - Manas Ranjan Gartia
- Department of Mechanical and Industrial Engineering, Louisiana State University, Baton Rouge, LA, United States.
| |
Collapse
|
45
|
Zhang Y, Ding X, Zhang X, Li Y, Xu R, Li HJ, Zuo D, Chen G. Unveiling the contribution of tumor-associated macrophages in driving epithelial-mesenchymal transition: a review of mechanisms and therapeutic Strategies. Front Pharmacol 2024; 15:1404687. [PMID: 39286635 PMCID: PMC11402718 DOI: 10.3389/fphar.2024.1404687] [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: 03/21/2024] [Accepted: 08/15/2024] [Indexed: 09/19/2024] Open
Abstract
Tumor-associated macrophages (TAMs), fundamental constituents of the tumor microenvironment (TME), significantly influence cancer development, primarily by promoting epithelial-mesenchymal transition (EMT). EMT endows cancer cells with increased motility, invasiveness, and resistance to therapies, marking a pivotal juncture in cancer progression. The review begins with a detailed exposition on the origins of TAMs and their functional heterogeneity, providing a foundational understanding of TAM characteristics. Next, it delves into the specific molecular mechanisms through which TAMs induce EMT, including cytokines, chemokines and stromal cross-talking. Following this, the review explores TAM-induced EMT features in select cancer types with notable EMT characteristics, highlighting recent insights and the impact of TAMs on cancer progression. Finally, the review concludes with a discussion of potential therapeutic targets and strategies aimed at mitigating TAM infiltration and disrupting the EMT signaling network, thereby underscoring the potential of emerging treatments to combat TAM-mediated EMT in cancer. This comprehensive analysis reaffirms the necessity for continued exploration into TAMs' regulatory roles within cancer biology to refine therapeutic approaches and improve patient outcomes.
Collapse
Affiliation(s)
- Yijia Zhang
- Department of Pharmacy, Taizhou Second People's Hospital (Mental Health Center affiliated to Taizhou University School of Medicine), Taizhou University, Taizhou, Zhejiang, China
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, China
| | - Xiaofei Ding
- Department of Pharmacology, Taizhou University, Taizhou, Zhejiang, China
| | - Xue Zhang
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, China
| | - Ye Li
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, China
| | - Rui Xu
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, China
| | - Hai-Jun Li
- Department of Pharmacy, Taizhou Second People's Hospital (Mental Health Center affiliated to Taizhou University School of Medicine), Taizhou University, Taizhou, Zhejiang, China
| | - Daiying Zuo
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, China
| | - Guang Chen
- Department of Pharmacy, Taizhou Second People's Hospital (Mental Health Center affiliated to Taizhou University School of Medicine), Taizhou University, Taizhou, Zhejiang, China
- Department of Pharmacology, Taizhou University, Taizhou, Zhejiang, China
| |
Collapse
|
46
|
Chen J, Sun JJ, Ma YW, Zhu MQ, Hu J, Lu QJ, Cai ZG. Cancer-associated fibroblasts derived exosomal LINC01833 promotes the occurrence of non-small cell lung cancer through miR-335-5p -VAPA axis. J Biochem Mol Toxicol 2024; 38:e23769. [PMID: 39152098 DOI: 10.1002/jbt.23769] [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: 01/22/2024] [Revised: 03/25/2024] [Accepted: 07/05/2024] [Indexed: 08/19/2024]
Abstract
Cancer-associated fibroblasts (CAFs) are an important component of the tumor microenvironment (TME) and can induce functional polarization of tumor macrophages. This study aimed to explore the effect of CAFs-derived exosome LINC01833 on the malignant biological behavior of non-small cell lung cancer (NSCLC) cells and its mechanism. Tumor tissues (n = 3) and adjacent noncancerous tissues (n = 3) were collected from patients with NSCLC, and fibroblasts (CAF, NF) were isolated from the two tissues. Expression of LINC01833/miR-335-5p/VAPA in NSCLC clinical tissues and cell lines was detected by RT-qPCR. Exosomes of CAFs and NFs were isolated by ultracentrifugation. Cell proliferation, migration, invasion, and M2 macrophage polarization were detected by MTT, transwell, wound-healing assay, and flow cytometry assay, while western blot was used to verify the expression of M2 macrophage polarization-related proteins. Tumor volume weight and M2 macrophage polarization were detected by tumor xenografts in nude mice. LINC01833 was highly expressed in NSCLC tumor tissues and cells. Knockdown of LINC01833 exosomes could significantly inhibit proliferation, migration, invasion of NSCLC cells, and M2 macrophage polarization of THP-1 cells, while simultaneous knockdown of miR-335-5p on the above basis could reverse the effect of knockdown of LINC01833. In vivo experiments also indicated that knockdown of LINC01833 exosomes suppressed tumor growth and M2 macrophage polarization. CAF-derived LINC01833 exosomes can promote the proliferation, migration and invasion of NSCLC cells and M2 macrophage polarization by inhibiting miR-335-5p and regulating VAPA activity.
Collapse
Affiliation(s)
- Jie Chen
- Naval Medical Center of PLA, Thoracic and Cardiac Surgery, Naval Medical University, Shanghai, China
| | - Jian-Jun Sun
- Naval Medical Center of PLA, Thoracic and Cardiac Surgery, Naval Medical University, Shanghai, China
| | - Ya-Wen Ma
- Department of Cardiology, Naval Medical Center of PLA, Naval Medical University, Shanghai, China
| | - Meng-Qin Zhu
- Naval Medical Center of PLA, Thoracic and Cardiac Surgery, Naval Medical University, Shanghai, China
| | - Jing Hu
- Naval Medical Center of PLA, Thoracic and Cardiac Surgery, Naval Medical University, Shanghai, China
| | - Qi-Jue Lu
- Naval Medical Center of PLA, Thoracic and Cardiac Surgery, Naval Medical University, Shanghai, China
| | - Zhi-Gang Cai
- Naval Medical Center of PLA, Thoracic and Cardiac Surgery, Naval Medical University, Shanghai, China
| |
Collapse
|
47
|
Zeng P, Shu LZ, Zhou YH, Huang HL, Wei SH, Liu WJ, Deng H. Stem Cell Division and Its Critical Role in Mammary Gland Development and Tumorigenesis: Current Progress and Remaining Challenges. Stem Cells Dev 2024; 33:449-467. [PMID: 38943275 DOI: 10.1089/scd.2024.0035] [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: 07/01/2024] Open
Abstract
The origin of breast cancer (BC) has traditionally been a focus of medical research. It is widely acknowledged that BC originates from immortal mammary stem cells and that these stem cells participate in two division modes: symmetric cell division (SCD) and asymmetrical cell division (ACD). Although both of these modes are key to the process of breast development and their imbalance is closely associated with the onset of BC, the molecular mechanisms underlying these phenomena deserve in-depth exploration. In this review, we first outline the molecular mechanisms governing ACD/SCD and analyze the role of ACD/SCD in various stages of breast development. We describe that the changes in telomerase activity, the role of polar proteins, and the stimulation of ovarian hormones subsequently lead to two distinct consequences: breast development or carcinogenesis. Finally, gene mutations, abnormalities in polar proteins, modulation of signal-transduction pathways, and alterations in the microenvironment disrupt the balance of BC stem cell division modes and cause BC. Important regulatory factors such as mammalian Inscuteable mInsc, Numb, Eya1, PKCα, PKCθ, p53, and IL-6 also play significant roles in regulating pathways of ACD/SCD and may constitute key targets for future research on stem cell division, breast development, and tumor therapy.
Collapse
MESH Headings
- Humans
- Female
- Breast Neoplasms/pathology
- Breast Neoplasms/metabolism
- Breast Neoplasms/genetics
- Animals
- Mammary Glands, Human/growth & development
- Mammary Glands, Human/pathology
- Mammary Glands, Human/cytology
- Mammary Glands, Human/metabolism
- Carcinogenesis/pathology
- Carcinogenesis/metabolism
- Carcinogenesis/genetics
- Stem Cells/metabolism
- Stem Cells/cytology
- Cell Division
- Neoplastic Stem Cells/metabolism
- Neoplastic Stem Cells/pathology
- Mammary Glands, Animal/growth & development
- Mammary Glands, Animal/cytology
- Mammary Glands, Animal/pathology
- Mammary Glands, Animal/metabolism
- Cell Transformation, Neoplastic/metabolism
- Cell Transformation, Neoplastic/genetics
- Cell Transformation, Neoplastic/pathology
- Signal Transduction
Collapse
Affiliation(s)
- Peng Zeng
- Department of Breast Surgery, Jiangxi Armed Police Corps Hospital, Nanchang, China
| | - Lin-Zhen Shu
- Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Yu-Hong Zhou
- Department of Breast Surgery, Jiangxi Armed Police Corps Hospital, Nanchang, China
| | - Hai-Lin Huang
- Department of Breast Surgery, Jiangxi Armed Police Corps Hospital, Nanchang, China
| | - Shu-Hua Wei
- Department of Breast Surgery, Jiangxi Armed Police Corps Hospital, Nanchang, China
| | - Wen-Jian Liu
- Department of Breast Surgery, Jiangxi Armed Police Corps Hospital, Nanchang, China
| | - Huan Deng
- Affiliated Rehabilitation Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- The Fourth Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Tumor Immunology Institute, Nanchang University, Nanchang, China
- The MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, Jiangxi Medical College, Nanchang University, Nanchang, China
| |
Collapse
|
48
|
Li J, Yao J. CD8 + T cell‑related KCTD5 contributes to malignant progression and unfavorable clinical outcome of patients with triple‑negative breast cancer. Mol Med Rep 2024; 30:166. [PMID: 39027992 PMCID: PMC11267436 DOI: 10.3892/mmr.2024.13290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 06/06/2024] [Indexed: 07/20/2024] Open
Abstract
Triple‑negative breast cancer (TNBC) is a highly aggressive and heterogeneous subtype of breast cancer that lacks expression of estrogen receptor, progesterone receptor, and HER2, making it more challenging to treat with targeted therapies. The present study aimed to identify CD8+ T cell‑associated genes, which could provide insight into the mechanisms underlying TNBC to facilitate developing novel immunotherapies. TNBC datasets were downloaded from public databases including The Cancer Genome Atlas, Molecular Taxonomy of Breast Cancer International Consortium, and Gene Expression Omnibus. Candidate genes were identified integrating weighted gene co‑expression network analysis (WGCNA), differential gene expression, protein‑protein‑interaction network construction and univariate Cox regression analysis. Kaplan‑Meier survival, multivariate Cox regression and receiver operating characteristic analysis were performed to evaluate the prognostic value of hub genes. Knockdown experiments, alongside wound healing, Cell Counting Kit‑8 and Transwell migration and invasion assays were performed. In total, seven gene modules were associated with CD8+ T cells using WGCNA, among which potassium channel tetramerization domain 5 (KCTD5) was significantly upregulated in TNBC samples and was associated with poor prognosis. KCTD5 expression inversely associated with infiltration ratios of 'Macrophages M1', 'Plasma cells', and 'γδ T cells', but positively with 'activated Mast cells', 'Macrophages M0', and 'Macrophages M2'. As an independent prognostic indicator for TNBC, KCTD5 was also associated with drug sensitivity and the expression of programmed cell death protein 1, Cytotoxic T‑Lymphocyte‑Associated Protein 4 (CTLA4), CD274), Cluster of Differentiation 86 (CD86), Lymphocyte‑Activation Gene 3 (LAG3), T Cell Immunoreceptor with Ig and ITIM Domains (TIGIT). Knockdown of KCTD5 significantly inhibited viability, migration and invasion of TNBC cells in vitro. KCTD5 was suggested to impact the tumor immune microenvironment by influencing the infiltration of immune cells and may serve as a potential therapeutic target for TNBC.
Collapse
Affiliation(s)
- Jia Li
- Department of Breast Surgical Oncology, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, Shanxi 030013, P.R. China
| | - Jingchun Yao
- Department of Head and Neck, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, Shanxi 030013, P.R. China
| |
Collapse
|
49
|
Zhang W, Wang M, Ji C, Liu X, Gu B, Dong T. Macrophage polarization in the tumor microenvironment: Emerging roles and therapeutic potentials. Biomed Pharmacother 2024; 177:116930. [PMID: 38878638 DOI: 10.1016/j.biopha.2024.116930] [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: 04/14/2024] [Revised: 05/31/2024] [Accepted: 06/09/2024] [Indexed: 07/28/2024] Open
Abstract
The tumor microenvironment (TME) is a combination of tumor cells and indigenous host stroma, which consists of tumor-infiltrating immune cells, endothelial cells, fibroblasts, pericytes, and non-cellular elements. Tumor-associated macrophages (TAMs) represent the major tumor-infiltrating immune cell type and are generally polarized into two functionally contradictory subtypes, namely classical activated M1 macrophages and alternatively activated M2 macrophages. Macrophage polarization refers to how macrophages are activated at a given time and space. The interplay between the TME and macrophage polarization can influence tumor initiation and progression, making TAM a potential target for cancer therapy. Here, we review the latest investigations on factors orchestrating macrophage polarization in the TME, how macrophage polarization affects tumor progression, and the perspectives in modulating macrophage polarization for cancer immunotherapy.
Collapse
Affiliation(s)
- Wenru Zhang
- Department of Natural Products Chemistry, Key Laboratory of Natural Products & Chemical Biology, Ministry of Education, School of Pharmaceutical Sciences, Shandong University, Jinan 250012, China
| | - Mengmeng Wang
- Department of Natural Products Chemistry, Key Laboratory of Natural Products & Chemical Biology, Ministry of Education, School of Pharmaceutical Sciences, Shandong University, Jinan 250012, China
| | - Chonghao Ji
- Department of Implantology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Xiaohui Liu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 2A Nanwei Road, Xicheng District, Beijing 100050, China
| | - Bowen Gu
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, United States.
| | - Ting Dong
- Department of Natural Products Chemistry, Key Laboratory of Natural Products & Chemical Biology, Ministry of Education, School of Pharmaceutical Sciences, Shandong University, Jinan 250012, China.
| |
Collapse
|
50
|
Zou Z, Luo T, Wang X, Wang B, Li Q. Exploring the interplay between triple-negative breast cancer stem cells and tumor microenvironment for effective therapeutic strategies. J Cell Physiol 2024; 239:e31278. [PMID: 38807378 DOI: 10.1002/jcp.31278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 03/28/2024] [Accepted: 04/05/2024] [Indexed: 05/30/2024]
Abstract
Triple-negative breast cancer (TNBC) is a highly aggressive and metastatic malignancy with poor treatment outcomes. The interaction between the tumor microenvironment (TME) and breast cancer stem cells (BCSCs) plays an important role in the development of TNBC. Owing to their ability of self-renewal and multidirectional differentiation, BCSCs maintain tumor growth, drive metastatic colonization, and facilitate the development of drug resistance. TME is the main factor regulating the phenotype and metastasis of BCSCs. Immune cells, cancer-related fibroblasts (CAFs), cytokines, mesenchymal cells, endothelial cells, and extracellular matrix within the TME form a complex communication network, exert highly selective pressure on the tumor, and provide a conducive environment for the formation of BCSC niches. Tumor growth and metastasis can be controlled by targeting the TME to eliminate BCSC niches or targeting BCSCs to modify the TME. These approaches may improve the treatment outcomes and possess great application potential in clinical settings. In this review, we summarized the relationship between BCSCs and the progression and drug resistance of TNBC, especially focusing on the interaction between BCSCs and TME. In addition, we discussed therapeutic strategies that target the TME to inhibit or eliminate BCSCs, providing valuable insights into the clinical treatment of TNBC.
Collapse
Affiliation(s)
- Zhuoling Zou
- Queen Mary College, Nanchang University, Nanchang, Jiangxi, China
| | - Tinglan Luo
- Department of Oncology, The Seventh People's Hospital of Chongqing (Affiliated Central Hospital of Chongqing University of Technology), Chongqing, China
| | - Xinyuan Wang
- Department of Clinical Medicine, The Second Clinical College of Chongqing Medicine University, Chongqing, China
| | - Bin Wang
- Department of Oncology, The Seventh People's Hospital of Chongqing (Affiliated Central Hospital of Chongqing University of Technology), Chongqing, China
| | - Qing Li
- Department of Oncology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|