1
|
Zhang X, Li P, Ji L, Zhang Y, Zhang Z, Guo Y, Zhang L, Jing S, Dong Z, Tian J, Yang L, Ding H, Yang E, Wang Z. A machine learning-based prognostic signature utilizing MSC proteomics for predicting bladder cancer prognosis and treatment response. Transl Oncol 2025; 54:102349. [PMID: 40073802 PMCID: PMC11950781 DOI: 10.1016/j.tranon.2025.102349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 01/23/2025] [Accepted: 03/03/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND Mesenchymal stem cells (MSCs), due to their tumor-targeting homing properties, are present in the tumor microenvironment (TME) and influence the biological behaviors of tumors. The purpose of this paper is to establish a signature based on the MSC secretome to predict the prognosis and treatment of bladder cancer (BLCA). METHODS The presence of MSCs in BLCA was validated through flow cytometry and multiplex fluorescence immunohistochemistry (mFIHC), and the relationships between MSCs and clinical characteristics were explored. Unsupervised clustering analysis was performed on BLCA according to the differential proteins detected in MSC-conditioned medium (MSCCM) using a cytokine array. Using the TCGA-BLCA, GSE32548, and GSE32894 datasets as background data, a risk signature was constructed according to the differential proteins in MSCCM through machine learning. For the risk groups with high and low prognoses, we calculated Kaplan-Meier (K-M) curves. Additionally, we explored the relationships between the signature and the tumor immune landscape, response to immunotherapy, and chemotherapy drugs. RESULTS Both flow cytometry and mFIHC confirmed the presence of MSCs in bladder tumors, and clinical samples revealed correlations between MSCs and the pathological grade, T stage, and Ki67 in BLCA. Based on differential proteins and unsupervised clustering analysis, BLCA patients were divided into two groups, and significant differences were found between these groups in terms of TME, immune response, and clinical treatments. Using machine learning, a signature was constructed with the combination algorithm Stepcox (both) + plsRcox, revealing significant survival differences between the high- and low-risk MSC groups. Regression analyses, along with ROC curves, further demonstrated that risk score independently predict the prognosis of patients with high predictive performance. Moreover, there were notable differences between the high- and low-risk groups in terms of the TME scores, immune infiltration, and immune checkpoints. For BLCA immunotherapy, the low-risk group suggested better efficacy, while conventional chemotherapy drugs such as gemcitabine and cisplatin might be less effective in the low-risk group. CONCLUSION The signature based on MSC secreted protein profiles could effectively predict the prognosis of BLCA and provided valuable guidance for treatment and drug resistance.
Collapse
Affiliation(s)
- Xinyu Zhang
- Institute of Urology, Lanzhou University Second Hospital, Key Laboratory of Gansu Province for Urological Diseases, Gansu Urological Clinical Center, Lanzhou, China
| | - Pan Li
- Institute of Urology, Lanzhou University Second Hospital, Key Laboratory of Gansu Province for Urological Diseases, Gansu Urological Clinical Center, Lanzhou, China
| | - Luhua Ji
- Institute of Urology, Lanzhou University Second Hospital, Key Laboratory of Gansu Province for Urological Diseases, Gansu Urological Clinical Center, Lanzhou, China
| | - Yuanfeng Zhang
- Institute of Urology, Lanzhou University Second Hospital, Key Laboratory of Gansu Province for Urological Diseases, Gansu Urological Clinical Center, Lanzhou, China
| | - Ze Zhang
- Institute of Urology, Lanzhou University Second Hospital, Key Laboratory of Gansu Province for Urological Diseases, Gansu Urological Clinical Center, Lanzhou, China
| | - Yufeng Guo
- Institute of Urology, Lanzhou University Second Hospital, Key Laboratory of Gansu Province for Urological Diseases, Gansu Urological Clinical Center, Lanzhou, China
| | - Luyang Zhang
- Institute of Urology, Lanzhou University Second Hospital, Key Laboratory of Gansu Province for Urological Diseases, Gansu Urological Clinical Center, Lanzhou, China
| | - Suoshi Jing
- Institute of Urology, Lanzhou University Second Hospital, Key Laboratory of Gansu Province for Urological Diseases, Gansu Urological Clinical Center, Lanzhou, China
| | - Zhilong Dong
- Institute of Urology, Lanzhou University Second Hospital, Key Laboratory of Gansu Province for Urological Diseases, Gansu Urological Clinical Center, Lanzhou, China
| | - Junqiang Tian
- Institute of Urology, Lanzhou University Second Hospital, Key Laboratory of Gansu Province for Urological Diseases, Gansu Urological Clinical Center, Lanzhou, China
| | - Li Yang
- Institute of Urology, Lanzhou University Second Hospital, Key Laboratory of Gansu Province for Urological Diseases, Gansu Urological Clinical Center, Lanzhou, China
| | - Hui Ding
- Institute of Urology, Lanzhou University Second Hospital, Key Laboratory of Gansu Province for Urological Diseases, Gansu Urological Clinical Center, Lanzhou, China
| | - Enguang Yang
- Institute of Urology, Lanzhou University Second Hospital, Key Laboratory of Gansu Province for Urological Diseases, Gansu Urological Clinical Center, Lanzhou, China.
| | - Zhiping Wang
- Institute of Urology, Lanzhou University Second Hospital, Key Laboratory of Gansu Province for Urological Diseases, Gansu Urological Clinical Center, Lanzhou, China.
| |
Collapse
|
2
|
Di Spirito A, Balkhi S, Vivona V, Mortara L. Key immune cells and their crosstalk in the tumor microenvironment of bladder cancer: insights for innovative therapies. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2025; 6:1002304. [PMID: 40177538 PMCID: PMC11964778 DOI: 10.37349/etat.2025.1002304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Accepted: 02/27/2025] [Indexed: 04/05/2025] Open
Abstract
Bladder cancer (BC) is a heterogeneous disease associated with high mortality if not diagnosed early. BC is classified into non-muscle-invasive BC (NMIBC) and muscle-invasive BC (MIBC), with MIBC linked to poor systemic therapy response and high recurrence rates. Current treatments include transurethral resection with Bacillus Calmette-Guérin (BCG) therapy for NMIBC and radical cystectomy with chemotherapy and/or immunotherapy for MIBC. The tumor microenvironment (TME) plays a critical role in cancer progression, metastasis, and therapeutic efficacy. A comprehensive understanding of the TME's complex interactions holds substantial translational significance for developing innovative treatments. The TME can contribute to therapeutic resistance, particularly in immune checkpoint inhibitor (ICI) therapies, where resistance arises from tumor-intrinsic changes or extrinsic TME factors. Recent advancements in immunotherapy highlight the importance of translational research to address these challenges. Strategies to overcome resistance focus on remodeling the TME to transform immunologically "cold" tumors, which lack immune cell infiltration, into "hot" tumors that respond better to immunotherapy. These strategies involve disrupting cancer-microenvironment interactions, inhibiting angiogenesis, and modulating immune components to enhance anti-tumor responses. Key mechanisms include cytokine involvement [e.g., interleukin-6 (IL-6)], phenotypic alterations in macrophages and natural killer (NK) cells, and the plasticity of cancer-associated fibroblasts (CAFs). Identifying potential therapeutic targets within the TME can improve outcomes for MIBC patients. This review emphasizes the TME's complexity and its impact on guiding novel therapeutic approaches, offering hope for better survival in MIBC.
Collapse
Affiliation(s)
- Anna Di Spirito
- Immunology and General Pathology Laboratory, Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy
| | - Sahar Balkhi
- Immunology and General Pathology Laboratory, Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy
| | - Veronica Vivona
- Immunology and General Pathology Laboratory, Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy
| | - Lorenzo Mortara
- Immunology and General Pathology Laboratory, Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy
| |
Collapse
|
3
|
Zhou S, Yang H. Radiotherapy modulates autophagy to reshape the tumor immune microenvironment to enhance anti-tumor immunity in esophageal cancer. Biochim Biophys Acta Rev Cancer 2025; 1880:189302. [PMID: 40120778 DOI: 10.1016/j.bbcan.2025.189302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 03/15/2025] [Accepted: 03/16/2025] [Indexed: 03/25/2025]
Abstract
The combination of radiotherapy and immunotherapy exerts synergistic antitumor in a range of human cancers, and also in esophageal cancer. Radiotherapy-induced tumor immune microenvironment (TIME) reprogramming is an essential basis for the synergistic antitumor between radiotherapy and immunotherapy. Radiotherapy can induce autophagy in tumor cells and immune cells of TIME, and autophagy activation is involved in the modification of immunological characteristics of TIME. The TIME landscape of esophageal cancer, especially ESCC, can be affected by radiotherapy or autophagy regulation. In this review, we depicted that local radiotherapy-induced autophagy could promote the maturation, migration, infiltration, and function of immune cells by complicated mechanisms to make TIME from immune "cold" to "hot", resulting in the synergistic antitumor of RT and IO. We argue that unraveling the relevance of radiotherapy-initiated autophagy to driving radiotherapy reprogramming TIME will open new ideas to explore new targets or more efficiently multimodal therapeutic interventions in ESCC.
Collapse
Affiliation(s)
- Suna Zhou
- Key Laboratory of Radiation Oncology of Taizhou, Department of Radiation Oncology, Key Laboratory of Minimally Invasive Techniques & Rapid Rehabilitation of Digestive System Tumor of Zhejiang Province, Taizhou Hospital Affiliated to Wenzhou Medical University, Taizhou, Zhejiang 317000, China
| | - Haihua Yang
- Key Laboratory of Radiation Oncology of Taizhou, Department of Radiation Oncology, Key Laboratory of Minimally Invasive Techniques & Rapid Rehabilitation of Digestive System Tumor of Zhejiang Province, Taizhou Hospital Affiliated to Wenzhou Medical University, Taizhou, Zhejiang 317000, China.
| |
Collapse
|
4
|
Yu F, Yu N, Zhang L, Xu X, Zhao Y, Cao Z, Wang F. Emodin Decreases Tumor-Associated Macrophages Accumulation and Suppresses Bladder Cancer Development by Inhibiting CXCL1 Secretion from Cancer-Associated Fibroblasts. Nutr Cancer 2025:1-16. [PMID: 40114381 DOI: 10.1080/01635581.2025.2480309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 03/07/2025] [Accepted: 03/10/2025] [Indexed: 03/22/2025]
Abstract
Tumor-associated macrophages (TAMs) and cancer-associated fibroblasts (CAFs) are the most abundant stromal cells in the bladder cancer (BC) microenvironment (TME). However, the detailed mechanisms underlying TAM-CAF communication and their contributions to BC progression remain incompletely understood. Emerging evidence shows that Emodin exerts anti-tumor effect on several tumor models by targeting TME. To date, the impact of Emodin on BC has not been previously reported. Our study firstly demonstrated that Emodin significantly inhibited tumor growth and reduced TAM accumulation in a murine BC model. Emodin markedly decreased serum levels of multiple chemokines in tumor-bearing mice, with CXCL1 showing the most pronounced reduction. Strikingly, Emodin selectively suppressed CXCL1 secretion in CAFs but not in TAMs or tumor cells. Furthermore, the decrease in TAM migration induced by Emodin was dependent on CAF-derived CXCL1. Using a subcutaneous tumor model, we found that Emodin failed to inhibit tumor growth when CXCL1-deficient CAFs were co-injected with tumor cells, underscoring the critical role of CXCL1 in this process. Bioinformatics analysis further revealed that elevated CXCL1 levels correlated negatively with invasive/metastatic potential and overall survival in BC patients. In conclusion, our findings establish that Emodin delays BC progression by disrupting CXCL1-mediated crosstalk between CAFs and TAMs.
Collapse
Affiliation(s)
- Fang Yu
- Department of Nutrition and Food Hygiene & Department of Health Education and Health Management, the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, Shaanxi Key Laboratory of Hazard Assessment and Protection in Environmental Health, Shaanxi Key Laboratory of Free Radical Biology and Medicine, Fourth Military Medical University, Xi'an, China
| | - Nan Yu
- Department of Ophthalmology, Heping Hospital affiliated with Changzhi Medical College, Changzhi, China
| | - Lei Zhang
- Department of Nutrition and Food Hygiene & Department of Health Education and Health Management, the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, Shaanxi Key Laboratory of Hazard Assessment and Protection in Environmental Health, Shaanxi Key Laboratory of Free Radical Biology and Medicine, Fourth Military Medical University, Xi'an, China
| | - Xiaona Xu
- Department of Nutrition and Food Hygiene & Department of Health Education and Health Management, the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, Shaanxi Key Laboratory of Hazard Assessment and Protection in Environmental Health, Shaanxi Key Laboratory of Free Radical Biology and Medicine, Fourth Military Medical University, Xi'an, China
| | - Yan Zhao
- Department of Basic Science, Fourth Military Medical University, Xi'an, China
| | - Zipeng Cao
- Department of Nutrition and Food Hygiene & Department of Health Education and Health Management, the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, Shaanxi Key Laboratory of Hazard Assessment and Protection in Environmental Health, Shaanxi Key Laboratory of Free Radical Biology and Medicine, Fourth Military Medical University, Xi'an, China
| | - Feng Wang
- Department of Nutrition and Food Hygiene & Department of Health Education and Health Management, the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, Shaanxi Key Laboratory of Hazard Assessment and Protection in Environmental Health, Shaanxi Key Laboratory of Free Radical Biology and Medicine, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
5
|
Wang H, Sun P, Yuan X, Xu Z, Jiang X, Xiao M, Yao X, Shi Y. Autophagy in tumor immune escape and immunotherapy. Mol Cancer 2025; 24:85. [PMID: 40102867 PMCID: PMC11921617 DOI: 10.1186/s12943-025-02277-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 02/24/2025] [Indexed: 03/20/2025] Open
Abstract
The immunotherapy targeting tumor immune escape mechanisms has become a critical strategy in anticancer treatment; however, the challenge of immune resistance remains significant. Autophagy, a cellular response to various stressors, involves the degradation of damaged proteins and organelles via lysosomal pathways, maintaining cellular homeostasis. This process not only supports tumor cell survival but also profoundly impacts the efficacy of cancer immunotherapies. The modulation of autophagy in tumor cells or immune cells exerts dual effects on tumor immune escape and immunotherapy. However, the mechanistic details of how autophagy influences the immune system and therapy remain inadequately understood. Given this complexity, a deeper understanding of the role of autophagy in the tumor-immune landscape could reveal novel therapeutic avenues. By manipulating autophagy appropriately, it may be possible to overcome immune resistance and enhance the effectiveness of immunotherapeutic strategies. This article summarizes the role of autophagy in tumor immunity, its relationship with immunotherapy, and the potential therapeutic benefits of targeting autophagy to strengthen antitumor immune responses and optimize the outcomes of immunotherapy.
Collapse
Affiliation(s)
- Huan Wang
- Department of Respiratory & Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Peng Sun
- Department of Respiratory & Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Xijing Yuan
- Department of Respiratory & Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Zhiyong Xu
- Department of Respiratory and Critical Care Medicine, Center for Oncology Medicine, the, Fourth Affiliated Hospital of School of Medicine, and Internation School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Xinyuan Jiang
- Department of Respiratory and Critical Care Medicine, Center for Oncology Medicine, the, Fourth Affiliated Hospital of School of Medicine, and Internation School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Mingshu Xiao
- Department of Respiratory and Critical Care Medicine, Center for Oncology Medicine, the, Fourth Affiliated Hospital of School of Medicine, and Internation School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Xin Yao
- Department of Respiratory & Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China.
| | - Yueli Shi
- Department of Respiratory and Critical Care Medicine, Center for Oncology Medicine, the, Fourth Affiliated Hospital of School of Medicine, and Internation School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China.
| |
Collapse
|
6
|
Li D, Wang J, Li X, Wang Z, Yu Q, Koh SB, Wu R, Ye L, Guo Y, Okoli U, Pati-Alam A, Mota E, Wei W, Yoo KH, Cho WC, Feng D, Heavey S. Interactions between radiotherapy resistance mechanisms and the tumor microenvironment. Crit Rev Oncol Hematol 2025; 210:104705. [PMID: 40107436 DOI: 10.1016/j.critrevonc.2025.104705] [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: 01/31/2025] [Revised: 03/11/2025] [Accepted: 03/12/2025] [Indexed: 03/22/2025] Open
Abstract
BACKGROUND Resistance to radiotherapy (RT) presents a significant clinical challenge in management of cancer. Recent evidence points to specific mechanisms of resistance within the tumor microenvironment (TME), which we aim to discuss, with the aim of overcoming the clinical challenge. METHODS We performed the narrative review using PubMed and Web of Science databases to identify studies that reported the regulative network and treatments of RT resistance from TME perspectives. RESULTS RT significantly changes the immune TME of cancers, which is closely appearing to play a key role in RT resistance (RTR) by modulating immune cell infiltration and function. Various phenotypes are involved in the development of RTR, such as autophagy, senescence, oxidative stress, cell polarization, ceramide metabolism, and angiogenesis in the TME. Key genes and pathways are also implicated in RTR, including immune and inflammatory cytokines, TGF-β, P53, the NF-κB pathway, the cGAS/STING pathway, the ERK and AKT pathway, and the STAT pathway. Based on the mechanism of RTR in the TME, many proposed routes to overcome RTR, several specifically target the TME including targeting fibroblast activation protein, exosomes management, nanomedicine, and immunotherapy. Many challenges in RT resistance still need to be further explored with emerging investigative methods, such as artificial intelligence, genetic technologies, and bioengineering. CONCLUSIONS The complex interactions between RT and TME significantly affect the efficiency of RT. Novel approaches to overcome this clinical difficulty are promising, which needs future work to further explore and identify better treatment strategies.
Collapse
Affiliation(s)
- Dengxiong Li
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jie Wang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xinrui Li
- Department of Rehabilitation, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Zhipeng Wang
- Department of Urology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Qingxin Yu
- Department of Pathology, Ningbo Clinical Pathology Diagnosis Center, Ningbo, Zhejiang 315211, China
| | - Siang Boon Koh
- Faculty of Health and Life Sciences, University of Bristol, Bristol, BS8 1TD, UK
| | - Ruicheng Wu
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Luxia Ye
- Department of Public Research Platform, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
| | - Yiqing Guo
- Department of Public Research Platform, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
| | - Uzoamaka Okoli
- Division of Surgery & Interventional Science, University College London, London, UK; Basic and Translational Cancer Research Group, Department of Pharmacology and Therapeutics, College of Medicine, University of Nigeria, Eastern part of Nigeria, Nsukka, Enugu, Nigeria
| | - Alisha Pati-Alam
- Division of Surgery & Interventional Science, University College London, London, UK
| | - Eduardo Mota
- Division of Surgery & Interventional Science, University College London, London, UK
| | - Wuran Wei
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Koo Han Yoo
- Department of Urology, Kyung Hee University, South Korea
| | - William C Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, Hong Kong Special Administrative Region of China.
| | - Dechao Feng
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, China; Division of Surgery & Interventional Science, University College London, London, UK.
| | - Susan Heavey
- Division of Surgery & Interventional Science, University College London, London, UK.
| |
Collapse
|
7
|
Chen H, Liu J, Cao Z, Li J, Zhang H, Yang Q, Cheng J, Shen Y, He K. Enhancing hepatocellular carcinoma therapy with DOX-loaded SiO 2 nanoparticles via mTOR-TFEB pathway autophagic flux inhibition. J Nanobiotechnology 2025; 23:27. [PMID: 39828690 PMCID: PMC11743218 DOI: 10.1186/s12951-025-03107-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 01/11/2025] [Indexed: 01/22/2025] Open
Abstract
Chemotherapeutic drugs often fail to provide long-term efficacy due to their lack of specificity and high toxicity. To enhance the biosafety and reduce the side effects of these drugs, various nanocarrier delivery systems have been developed. In this study, we loaded the anticancer drug doxorubicin (DOX) and an MRI contrast agent into silica nanoparticles, coating them with pH-responsive and tumor cell-targeting polymers. These polymers enable the carrier to achieve targeted delivery and controlled drug release in acidic environments. This integrated diagnostic and therapeutic strategy successfully achieved both the diagnosis and treatment of liver cancer. Additionally, we demonstrated that the nanocarrier inhibits autophagic flux in liver cancer cells by targeting the autophagy-lysosome pathway and regulating the nuclear translocation of TFEB, thereby promoting tumor cell death. This novel diagnostic-integrated nanocarrier is expected to be a promising tool for targeted liver cancer treatment.
Collapse
Affiliation(s)
- Huanyu Chen
- Imaging Center, The Third Affiliated Hospital of Anhui Medical University, Hefei, 230061, China
- The Fifth Clinical Medical School of Anhui Medical University, Hefei, 230032, China
| | - Jun Liu
- School of Basic Medical Sciences and Biopharmaceutical Research Institute, Anhui Medical University, Hefei, 230032, China
| | - Zhichao Cao
- Imaging Center, The Third Affiliated Hospital of Anhui Medical University, Hefei, 230061, China
- Wannan Medical College, Wuhu, 241002, China
| | - Jiajia Li
- Central Laboratory, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Hong Zhang
- Imaging Center, The Third Affiliated Hospital of Anhui Medical University, Hefei, 230061, China
- Wannan Medical College, Wuhu, 241002, China
| | - Qianqian Yang
- Imaging Center, The Third Affiliated Hospital of Anhui Medical University, Hefei, 230061, China
- The Fifth Clinical Medical School of Anhui Medical University, Hefei, 230032, China
| | - Jian Cheng
- Suzhou Institute for Advanced Research, University of Science and Technology of China, Suzhou, 215123, China.
| | - Yuxian Shen
- School of Basic Medical Sciences and Biopharmaceutical Research Institute, Anhui Medical University, Hefei, 230032, China.
| | - Kewu He
- Imaging Center, The Third Affiliated Hospital of Anhui Medical University, Hefei, 230061, China.
- The Fifth Clinical Medical School of Anhui Medical University, Hefei, 230032, China.
| |
Collapse
|
8
|
Fusco C, Di Rella F, Liotti A, Colamatteo A, Ferrara AL, Gigantino V, Collina F, Esposito E, Donzelli I, Porcellini A, Feola A, Micillo T, Perna F, Garziano F, Maniscalco GT, Varricchi G, Mottola M, Zuccarelli B, De Simone B, di Bonito M, Matarese G, Accurso A, Pontillo M, Russo D, Insabato L, Spaziano A, Cantone I, Pezone A, De Rosa V. CD4 +FOXP3Exon2 + regulatory T cell frequency predicts breast cancer prognosis and survival. SCIENCE ADVANCES 2025; 11:eadr7934. [PMID: 39813341 PMCID: PMC11734725 DOI: 10.1126/sciadv.adr7934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 12/13/2024] [Indexed: 01/18/2025]
Abstract
CD4+FOXP3+ regulatory T cells (Tregs) suppress immune responses to tumors, and their accumulation in the tumor microenvironment (TME) correlates with poor clinical outcome in several cancers, including breast cancer (BC). However, the properties of intratumoral Tregs remain largely unknown. Here, we found that a functionally distinct subpopulation of Tregs, expressing the FOXP3 Exon2 splicing variants, is prominent in patients with hormone receptor-positive BC with poor prognosis. Notably, a comprehensive examination of the TCGA validated FOXP3E2 as an independent prognostic marker in all other BC subtypes. We found that FOXP3E2 expression underlies BCs with defective mismatch repair and a stem-like signature and highlights pathways involved in tumor survival. Last, we found that the TME induces FOXP3E2 through the CXCL12/CXCR4 axis and confirmed the higher immunosuppressive capacity of FOXP3E2+ Tregs derived from patients with BC. Our study suggests that FOXP3E2+ Tregs might be used as an independent biomarker to predict BC prognosis and survival and to develop super-targeted immunotherapies.
Collapse
Affiliation(s)
- Clorinda Fusco
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli “Federico II”, Napoli, Italy
| | - Francesca Di Rella
- Oncologia Clinica Sperimentale di Senologia, Istituto Nazionale Tumori, IRCCS, Fondazione Pascale, Napoli, Italy
| | - Antonietta Liotti
- Istituto per l’Endocrinologia e l’Oncologia Sperimentale “G. Salvatore”, IEOS-CNR, Napoli, Italy
- Dipartimento di Scienze Mediche Traslazionali, Università degli Studi di Napoli “Federico II”, Napoli 80131, Italy
| | - Alessandra Colamatteo
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli “Federico II”, Napoli, Italy
| | - Anne Lise Ferrara
- Dipartimento di Scienze Mediche Traslazionali, Università degli Studi di Napoli “Federico II”, Napoli 80131, Italy
| | - Vincenzo Gigantino
- Unità di Anatomia Patologica, Istituto Nazionale Tumori, IRCCS, Fondazione Pascale, Napoli, Italy
| | - Francesca Collina
- Unità di Anatomia Patologica, Istituto Nazionale Tumori, IRCCS, Fondazione Pascale, Napoli, Italy
| | - Emanuela Esposito
- Chirurgia Oncologica di Senologia, Istituto Nazionale Tumori, IRCCS, Fondazione Pascale, Napoli, Italy
| | - Ivana Donzelli
- Chirurgia Oncologica di Senologia, Istituto Nazionale Tumori, IRCCS, Fondazione Pascale, Napoli, Italy
| | - Antonio Porcellini
- Dipartimento di Biologia, Complesso Universitario di Monte Sant’Angelo, Università di Napoli “Federico II”, Napoli 80126, Italy
| | - Antonia Feola
- Dipartimento di Biologia, Complesso Universitario di Monte Sant’Angelo, Università di Napoli “Federico II”, Napoli 80126, Italy
| | - Teresa Micillo
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli “Federico II”, Napoli, Italy
| | - Francesco Perna
- Dipartimento di Medicina Clinica e Chirurgia, Università degli Studi di Napoli “Federico II”, Napoli 80131, Italy
| | - Federica Garziano
- U.O.C Biochimica Clinica Azienda Ospedaliera Specialistica dei Colli Monaldi-Cotugno-C.T.O. Presidio Monaldi, Napoli, Italy
| | - Giorgia Teresa Maniscalco
- Clinica Neurologica e Unità Stroke, Centro Sclerosi Multipla, Ospedale “A.Cardarelli”, Napoli 80131, Italy
| | - Gilda Varricchi
- Istituto per l’Endocrinologia e l’Oncologia Sperimentale “G. Salvatore”, IEOS-CNR, Napoli, Italy
- Dipartimento di Scienze Mediche Traslazionali, Università degli Studi di Napoli “Federico II”, Napoli 80131, Italy
| | - Maria Mottola
- U.O.C Medicina Trasfusionale, Azienda Ospedaliera Specialistica dei Colli, Napoli 80131, Italy
| | - Bruno Zuccarelli
- U.O.C Medicina Trasfusionale, Azienda Ospedaliera Specialistica dei Colli, Napoli 80131, Italy
| | - Bruna De Simone
- Dipartimento di Sanità Pubblica, Università degli Studi di Napoli “Federico II”, Napoli 80131, Italy
| | - Maurizio di Bonito
- Unità di Anatomia Patologica, Istituto Nazionale Tumori, IRCCS, Fondazione Pascale, Napoli, Italy
| | - Giuseppe Matarese
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli “Federico II”, Napoli, Italy
- Istituto per l’Endocrinologia e l’Oncologia Sperimentale “G. Salvatore”, IEOS-CNR, Napoli, Italy
| | - Antonello Accurso
- Dipartimento di Chirurgia Generale, Oncologica, Bariatrica e Metabolica, Università degli Studi di Napoli “Federico II”, Napoli 80131, Italy
| | - Martina Pontillo
- Dipartimento di Chirurgia Generale, Oncologica, Bariatrica e Metabolica, Università degli Studi di Napoli “Federico II”, Napoli 80131, Italy
| | - Daniela Russo
- Unità di Anatomia Patologica, Dipartimento di Scienze Biomediche Avanzate, Facoltà di Medicina, Università degli Studi di Napoli “Federico II”, Napoli 80131, Italy
| | - Luigi Insabato
- Unità di Anatomia Patologica, Dipartimento di Scienze Biomediche Avanzate, Facoltà di Medicina, Università degli Studi di Napoli “Federico II”, Napoli 80131, Italy
| | - Alessandra Spaziano
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli “Federico II”, Napoli, Italy
| | - Irene Cantone
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli “Federico II”, Napoli, Italy
- Istituto per l’Endocrinologia e l’Oncologia Sperimentale “G. Salvatore”, IEOS-CNR, Napoli, Italy
| | - Antonio Pezone
- Dipartimento di Biologia, Complesso Universitario di Monte Sant’Angelo, Università di Napoli “Federico II”, Napoli 80126, Italy
| | - Veronica De Rosa
- Istituto per l’Endocrinologia e l’Oncologia Sperimentale “G. Salvatore”, IEOS-CNR, Napoli, Italy
| |
Collapse
|
9
|
Zeng X, Chen Y, Wang J, He M, Qiu J, Huang Y. Targeting autophagy to enhance chemotherapy and immunotherapy in oral cancer. Front Immunol 2025; 15:1535649. [PMID: 39840028 PMCID: PMC11747659 DOI: 10.3389/fimmu.2024.1535649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 12/17/2024] [Indexed: 01/23/2025] Open
Abstract
Oral cancer is a highly malignant disease characterized by recurrence, metastasis, and poor prognosis. Autophagy, a catabolic process induced under stress conditions, has been shown to play a dual role in oral cancer development and therapy. Recent studies have identified that autophagy activation in oral epithelial cells suppresses cancer cell survival by inhibiting key pathways such as the mammalian target of rapamycin (mTOR) and mitogen-activated protein kinase (MAPK), while activating the adenosine monophosphate-activated protein kinase (AMPK) pathway. Inducing autophagy promotes degradation of eukaryotic initiation factor 4E, thus reducing metastasis and enhancing the efficacy of chemotherapy, radiotherapy, and immunotherapy. Furthermore, autophagy induction can modulate the tumor immune microenvironment and enhance antitumor immunity. This review comprehensively summarizes the relationship between autophagy and oral cancer, focusing on its mechanisms and therapeutic potential when combined with conventional treatments. While promising, the precise mechanisms and clinical applications of autophagy inducers in oral cancer therapy remain to be elucidated, offering new directions for future research to improve treatment outcomes and reduce recurrence.
Collapse
Affiliation(s)
- Xiaoli Zeng
- Department of Oncology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
- Jiangxi "Flagship" Oncology Department of Synergy for Chinese and Western Medicine, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
- Department of Oncology, Jiangxi Clinical Medical Research Center for Cancer, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Yue Chen
- Department of Oncology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
- Department of Oncology, Jiangxi Clinical Medical Research Center for Cancer, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Jing Wang
- Department of Oncology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
- Department of Oncology, Jiangxi Clinical Medical Research Center for Cancer, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Miao He
- Department of Oncology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
- Department of Oncology, Jiangxi Clinical Medical Research Center for Cancer, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Junyao Qiu
- Department of Gastroenterology, The Affiliated Ganzhou Hospital of Nanchang University, Ganzhou, Jiangxi, China
| | - Yun Huang
- Department of Otolaryngology, The Affiliated Ganzhou Hospital of Nanchang University, Ganzhou, Jiangxi, China
| |
Collapse
|
10
|
Lu C, Zhang Y, Sun C, Na Y, Sun H, Ma J, Wang X, Cang X. Stromal Cell Derived Factor-1 Promotes Hepatic Insulin Resistance via Inhibiting Hepatocyte Lipophagy. J Cell Mol Med 2025; 29:e70352. [PMID: 39855896 PMCID: PMC11761003 DOI: 10.1111/jcmm.70352] [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: 06/06/2024] [Revised: 12/26/2024] [Accepted: 01/02/2025] [Indexed: 01/27/2025] Open
Abstract
Saturated fatty acid (SFA) accumulation in liver decreases hepatocyte lipophagy, a type of selective autophagy that degrades intracellular lipid droplets, leading to hepatic insulin resistance (IR), which contributes to simultaneous increases in liver glucose production and fat synthesis, resulting in hyperglycemia and dyslipidemia traits of type 2 diabetes mellitus (T2DM). Stromal cell derived factor-1 (SDF-1), a cytokine produced by hepatocytes, inhibits autophagy. In this study, we evaluated the hypothesis that SDF-1 promoted hepatic IR via inhibiting hepatocyte lipophagy during T2DM. Furthermore, we probed the downstream pathway participating in the role of SDF-1. The results showed that the neutralising of SDF-1 improved hepatic IR via promoting hepatocyte lipophagy in a mouse high-fat and high sucrose diet (HFHSD)-induced T2DM model. In vitro, SDF-1 expression and release increased in palmitic acid (PA, a kind of SFA)-treated hepatocytes. Meanwhile, SDF-1 bound to up-regulated C-X-C chemokine receptor type 4 (CXCR4) and C-X-C chemokine receptor type 7 (CXCR7) on PA-treated hepatocytes. Subsequently, SDF-1 inhibited lipophagy in PA-treated hepatocytes via CXCR4, rather than CXCR7. Finally, SDF-1/CXCR4/protein kinase B (AKT)/mechanistic target of rapamycin (mTOR) pathway-inhibited lipophagy promotes PA-induced hepatocyte IR. Collectively, this study discovered that SDF-1 might inhibit lipophagy in SFA-treated hepatocytes to promote hepatic IR via CXCR4/AKT/mTOR pathway.
Collapse
Affiliation(s)
- Chunfeng Lu
- Department of EndocrinologySecondary Affiliated Hospital of Nantong University and the First People's Hospital of NantongNantongJiangsuChina
| | - Yuting Zhang
- Department of PathologyLixiang Eye Hospital of Soochow UniversitySuzhouJiangsuChina
| | - Cuilian Sun
- Department of Pathogen Biology, Medical CollegeNantong UniversityNantongJiangsuChina
| | - Yuhang Na
- Department of Pathogen Biology, Medical CollegeNantong UniversityNantongJiangsuChina
| | - Haotian Sun
- Department of PathologyLixiang Eye Hospital of Soochow UniversitySuzhouJiangsuChina
| | - Jianhua Ma
- Department of Endocrinology, Nanjing First HospitalNanjing Medical UniversityNanjingJiangsuChina
| | - Xueqin Wang
- Department of EndocrinologySecondary Affiliated Hospital of Nantong University and the First People's Hospital of NantongNantongJiangsuChina
| | - Xiaomin Cang
- Department of EndocrinologySecondary Affiliated Hospital of Nantong University and the First People's Hospital of NantongNantongJiangsuChina
| |
Collapse
|
11
|
Zhan Y, Weng M, Guo Y, Lv D, Zhao F, Yan Z, Jiang J, Xiao Y, Yao L. Identification and validation of the nicotine metabolism-related signature of bladder cancer by bioinformatics and machine learning. Front Immunol 2024; 15:1465638. [PMID: 39742262 PMCID: PMC11685211 DOI: 10.3389/fimmu.2024.1465638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 11/29/2024] [Indexed: 01/03/2025] Open
Abstract
Background Several studies indicate that smoking is one of the major risk factors for bladder cancer. Nicotine and its metabolites, the main components of tobacco, have been found to be strongly linked to the occurrence and progression of bladder cancer. However, the function of nicotine metabolism-related genes (NRGs) in bladder urothelial carcinoma (BLCA) are still unclear. Methods NRGs were collected from MSigDB to identify the clusters associated with nicotine metabolism. Prognostic differentially expressed genes (DEGs) were filtered via differentially expression analysis and univariate Cox regression analysis. Integrative machine learning combination based on 10 machine learning algorithms was used for the construction of robust signature. Subsequently, the clinical application of signature in terms of prognosis, tumor microenvironment (TME) as well as immunotherapy was comprehensively evaluated. Finally, the biology function of the signature gene was further verified via CCK-8, transwell migration and colony formation. Results Three clusters associated with nicotine metabolism were discovered with distinct prognosis and immunological patterns. A four gene-signature was developed by random survival forest (RSF) method with highest average Harrell's concordance index (C-index) of 0.763. The signature exhibited a reliable and accurate performance in prognostic prediction across TCGA-train, TCGA-test and GSE32894 cohorts. Furthermore, the signature showed highly correlation with clinical characteristics, TME and immunotherapy responses. Suppression of MKRN1 was found to reduce the migration and proliferation of bladder cancer cell. In addition, enhanced migration and proliferation caused by nicotine was blocked down by loss of MKRN1. Conclusions The novel nicotine metabolism-related signature may provide valuable insights into clinical prognosis and potential benefits of immunotherapy in bladder cancer patients.
Collapse
Affiliation(s)
- Yating Zhan
- Department of Blood Transfusion, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Min Weng
- Department of Urology, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Yangyang Guo
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Dingfeng Lv
- Department of Blood Transfusion, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Feng Zhao
- Department of Blood Transfusion, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Zejun Yan
- Department of Urology, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Junhui Jiang
- Department of Urology, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Yanyi Xiao
- Department of Thyroid and Breast Surgery, The Second Affiliated Hospital of Shanghai University, Wenzhou, China
| | - Lili Yao
- Department of Ultrasonography, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
12
|
Song D, Wu Y, Li J, Liu J, Yi Z, Wang X, Sun J, Li L, Wu Q, Chen Y, Fang H, Luan T, Du H, Huang J, Peng W, Wei Y, Li F, Li Q, Zhang L, Zhu Y, Wan J, Ren G, Li H. Insulin-like growth factor 2 drives fibroblast-mediated tumor immunoevasion and confers resistance to immunotherapy. J Clin Invest 2024; 134:e183366. [PMID: 39545420 PMCID: PMC11563680 DOI: 10.1172/jci183366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 09/20/2024] [Indexed: 11/17/2024] Open
Abstract
T cell exclusion is crucial in enabling tumor immune evasion and immunotherapy resistance. However, the key genes driving this process remain unclear. We uncovered a notable increase of insulin-like growth factor 2 (IGF2) in immune-excluded tumors, predominantly secreted by cancer-associated fibroblasts (CAFs). Using mice with systemic or fibroblast-specific deletion of IGF2, we demonstrated that IGF2 deficiency enhanced the infiltration and cytotoxic activity of CD8+ T cells, leading to a reduction in tumor burden. Integration of spatial and single-cell transcriptomics revealed that IGF2 promoted interaction between CAFs and T cells via CXCL12 and programmed death ligand 1 (PD-L1). Mechanistically, autocrine IGF2 activated PI3K/AKT signaling by binding to the IGF1 receptor (IGF1R) on CAFs, which was required for the immunosuppressive functions of CAFs. Furthermore, genetic ablation of IGF2 or targeted inhibition of the IGF2/IGF1R axis with the inhibitor linsitinib markedly boosted the response to immune checkpoint blockade. Clinically, elevated levels of IGF2 in tumors or plasma correlated with an adverse prognosis and reduced efficacy of anti-programmed death 1 treatment. Together, these results highlight the pivotal role of IGF2 in promoting CAF-mediated immunoevasion, indicating its potential as a biomarker and therapeutic target in immunotherapy.
Collapse
Affiliation(s)
- Daqiang Song
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Pharmacology, Chongqing Medical University, Chongqing, China
| | - Yushen Wu
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Oncology
| | - Jie Li
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jiazhou Liu
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Breast and Thyroid Surgery, and
| | - Ziying Yi
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaoyu Wang
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jiazheng Sun
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Liuying Li
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qianxue Wu
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yuru Chen
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Huiying Fang
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Tiankuo Luan
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | | | - Jing Huang
- Department of Respiratory, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Weiyan Peng
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yuxian Wei
- Department of Breast and Thyroid Surgery, and
| | - Fan Li
- Department of Breast and Thyroid Surgery, and
| | - Qin Li
- Department of Oncology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Li Zhang
- Department of Pathophysiology and
| | - Yong Zhu
- Research Institute of Life Sciences, Chongqing Medical University, Chongqing, China
| | - Jingyuan Wan
- Department of Pharmacology, Chongqing Medical University, Chongqing, China
| | - Guosheng Ren
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Breast and Thyroid Surgery, and
| | - Hongzhong Li
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
13
|
Xie Q, Liu X, Liu R, Pan J, Liang J. Cellular mechanisms of combining innate immunity activation with PD-1/PD-L1 blockade in treatment of colorectal cancer. Mol Cancer 2024; 23:252. [PMID: 39529058 PMCID: PMC11555832 DOI: 10.1186/s12943-024-02166-w] [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: 10/03/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024] Open
Abstract
PD-1/PD-L1 blockade therapies have displayed extraordinary clinical efficacy for melanoma, renal, bladder and lung cancer; however, only a minority of colorectal cancer (CRC) patients benefit from these treatments. The efficacy of PD-1/PD-L1 blockade in CRC is limited by the complexities of tumor microenvironment. PD-1/PD-L1 blockade immunotherapy is based on T cell-centered view of tumor immunity. However, the onset and maintenance of T cell responses and the development of long-lasting memory T cells depend on innate immune responses. Acknowledging the pivotal role of innate immunity in anti-tumor immune response, this review encapsulates the employment of combinational therapies those involve PD-1/PD-L1 blockade alongside the activation of innate immunity and explores the underlying cellular mechanisms, aiming to harnessing innate immune responses to induce long-lasting tumor control for CRC patients who received PD-1/PD-L1 blockade therapy.
Collapse
Affiliation(s)
- Qi Xie
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Lung Cancer Institute, Jinan, 250014, China
| | - Xiaolin Liu
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Lung Cancer Institute, Jinan, 250014, China
| | - Rengyun Liu
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Jingxuan Pan
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060, China.
| | - Jing Liang
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Lung Cancer Institute, Jinan, 250014, China.
| |
Collapse
|
14
|
Li X, Sun H, Zhang L, Liang H, Zhang B, Yang J, Peng X, Sun J, Zhou Y, Zhai M, Jiang L, Zhu H, Duan W. GDF15 attenuates sepsis-induced myocardial dysfunction by inhibiting cardiomyocytes ferroptosis via the SOCS1/GPX4 signaling pathway. Eur J Pharmacol 2024; 982:176894. [PMID: 39147013 DOI: 10.1016/j.ejphar.2024.176894] [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: 03/11/2024] [Revised: 08/11/2024] [Accepted: 08/12/2024] [Indexed: 08/17/2024]
Abstract
Sepsis is a systemic inflammatory response syndrome triggered by infection, presenting with symptoms such as fever, increased heart rate, and low blood pressure. In severe cases, it can lead to multiple organ dysfunction, posing a life-threatening risk. Sepsis-induced cardiomyopathy (SIC) is a critical factor in the poor prognosis of septic patients, leading to myocardial dysfunction characterized by cell death, inflammation, and diminished cardiac function. Ferroptosis, an iron-dependent form of programmed cell death, is a key mechanism causing cardiomyocyte damage in SIC. Growth differentiation factor 15 (GDF15), a member of the TGF-β superfamily, is associated with various cardiovascular diseases and can inhibit oxidative stress, reduce reactive oxygen species (ROS), and suppress ferroptosis. Elevated serum GDF15 levels in sepsis are correlated with organ injuries, suggesting its potential as a therapeutic target. However, its role and mechanisms in SIC remain unclear. Glutathione peroxidase 4 (GPX4), the only enzyme capable of reducing lipid peroxides within cells, protects cells by reducing lipid peroxidation levels and inhibiting ferroptosis. Investigating the regulatory factors of GPX4 may provide a theoretical basis for SIC treatment. In this study, a mouse SIC model revealed that elevated GDF15 exerts a protective effect. Antagonizing GDF15 exacerbates myocardial damage. Through transcriptomic analysis and other methods, we confirmed that GDF15 inhibits the expression of SOCS1 by activating the ALK5-SMAD2/3 pathway, thereby activates the JAK2/STAT3 pathway, promotes the transcription of GPX4, inhibits ferroptosis in cardiomyocytes, and plays a myocardial protective role in SIC.
Collapse
Affiliation(s)
- Xiayun Li
- College of Life Sciences, Northwest University, Xi'an, 710069, China; Department of Cardiovascular Surgery, Xijing Hospital, The Air Force Medical University, Xi'an, 710032, China
| | - He Sun
- Department of Cardiovascular Surgery, Xijing Hospital, The Air Force Medical University, Xi'an, 710032, China
| | - Liyun Zhang
- Department of Cardiovascular Surgery, Xijing Hospital, The Air Force Medical University, Xi'an, 710032, China
| | - Hongliang Liang
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, 94305, USA; Stanford Cardiovascular Institute, Stanford University, Stanford, CA, 94305, USA
| | - Bin Zhang
- Department of Cardiovascular Surgery, Xijing Hospital, The Air Force Medical University, Xi'an, 710032, China; Department of Surgery, The 954th Hospital of the Chinese People's Liberation Army, Shannan, 856100, China
| | - Jiachang Yang
- Department of Cardiovascular Surgery, Xijing Hospital, The Air Force Medical University, Xi'an, 710032, China
| | - Xiangyan Peng
- School of Medicine, Northwest University, Xi'an, 710069, China
| | - Jingwei Sun
- Department of Cardiovascular Surgery, Xijing Hospital, The Air Force Medical University, Xi'an, 710032, China
| | - Yang Zhou
- College of Life Sciences, Northwest University, Xi'an, 710069, China
| | - Mengen Zhai
- Department of Cardiovascular Surgery, Xijing Hospital, The Air Force Medical University, Xi'an, 710032, China
| | - Liqing Jiang
- Department of Cardiovascular Surgery, Xijing Hospital, The Air Force Medical University, Xi'an, 710032, China.
| | - Hanzhao Zhu
- Department of Cardiovascular Surgery, Xijing Hospital, The Air Force Medical University, Xi'an, 710032, China.
| | - Weixun Duan
- Department of Cardiovascular Surgery, Xijing Hospital, The Air Force Medical University, Xi'an, 710032, China.
| |
Collapse
|
15
|
Wang T, Tian L, Wei B, Li J, Zhang C, Long R, Zhu X, Zhang Y, Wang B, Tang G, Yang J, Guo Y. Effect of fibroblast heterogeneity on prognosis and drug resistance in high-grade serous ovarian cancer. Sci Rep 2024; 14:26617. [PMID: 39496775 PMCID: PMC11535537 DOI: 10.1038/s41598-024-77630-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 10/23/2024] [Indexed: 11/06/2024] Open
Abstract
Tumor heterogeneity is associated with poor prognosis and drug resistance, leading to therapeutic failure. Here, we used tumor evolution analysis to determine the intra- and intertumoral heterogeneity of high-grade serous ovarian cancer (HGSOC) and analyze the correlation between tumor heterogeneity and prognosis, as well as chemotherapy response, through single-cell and spatial transcriptomic analysis. We collected and curated 28 HGSOC patients' single-cell transcriptomic data from five datasets. Then, we developed a novel text-mining-based machine-learning approach to deconstruct the evolutionary patterns of tumor cell functions. We then identified key tumor-related genes within different evolutionary branches, characterized the microenvironmental cell compositions that various functional tumor cells depend on, and analyzed the intra- and intertumoral heterogeneity as well as the tumor microenvironments. These analyses were conducted in relation to the prognosis and chemotherapy response in HGSOC patients. We validated our findings in two spatial and seven bulk transcriptomic datasets (total: 1,030 patients). Using transcriptomic clusters as proxies for functional clonality, we identified a significant increase in tumor cell state heterogeneity that was strongly correlated with patient prognosis and treatment response. Furthermore, increased intra- and intertumoral functional clonality was associated with the characteristics of cancer-associated fibroblasts (CAFs). The spatial proximity between CXCL12-positive CAFs and tumor cells, mediated through the CXCL12/CXCR4 interaction, was highly positively correlated with poor prognosis and chemotherapy resistance in HGSOC. Finally, we constructed a panel of 24 genes through statistical modeling that correlate with CXCL12-positive fibroblasts and can predict both prognosis and the response to chemotherapy in HGSOC patients. Our study offers insights into the collective behavior of tumor cell communities in HGSOC, as well as potential drivers of tumor evolution in response to therapy. There was a strong association between CXCL12-positive fibroblasts and tumor progression, as well as treatment outcomes.
Collapse
Affiliation(s)
- Tingjie Wang
- Department of Molecular Pathology, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan, People's Republic of China
- Henan Key Laboratory of Molecular Pathology, Zhengzhou, People's Republic of China
| | - Lingxi Tian
- MOE Key Laboratory of Intelligent Biomanufacturing, School of Bioengineering, Dalian University of Technology, Dalian, 116024, People's Republic of China
| | - Bing Wei
- Department of Molecular Pathology, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan, People's Republic of China
- Henan Key Laboratory of Molecular Pathology, Zhengzhou, People's Republic of China
| | - Jun Li
- Department of Molecular Pathology, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan, People's Republic of China
- Henan Key Laboratory of Molecular Pathology, Zhengzhou, People's Republic of China
| | - Cuiyun Zhang
- Department of Molecular Pathology, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan, People's Republic of China
- Henan Key Laboratory of Molecular Pathology, Zhengzhou, People's Republic of China
| | - Ruitao Long
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Xiaofei Zhu
- Department of Clinical Laboratory, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, People's Republic of China
- Henan Key Laboratory of Immunology and Targeted Drugs, Xinxiang Key Laboratory of Tumor Microenvironment and Immunotherapy, School of Medical Technology, Xinxiang Medical University, Xinxiang, People's Republic of China
| | - Yougai Zhang
- Department of Molecular Pathology, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan, People's Republic of China
- Henan Key Laboratory of Molecular Pathology, Zhengzhou, People's Republic of China
| | - Bo Wang
- Department of Molecular Pathology, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan, People's Republic of China
- Henan Key Laboratory of Molecular Pathology, Zhengzhou, People's Republic of China
| | - Guangbo Tang
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Jun Yang
- MOE Key Laboratory of Intelligent Biomanufacturing, School of Bioengineering, Dalian University of Technology, Dalian, 116024, People's Republic of China.
| | - Yongjun Guo
- Department of Molecular Pathology, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan, People's Republic of China.
- Henan Key Laboratory of Molecular Pathology, Zhengzhou, People's Republic of China.
| |
Collapse
|
16
|
Yuan Z, He J, Li Z, Fan B, Zhang L, Man X. Targeting autophagy in urological system cancers: From underlying mechanisms to therapeutic implications. Biochim Biophys Acta Rev Cancer 2024; 1879:189196. [PMID: 39426690 DOI: 10.1016/j.bbcan.2024.189196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 09/27/2024] [Accepted: 10/07/2024] [Indexed: 10/21/2024]
Abstract
The urological system, including kidneys, ureters, bladder, urethra and prostate is known to be vital for blood filtration, waste elimination and electrolyte balance. Notably, urological system cancers represent a significant portion of global cancer diagnoses and mortalities. The current therapeutic strategies for early-stage cancer primarily involve resection surgery, which significantly affects the quality of life of patients, whereas advanced-stage cancer often relies on less effective chemo- or radiotherapy. Recently, accumulating evidence has revealed that autophagy, a crucial process in which excess organelles or inclusions within cells are removed to maintain cell homeostasis, has numerous links to urological system cancers. In this review, we focus on summarizing the underlying two-sided mechanisms of autophagy in urological system cancers. We also review the current clinical drugs targeting autophagy, which demonstrate significant potential in improving treatment outcomes for urological system cancers. In addition, we provide an overview of the research status of novel small molecule compounds targeting autophagy that are in the preclinical stages of investigation. Furthermore, drug combinations based on autophagy modulation strategies in urological system cancers are systematically summarized and discussed. These findings provide comprehensive new insight for the future discovery of more autophagy-related drug candidates.
Collapse
Affiliation(s)
- Ziyue Yuan
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Jiani He
- Department of Urology, Department of Surgical Oncology and Breast Surgery, Institute of Urology, The First Hospital of China Medical University, Shenyang 110001, China
| | - Zhijia Li
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Bo Fan
- Department of Urology, Institute of Precision Drug Innovation and Cancer Center, Second Affiliated Hospital of Dalian Medical University, Dalian 116023, China.
| | - Lan Zhang
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China; Department of Urology, Institute of Precision Drug Innovation and Cancer Center, Second Affiliated Hospital of Dalian Medical University, Dalian 116023, China.
| | - Xiaojun Man
- Department of Urology, Department of Surgical Oncology and Breast Surgery, Institute of Urology, The First Hospital of China Medical University, Shenyang 110001, China.
| |
Collapse
|
17
|
Ma Z, Zhou F, Jin H, Wu X. Crosstalk between CXCL12/CXCR4/ACKR3 and the STAT3 Pathway. Cells 2024; 13:1027. [PMID: 38920657 PMCID: PMC11201928 DOI: 10.3390/cells13121027] [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: 04/25/2024] [Revised: 06/09/2024] [Accepted: 06/10/2024] [Indexed: 06/27/2024] Open
Abstract
The reciprocal modulation between the CXCL12/CXCR4/ACKR3 axis and the STAT3 signaling pathway plays a crucial role in the progression of various diseases and neoplasms. Activation of the CXCL12/CXCR4/ACKR3 axis triggers the STAT3 pathway through multiple mechanisms, while the STAT3 pathway also regulates the expression of CXCL12. This review offers a thorough and systematic analysis of the reciprocal regulatory mechanisms between the CXCL12/CXCR4/ACKR3 signaling axis and the STAT3 signaling pathway in the context of diseases, particularly tumors. It explores the potential clinical applications in tumor treatment, highlighting possible therapeutic targets and novel strategies for targeted tumor therapy.
Collapse
Affiliation(s)
| | | | | | - Xiaoming Wu
- Laboratory of Molecular Genetics of Aging & Tumor, Medical School, Kunming University of Science and Technology, Chenggong Campus, 727 South Jingming Road, Kunming 650500, China; (Z.M.); (F.Z.); (H.J.)
| |
Collapse
|
18
|
Hsu SK, Chou CK, Lin IL, Chang WT, Kuo IY, Chiu CC. Deubiquitinating enzymes: potential regulators of the tumor microenvironment and implications for immune evasion. Cell Commun Signal 2024; 22:259. [PMID: 38715050 PMCID: PMC11075295 DOI: 10.1186/s12964-024-01633-7] [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: 02/26/2024] [Accepted: 04/24/2024] [Indexed: 05/12/2024] Open
Abstract
Ubiquitination and deubiquitination are important forms of posttranslational modification that govern protein homeostasis. Deubiquitinating enzymes (DUBs), a protein superfamily consisting of more than 100 members, deconjugate ubiquitin chains from client proteins to regulate cellular homeostasis. However, the dysregulation of DUBs is reportedly associated with several diseases, including cancer. The tumor microenvironment (TME) is a highly complex entity comprising diverse noncancerous cells (e.g., immune cells and stromal cells) and the extracellular matrix (ECM). Since TME heterogeneity is closely related to tumorigenesis and immune evasion, targeting TME components has recently been considered an attractive therapeutic strategy for restoring antitumor immunity. Emerging studies have revealed the involvement of DUBs in immune modulation within the TME, including the regulation of immune checkpoints and immunocyte infiltration and function, which renders DUBs promising for potent cancer immunotherapy. Nevertheless, the roles of DUBs in the crosstalk between tumors and their surrounding components have not been comprehensively reviewed. In this review, we discuss the involvement of DUBs in the dynamic interplay between tumors, immune cells, and stromal cells and illustrate how dysregulated DUBs facilitate immune evasion and promote tumor progression. We also summarize potential small molecules that target DUBs to alleviate immunosuppression and suppress tumorigenesis. Finally, we discuss the prospects and challenges regarding the targeting of DUBs in cancer immunotherapeutics and several urgent problems that warrant further investigation.
Collapse
Affiliation(s)
- Sheng-Kai Hsu
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung, 807, Taiwan
| | - Chon-Kit Chou
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Science, University of Macau, Macau SAR, 999078, P.R. China
| | - I-Ling Lin
- Department of Medical Laboratory Science and Biotechnology, Kaohsiung Medical University, Kaohsiung, 807, Taiwan
| | - Wen-Tsan Chang
- Division of General and Digestive Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung, 807, Taiwan
- Department of Surgery, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 807, Taiwan
- Center for Cancer Research, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, 807, Taiwan
| | - I-Ying Kuo
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung, 807, Taiwan.
| | - Chien-Chih Chiu
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung, 807, Taiwan.
- Center for Cancer Research, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, 807, Taiwan.
- Department of Biological Sciences, National Sun Yat-Sen University, Kaohsiung, 804, Taiwan.
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung, 807, Taiwan.
| |
Collapse
|
19
|
Dong C, Hui P, Wu Z, Li J, Man X. CircRNA LOC729852 promotes bladder cancer progression by regulating macrophage polarization and recruitment via the miR-769-5p/IL-10 axis. J Cell Mol Med 2024; 28:e18225. [PMID: 38506082 PMCID: PMC10951884 DOI: 10.1111/jcmm.18225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 02/19/2024] [Accepted: 02/23/2024] [Indexed: 03/21/2024] Open
Abstract
Circular RNAs (circRNAs) function as tumour promoters or suppressors in bladder cancer (BLCA) by regulating genes involved in macrophage recruitment and polarization. However, the underlying mechanisms are largely unknown. The aim of this study was to determine the biological role of circLOC729852 in BLCA. CircLOC729852 was upregulated in BLCA tissues and correlated with increased proliferation, migration and epithelial mesenchymal transition (EMT) of BCLA cells. MiR-769-5p was identified as a target for circLOC729852, which can upregulate IL-10 expression by directly binding to and suppressing miR-769-5p. Furthermore, our results indicated that the circLOC729852/miR-769-5p/IL-10 axis modulates autophagy signalling in BLCA cells and promotes the recruitment and M2 polarization of TAMs by activating the JAK2/STAT3 signalling pathway. In addition, circLOC729852 also promoted the growth of BLCA xenografts and M2 macrophage infiltration in vivo. Thus, circLOC729852 functions as an oncogene in BLCA by inducing secretion of IL-10 by the M2 TAMs, which then facilitates tumour cell growth and migration. Taken together, circLOC729852 is a potential diagnostic biomarker and therapeutic target for BLCA.
Collapse
Affiliation(s)
- Changming Dong
- Department of Urology, China Medical UniversityThe First Hospital of China Medical UniversityShenyangLiaoningChina
- Department of UrologyThe First Hospital of China Medical UniversityShenyangLiaoningPR China
| | - Pengyu Hui
- Department of UrologyThe Second Affiliated Hospital of Xi'an Medical UniversityXi'anShaanxiChina
| | - Zhengqi Wu
- Department of Urology, China Medical UniversityThe First Hospital of China Medical UniversityShenyangLiaoningChina
| | - Jianfeng Li
- Department of Urology, China Medical UniversityThe First Hospital of China Medical UniversityShenyangLiaoningChina
| | - Xiaojun Man
- Department of Urology, China Medical UniversityThe First Hospital of China Medical UniversityShenyangLiaoningChina
- Department of UrologyThe First Hospital of China Medical UniversityShenyangLiaoningPR China
| |
Collapse
|