1
|
Du SL, Zhou YT, Hu HJ, Lin L, Zhang ZQ. Silica-induced ROS in alveolar macrophages and its role on the formation of pulmonary fibrosis via polarizing macrophages into M2 phenotype: a review. Toxicol Mech Methods 2025; 35:89-100. [PMID: 39223849 DOI: 10.1080/15376516.2024.2400323] [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: 06/12/2024] [Revised: 08/24/2024] [Accepted: 08/29/2024] [Indexed: 09/04/2024]
Abstract
Alveolar macrophages (AMs), the first line against the invasion of foreign invaders, play a predominant role in the pathogenesis of silicosis. Studies have shown that inhaled silica dust is recognized and engulfed by AMs, resulting in the production of large amounts of silica-induced reactive oxygen species (ROS), including particle-derived ROS and macrophage-derived ROS. These ROS change the microenvironment of the AMs where the macrophage phenotype is stimulated to swift from M0 to M1 and/or M2, and ultimately emerge as the M2 phenotype to trigger silicosis. This is a complex process accompanied by various molecular biological events. Unfortunately, the detailed processes and mechanisms have not been systematically described. In this review, we first systematically introduce the process of ROS induced by silica in AMs. Then, describe the role and molecular mechanism of M2-type macrophage polarization caused by silica-induced ROS. Finally, we review the mechanism of pulmonary fibrosis induced by M2 polarized AMs. We conclude that silica-induced ROS initiate the fibrotic process of silicosis by inducing macrophage into M2 phenotype, and that targeted intervention of silica-induced ROS in AMs can reprogram the macrophage polarization and ameliorate the pathogenesis of silicosis.
Collapse
Affiliation(s)
- Shu-Ling Du
- School of Public Health, Shandong Second Medical University, Weifang, China
- School of Public Health, Jining Medical University, Jining, China
| | - Yu-Ting Zhou
- School of Public Health, Jining Medical University, Jining, China
| | - Hui-Jie Hu
- School of Public Health, Shandong Second Medical University, Weifang, China
- School of Public Health, Jining Medical University, Jining, China
| | - Li Lin
- School of Public Health, Jining Medical University, Jining, China
| | - Zhao-Qiang Zhang
- School of Public Health, Jining Medical University, Jining, China
| |
Collapse
|
2
|
Limonta P, Marchesi S, Giannitti G, Casati L, Fontana F. The biological function of extracellular vesicles in prostate cancer and their clinical application as diagnostic and prognostic biomarkers. Cancer Metastasis Rev 2024; 43:1611-1627. [PMID: 39316264 PMCID: PMC11554767 DOI: 10.1007/s10555-024-10210-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 09/05/2024] [Indexed: 09/25/2024]
Abstract
Prostate cancer (PCa) is one of the most commonly diagnosed malignancies and main causes of cancer-related deaths worldwide. It is characterized by high heterogeneity, ranging from slow-growing tumor to metastatic disease. Since both therapy selection and outcome strongly rely on appropriate patient stratification, it is crucial to differentiate benign from more aggressive conditions using new and improved diagnostic and prognostic biomarkers. Extracellular vesicles (EVs) are membrane-coated particles carrying a specific biological cargo composed of nucleic acids, proteins, and metabolites. Here, we provide an overview of the role of EVs in PCa, focusing on both their biological function and clinical value. Specifically, we summarize the oncogenic role of EVs in mediating the interactions with PCa microenvironment as well as the horizontal transfer of metastatic traits and drug resistance between PCa cells. Furthermore, we discuss the potential usage of EVs as innovative tools for PCa diagnosis and prognosis.
Collapse
Affiliation(s)
- Patrizia Limonta
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università Degli Studi Di Milano, Milan, Italy
| | - Sara Marchesi
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università Degli Studi Di Milano, Milan, Italy
| | - Gaia Giannitti
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università Degli Studi Di Milano, Milan, Italy
| | - Lavinia Casati
- Department of Health Sciences, Università Degli Studi Di Milano, Milan, Italy
| | - Fabrizio Fontana
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università Degli Studi Di Milano, Milan, Italy.
| |
Collapse
|
3
|
Pakula H, Pederzoli F, Fanelli GN, Nuzzo PV, Rodrigues S, Loda M. Deciphering the Tumor Microenvironment in Prostate Cancer: A Focus on the Stromal Component. Cancers (Basel) 2024; 16:3685. [PMID: 39518123 PMCID: PMC11544791 DOI: 10.3390/cancers16213685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 10/25/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024] Open
Abstract
Prostate cancer progression is significantly affected by its tumor microenvironment, in which mesenchymal cells play a crucial role. Stromal cells are modified by cancer mutations, response to androgens, and lineage plasticity, and in turn, engage with epithelial tumor cells via a complex array of signaling pathways and ligand-receptor interactions, ultimately affecting tumor growth, immune interaction, and response to therapy. The metabolic rewiring and interplay in the microenvironment play an additional role in affecting the growth and progression of prostate cancer. Finally, therapeutic strategies and novel clinical trials with agents that target the stromal microenvironment or disrupt the interaction between cellular compartments are described. This review underscores cancer-associated fibroblasts as essential contributors to prostate cancer biology, emphasizing their potential as prognostic indicators and therapeutic targets.
Collapse
Affiliation(s)
- Hubert Pakula
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY 10021, USA; (H.P.); (F.P.); (G.N.F.); (P.V.N.); (S.R.)
| | - Filippo Pederzoli
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY 10021, USA; (H.P.); (F.P.); (G.N.F.); (P.V.N.); (S.R.)
| | - Giuseppe Nicolò Fanelli
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY 10021, USA; (H.P.); (F.P.); (G.N.F.); (P.V.N.); (S.R.)
| | - Pier Vitale Nuzzo
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY 10021, USA; (H.P.); (F.P.); (G.N.F.); (P.V.N.); (S.R.)
| | - Silvia Rodrigues
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY 10021, USA; (H.P.); (F.P.); (G.N.F.); (P.V.N.); (S.R.)
| | - Massimo Loda
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY 10021, USA; (H.P.); (F.P.); (G.N.F.); (P.V.N.); (S.R.)
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, Belfer Research Building, 413 East 69th Street, New York, NY 10021, USA
- Department of Oncologic Pathology, Dana-Farber Cancer Institute and Harvard Medical School, 450 Brookline Ave, Boston, MA 02215, USA
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford OX1 2JD, UK
| |
Collapse
|
4
|
Dai Q, Peng Y, He P, Wu X. Interactions and communications in the prostate tumour microenvironment: evolving towards effective cancer therapy. J Drug Target 2024:1-21. [PMID: 39445641 DOI: 10.1080/1061186x.2024.2418344] [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: 09/02/2024] [Revised: 10/02/2024] [Accepted: 10/14/2024] [Indexed: 10/25/2024]
Abstract
Prostate cancer is one of the most common malignancies in men. The tumour microenvironment (TME) has a critical role in the initiation, progression, and metastasis of prostate cancer. TME contains various cell types, including cancer-associated fibroblasts (CAFs), endothelial cells, immune cells such as macrophages, lymphocytes B and T, natural killer (NK) cells, and other proteins such as extracellular matrix (ECM) components. The interactions and communications between these cells within the TME are crucial for the growth and response of various solid tumours, such as prostate cancer to different anticancer modalities. In this review article, we exemplify the various mechanisms by which the TME influences prostate cancer progression. The roles of different cells, cytokines, chemokines, and growth factors in modulating the immune response and prostate tumour growth will be discussed. The impact of these cells and factors and other ECM components on tumour cell invasion and metastasis will also be discussed. We explain how these interactions in TME can affect the response of prostate cancer to therapy. We also highlight the importance of understanding these interactions to develop novel therapeutic approaches for prostate cancer.
Collapse
Affiliation(s)
- Qiang Dai
- Department of Urology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Yanling Peng
- Department of Urology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Peng He
- Department of Urology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Xiaojun Wu
- Department of Urology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| |
Collapse
|
5
|
Chen L, Xu YX, Wang YS, Ren YY, Dong XM, Wu P, Xie T, Zhang Q, Zhou JL. Prostate cancer microenvironment: multidimensional regulation of immune cells, vascular system, stromal cells, and microbiota. Mol Cancer 2024; 23:229. [PMID: 39395984 PMCID: PMC11470719 DOI: 10.1186/s12943-024-02137-1] [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: 08/14/2024] [Accepted: 09/23/2024] [Indexed: 10/14/2024] Open
Abstract
BACKGROUND Prostate cancer (PCa) is one of the most prevalent malignancies in males worldwide. Increasing research attention has focused on the PCa microenvironment, which plays a crucial role in tumor progression and therapy resistance. This review aims to provide a comprehensive overview of the key components of the PCa microenvironment, including immune cells, vascular systems, stromal cells, and microbiota, and explore their implications for diagnosis and treatment. METHODS Keywords such as "prostate cancer", "tumor microenvironment", "immune cells", "vascular system", "stromal cells", and "microbiota" were used for literature retrieval through online databases including PubMed and Web of Science. Studies related to the PCa microenvironment were selected, with a particular focus on those discussing the roles of immune cells, vascular systems, stromal cells, and microbiota in the development, progression, and treatment of PCa. The selection criteria prioritized peer-reviewed articles published in the last five years, aiming to summarize and analyze the latest research advancements and clinical relevance regarding the PCa microenvironment. RESULTS The PCa microenvironment is highly complex and dynamic, with immune cells contributing to immunosuppressive conditions, stromal cells promoting tumor growth, and microbiota potentially affecting androgen metabolism. Vascular systems support angiogenesis, which fosters tumor expansion. Understanding these components offers insight into the mechanisms driving PCa progression and opens avenues for novel therapeutic strategies targeting the tumor microenvironment. CONCLUSIONS A deeper understanding of the PCa microenvironment is crucial for advancing diagnostic techniques and developing precision therapies. This review highlights the potential of targeting the microenvironment to improve patient outcomes, emphasizing its significance in the broader context of PCa research and treatment innovation.
Collapse
Affiliation(s)
- Lin Chen
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
| | - Yu-Xin Xu
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
| | - Yuan-Shuo Wang
- School of Pharmacy, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Ying-Ying Ren
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
| | - Xue-Man Dong
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
| | - Pu Wu
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
| | - Tian Xie
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China.
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China.
| | - Qi Zhang
- Department of Urology, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang, 310014, China.
| | - Jian-Liang Zhou
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China.
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China.
| |
Collapse
|
6
|
Vecchiotti D, Clementi L, Cornacchia E, Di Vito Nolfi M, Verzella D, Capece D, Zazzeroni F, Angelucci A. Evidence of the Link between Stroma Remodeling and Prostate Cancer Prognosis. Cancers (Basel) 2024; 16:3215. [PMID: 39335188 PMCID: PMC11430343 DOI: 10.3390/cancers16183215] [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: 08/11/2024] [Revised: 09/18/2024] [Accepted: 09/18/2024] [Indexed: 09/30/2024] Open
Abstract
Prostate cancer (PCa), the most commonly diagnosed cancer in men worldwide, is particularly challenging for oncologists when a precise prognosis needs to be established. Indeed, the entire clinical management in PCa has important drawbacks, generating an intense debate concerning the possibility to individuate molecular biomarkers able to avoid overtreatment in patients with pathological indolent cancers. To date, the paradigmatic change in the view of cancer pathogenesis prompts to look for prognostic biomarkers not only in cancer epithelial cells but also in the tumor microenvironment. PCa ecology has been defined with increasing details in the last few years, and a number of promising key markers associated with the reactive stroma are now available. Here, we provide an updated description of the most biologically significant and cited prognosis-oriented microenvironment biomarkers derived from the main reactive processes during PCa pathogenesis: tissue adaptations, inflammatory response and metabolic reprogramming. Proposed biomarkers include factors involved in stromal cell differentiation, cancer-normal cell crosstalk, angiogenesis, extracellular matrix remodeling and energy metabolism.
Collapse
Affiliation(s)
- Davide Vecchiotti
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Letizia Clementi
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Emanuele Cornacchia
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Mauro Di Vito Nolfi
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Daniela Verzella
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Daria Capece
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Francesca Zazzeroni
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Adriano Angelucci
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| |
Collapse
|
7
|
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
|
8
|
Bossio S, Urlandini L, Perri A, Conforti F, Aversa A, Di Agostino S, Rago V. Prostate Cancer: Emerging Modifiable Risk Factors and Therapeutic Strategies in the Management of Advanced Cancer. Life (Basel) 2024; 14:1094. [PMID: 39337878 PMCID: PMC11433263 DOI: 10.3390/life14091094] [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: 07/24/2024] [Revised: 08/18/2024] [Accepted: 08/29/2024] [Indexed: 09/30/2024] Open
Abstract
Prostate cancer (PCa) is the third highest cause of cancer death in men. PCa is a very heterogeneous tumor form in terms of grade, phenotypes, and genetics, often accompanied by complex networks. PCa is characterized by slow growth that does not compromise the patient's quality of life, unlike more aggressive forms showing rapid growth and progression. Early diagnosis, even for the most aggressive forms, increases the possibilities of cure with less aggressive treatments and fewer side effects. However, it is important to know how to decrease the exposure to modifiable risk factors, including diet, sedentary life, smoking and alcohol, can represent an effective tool to reduce the incidence of PCa. In addition, the chronic exposure to environmental factors, most of which act as endocrine disruptors, is the focus of recent studies for their potential role in promoting the onset and progression of PCa. Although molecular therapies and clinical trials for biomarker identification have been introduced into the management of PCa, these still lag behind research performed in other solid tumors. This review provides an overview of the modifiable factors of PCa, linked to lifestyle and environmental pollutants, which together with the development of new therapeutic targets, can reduce the incidence of PCa and improve the quality of life of patients.
Collapse
Affiliation(s)
- Sabrina Bossio
- Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, 88100 Catanzaro, Italy; (S.B.); (A.A.)
| | - Lidia Urlandini
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy; (L.U.); (V.R.)
| | - Anna Perri
- Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, 88100 Catanzaro, Italy; (S.B.); (A.A.)
| | | | - Antonio Aversa
- Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, 88100 Catanzaro, Italy; (S.B.); (A.A.)
| | - Silvia Di Agostino
- Department of Health Sciences, Magna Graecia University of Catanzaro, 88100 Catanzaro, Italy;
| | - Vittoria Rago
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy; (L.U.); (V.R.)
| |
Collapse
|
9
|
Ullah A, Jiao W, Shen B. The role of proinflammatory cytokines and CXC chemokines (CXCL1-CXCL16) in the progression of prostate cancer: insights on their therapeutic management. Cell Mol Biol Lett 2024; 29:73. [PMID: 38745115 PMCID: PMC11094955 DOI: 10.1186/s11658-024-00591-9] [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: 08/30/2023] [Accepted: 05/06/2024] [Indexed: 05/16/2024] Open
Abstract
Reproductive cancers are malignancies that develop in the reproductive organs. One of the leading cancers affecting the male reproductive system on a global scale is prostate cancer (PCa). The negative consequences of PCa metastases endure and are severe, significantly affecting mortality and life quality for those who are affected. The association between inflammation and PCa has captured interest for a while. Inflammatory cells, cytokines, CXC chemokines, signaling pathways, and other elements make up the tumor microenvironment (TME), which is characterized by inflammation. Inflammatory cytokines and CXC chemokines are especially crucial for PCa development and prognosis. Cytokines (interleukins) and CXC chemokines such as IL-1, IL-6, IL-7, IL-17, TGF-β, TNF-α, CXCL1-CXCL6, and CXCL8-CXCL16 are thought to be responsible for the pleiotropic effects of PCa, which include inflammation, progression, angiogenesis, leukocyte infiltration in advanced PCa, and therapeutic resistance. The inflammatory cytokine and CXC chemokines systems are also promising candidates for PCa suppression and immunotherapy. Therefore, the purpose of this work is to provide insight on how the spectra of inflammatory cytokines and CXC chemokines evolve as PCa develops and spreads. We also discussed recent developments in our awareness of the diverse molecular signaling pathways of these circulating cytokines and CXC chemokines, as well as their associated receptors, which may one day serve as PCa-targeted therapies. Moreover, the current status and potential of theranostic PCa therapies based on cytokines, CXC chemokines, and CXC receptors (CXCRs) are examined.
Collapse
Affiliation(s)
- Amin Ullah
- Joint Laboratory of Artificial Intelligence for Critical Care Medicine, Department of Critical Care Medicine and Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Wang Jiao
- Joint Laboratory of Artificial Intelligence for Critical Care Medicine, Department of Critical Care Medicine and Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Bairong Shen
- Joint Laboratory of Artificial Intelligence for Critical Care Medicine, Department of Critical Care Medicine and Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
10
|
Bian X, Wang W, Abudurexiti M, Zhang X, Ma W, Shi G, Du L, Xu M, Wang X, Tan C, Sun H, He X, Zhang C, Zhu Y, Zhang M, Ye D, Wang J. Integration Analysis of Single-Cell Multi-Omics Reveals Prostate Cancer Heterogeneity. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2305724. [PMID: 38483933 PMCID: PMC11095148 DOI: 10.1002/advs.202305724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 02/25/2024] [Indexed: 05/16/2024]
Abstract
Prostate cancer (PCa) is an extensive heterogeneous disease with a complex cellular ecosystem in the tumor microenvironment (TME). However, the manner in which heterogeneity is shaped by tumors and stromal cells, or vice versa, remains poorly understood. In this study, single-cell RNA sequencing, spatial transcriptomics, and bulk ATAC-sequence are integrated from a series of patients with PCa and healthy controls. A stemness subset of club cells marked with SOX9highARlow expression is identified, which is markedly enriched after neoadjuvant androgen-deprivation therapy (ADT). Furthermore, a subset of CD8+CXCR6+ T cells that function as effector T cells is markedly reduced in patients with malignant PCa. For spatial transcriptome analysis, machine learning and computational intelligence are comprehensively utilized to identify the cellular diversity of prostate cancer cells and cell-cell communication in situ. Macrophage and neutrophil state transitions along the trajectory of cancer progression are also examined. Finally, the immunosuppressive microenvironment in advanced PCa is found to be associated with the infiltration of regulatory T cells (Tregs), potentially induced by an FAP+ fibroblast subset. In summary, the cellular heterogeneity is delineated in the stage-specific PCa microenvironment at single-cell resolution, uncovering their reciprocal crosstalk with disease progression, which can be helpful in promoting PCa diagnosis and therapy.
Collapse
Affiliation(s)
- Xiaojie Bian
- Department of UrologyFudan University Shanghai Cancer CenterDepartment of OncologyShanghai Medical CollegeFudan UniversityShanghai200032China
- Cancer InstituteShanghai Urological Cancer InstituteFudan University Shanghai Cancer CenterDepartment of OncologyShanghai Medical CollegeFudan UniversityShanghai200032China
| | - Wenfeng Wang
- Cancer InstituteShanghai Urological Cancer InstituteFudan University Shanghai Cancer CenterDepartment of OncologyShanghai Medical CollegeFudan UniversityShanghai200032China
| | - Mierxiati Abudurexiti
- Cancer InstituteShanghai Urological Cancer InstituteFudan University Shanghai Cancer CenterDepartment of OncologyShanghai Medical CollegeFudan UniversityShanghai200032China
- Department of UrologyShanghai Pudong New Area Gongli HospitalShanghai200135China
| | - Xingming Zhang
- Cancer InstituteShanghai Urological Cancer InstituteFudan University Shanghai Cancer CenterDepartment of OncologyShanghai Medical CollegeFudan UniversityShanghai200032China
| | - Weiwei Ma
- Department of UrologyFudan University Shanghai Cancer CenterDepartment of OncologyShanghai Medical CollegeFudan UniversityShanghai200032China
- Cancer InstituteShanghai Urological Cancer InstituteFudan University Shanghai Cancer CenterDepartment of OncologyShanghai Medical CollegeFudan UniversityShanghai200032China
| | - Guohai Shi
- Department of UrologyFudan University Shanghai Cancer CenterDepartment of OncologyShanghai Medical CollegeFudan UniversityShanghai200032China
| | - Leilei Du
- Cancer InstituteShanghai Urological Cancer InstituteFudan University Shanghai Cancer CenterDepartment of OncologyShanghai Medical CollegeFudan UniversityShanghai200032China
| | - Midie Xu
- Department of PathologyFudan University Shanghai Cancer CenterShanghai200032China
| | - Xin Wang
- Department of PathologyFudan University Shanghai Cancer CenterShanghai200032China
| | - Cong Tan
- Department of PathologyFudan University Shanghai Cancer CenterShanghai200032China
| | - Hui Sun
- Department of PathologyFudan University Shanghai Cancer CenterShanghai200032China
| | - Xiadi He
- Department of Cancer BiologyDana‐Farber Cancer InstituteBostonMA02215USA
- Department of Biological Chemistry and Molecular PharmacologyHarvard Medical SchoolBostonMA02115USA
| | - Chenyue Zhang
- Department of Integrated TherapyFudan University Shanghai Cancer CenterShanghai200032China
| | - Yao Zhu
- Department of UrologyFudan University Shanghai Cancer CenterDepartment of OncologyShanghai Medical CollegeFudan UniversityShanghai200032China
- Cancer InstituteShanghai Urological Cancer InstituteFudan University Shanghai Cancer CenterDepartment of OncologyShanghai Medical CollegeFudan UniversityShanghai200032China
| | - Min Zhang
- Pediatric Translational Medicine Institute and Pediatric Congenital Heart Disease InstituteShanghai Children's Medical CenterShanghai Jiao Tong University School of MedicineShanghai200127China
| | - Dingwei Ye
- Department of UrologyFudan University Shanghai Cancer CenterDepartment of OncologyShanghai Medical CollegeFudan UniversityShanghai200032China
- Cancer InstituteShanghai Urological Cancer InstituteFudan University Shanghai Cancer CenterDepartment of OncologyShanghai Medical CollegeFudan UniversityShanghai200032China
| | - Jianhua Wang
- Cancer InstituteShanghai Urological Cancer InstituteFudan University Shanghai Cancer CenterDepartment of OncologyShanghai Medical CollegeFudan UniversityShanghai200032China
| |
Collapse
|
11
|
Lai C, Wu Z, Li Z, Huang X, Hu Z, Yu H, Yuan Z, Shi J, Hu J, Mulati Y, Liu C, Xu K. Single-cell analysis extracted CAFs-related genes to established online app to predict clinical outcome and radiotherapy prognosis of prostate cancer. Clin Transl Oncol 2024; 26:1240-1255. [PMID: 38070051 DOI: 10.1007/s12094-023-03348-6] [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: 09/14/2023] [Accepted: 11/03/2023] [Indexed: 04/20/2024]
Abstract
BACKGROUND Cancer-associated fibroblasts (CAFs) play a significant role in regulating the clinical outcome and radiotherapy prognosis of prostate cancer (PCa). The aim of this study is to identify CAFs-related genes (CAFsRGs) using single-cell analysis and evaluate their potential for predicting the prognosis and radiotherapy prognosis in PCa. METHODS We acquire transcriptome and single-cell RNA sequencing (scRNA-seq) results of PCa and normal adjacent tissues from The GEO and TCGA databases. The "MCPcounter" and "EPIC" R packages were used to assess the infiltration level of CAFs and examine their correlation with PCa prognosis. ScRNA-seq and differential gene expression analyses were used to extract CAFsRGs. We also applied COX and LASSO analysis to further construct a risk score (CAFsRS) to assess biochemical recurrence-free survival (BRFS) and radiotherapy prognosis of PCa. The predictive efficacy of CAFsRS was evaluated by ROC curves and subgroup analysis. Finally, we integrated the CAFsRS gene signature with relevant clinical features to develop a nomogram, enhancing the predictive accuracy. RESULTS The abundance of CAFs is associated with a poor prognosis of PCa patients. ScRNA-seq and differential gene expression analysis revealed 323 CAFsRGs. After COX and LASSO analysis, we obtained seven CAFsRGs with prognostic significance (PTGS2, FKBP10, ENG, CDH11, COL5A1, COL5A2, and SRD5A2). Additionally, we established a risk score model based on the training set (n = 257). The ROC curve was used to confirm the performance of CAFsRS (The AUC values for 1, 3 and 5-year survival were determined to be 0.732, 0.773, and 0.775, respectively.). The testing set (n = 129), GSE70770 set (n = 199) and GSE116918 set (n = 248) revealed that the model exhibited exceptional predictive performance. This was also confirmed by clinical subgroup analysis. The violin plot demonstrated a statistically significant disparity in the CAFs infiltrations between the high-risk and low-risk groups of CAFsRS. Further analysis confirmed that both CAFsRS and T stage were independent prognostic factors for PCa. The nomogram was then established and its excellent predictive performance was demonstrated through calibration and ROC curves. Finally, we developed an online prognostic prediction app ( https://sysu-symh-cafsnomogram.streamlit.app/ ) to facilitate the practical application of the nomogram. CONCLUSIONS The prognostic prediction risk score model we constructed could accurately predict BRFS and radiotherapy prognosis PCa, which can provide new ideas for clinicians to develop personalized PCa treatment and follow-up programs.
Collapse
Affiliation(s)
- Cong Lai
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510000, Guangdong, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510000, Guangdong, China
| | - Zhikai Wu
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510000, Guangdong, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510000, Guangdong, China
| | - Zhuohang Li
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510000, Guangdong, China
- Guangdong Provincial Clinical Research Center for Urological Diseases, Guangzhou, 510000, Guangdong, China
| | - Xin Huang
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510000, Guangdong, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510000, Guangdong, China
| | - Zhensheng Hu
- Department of Medical Informatics, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510000, Guangdong, China
| | - Hao Yu
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510000, Guangdong, China
- Guangdong Provincial Clinical Research Center for Urological Diseases, Guangzhou, 510000, Guangdong, China
| | - Zhihan Yuan
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510000, Guangdong, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510000, Guangdong, China
| | - Juanyi Shi
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510000, Guangdong, China
- Guangdong Provincial Clinical Research Center for Urological Diseases, Guangzhou, 510000, Guangdong, China
| | - Jintao Hu
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510000, Guangdong, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510000, Guangdong, China
| | - Yelisudan Mulati
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510000, Guangdong, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510000, Guangdong, China
| | - Cheng Liu
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510000, Guangdong, China.
- Guangdong Provincial Clinical Research Center for Urological Diseases, Guangzhou, 510000, Guangdong, China.
| | - Kewei Xu
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510000, Guangdong, China.
- Guangdong Provincial Clinical Research Center for Urological Diseases, Guangzhou, 510000, Guangdong, China.
- Sun Yat-Sen College of Medical Science, Sun Yat-Sen University, Shenzhen, 518000, Guangdong, China.
| |
Collapse
|
12
|
Zhang S, Jia X, Dai H, Zhu X, Song W, Bian S, Wu H, Chen S, Tang Y, Chen J, Jin C, Zhou M, Xie H, Zheng S, Song P. SERPINE2 promotes liver cancer metastasis by inhibiting c-Cbl-mediated EGFR ubiquitination and degradation. Cancer Commun (Lond) 2024; 44:384-407. [PMID: 38407942 PMCID: PMC10958675 DOI: 10.1002/cac2.12527] [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: 08/07/2023] [Revised: 02/03/2024] [Accepted: 02/08/2024] [Indexed: 02/27/2024] Open
Abstract
BACKGROUND Liver cancer is a malignancy with high morbidity and mortality rates. Serpin family E member 2 (SERPINE2) has been reported to play a key role in the metastasis of many tumors. In this study, we aimed to investigate the potential mechanism of SERPINE2 in liver cancer metastasis. METHODS The Cancer Genome Atlas database (TCGA), including DNA methylation and transcriptome sequencing data, was utilized to identify the crucial oncogene associated with DNA methylation and cancer progression in liver cancer. Data from the TCGA and RNA sequencing for 94 pairs of liver cancer tissues were used to explore the correlation between SERPINE2 expression and clinical parameters of patients. DNA methylation sequencing was used to detect the DNA methylation levels in liver cancer tissues and cells. RNA sequencing, cytokine assays, immunoprecipitation (IP) and mass spectrometry (MS) assays, protein stability assays, and ubiquitination assays were performed to explore the regulatory mechanism of SERPINE2 in liver cancer metastasis. Patient-derived xenografts and tumor organoid models were established to determine the role of SERPINE2 in the treatment of liver cancer using sorafenib. RESULTS Based on the public database screening, SERPINE2 was identified as a tumor promoter regulated by DNA methylation. SERPINE2 expression was significantly higher in liver cancer tissues and was associated with the dismal prognosis in patients with liver cancer. SERPINE2 promoted liver cancer metastasis by enhancing cell pseudopodia formation, cell adhesion, cancer-associated fibroblast activation, extracellular matrix remodeling, and angiogenesis. IP/MS assays confirmed that SERPINE2 activated epidermal growth factor receptor (EGFR) and its downstream signaling pathways by interacting with EGFR. Mechanistically, SERPINE2 inhibited EGFR ubiquitination and maintained its protein stability by competing with the E3 ubiquitin ligase, c-Cbl. Additionally, EGFR was activated in liver cancer cells after sorafenib treatment, and SERPINE2 knockdown-induced EGFR downregulation significantly enhanced the therapeutic efficacy of sorafenib against liver cancer. Furthermore, we found that SERPINE2 knockdown also had a sensitizing effect on lenvatinib treatment. CONCLUSIONS SERPINE2 promoted liver cancer metastasis by preventing EGFR degradation via c-Cbl-mediated ubiquitination, suggesting that inhibition of the SERPINE2-EGFR axis may be a potential target for liver cancer treatment.
Collapse
|
13
|
Zong S, Gao J. Identifying the tumor immune microenvironment-associated prognostic genes for prostate cancer. Discov Oncol 2024; 15:42. [PMID: 38376699 PMCID: PMC10879074 DOI: 10.1007/s12672-023-00856-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 12/29/2023] [Indexed: 02/21/2024] Open
Abstract
PURPOSE This study aimed to explore novel tumor immune microenvironment (TIME)-associated biomarkers in prostate adenocarcinoma (PRAD). METHODS PRAD RNA-sequencing data were obtained from UCSC Xena database as the training dataset. The ESTIMATE package was used to evaluate stromal, immune, and tumor purity scores. Differentially expressed genes (DEGs) related to TIME were screened using the immune and stromal scores. Gene functions were analyzed using DAVID. The LASSO method was performed to screen prognostic TIME-related genes. Kaplan-Meier curves were used to evaluate the prognosis of samples. The correlation between the screened genes and immune cell infiltration was explored using Tumor IMmune Estimation Resource. The GSE70768 dataset from the Gene Expression Omnibus was used to validate the expression of the screened genes. RESULTS The ESTIMATE results revealed that high immune, stromal, and ESTIMATE scores and low tumor purity had better prognoses. Function analysis indicated that DEGs are involved in the cytokine-cytokine receptor interaction signaling pathway. In TIME-related DEGs, METTL7B, HOXB8, and TREM1 were closely related to the prognosis. Samples with low expression levels of METTL7B, HOXB8, and TREM1 had better survival times. Similarly, both the validation dataset and qRT-PCR suggested that METTL7B, HOXB8, and TREM1 were significantly decreased. The three genes showed a positive correlation with immune infiltration. CONCLUSIONS This study identified three TIME-related genes, namely, METTL7B, HOXB8, and TREM1, which correlated with the prognosis of patients with PRAD. Targeting the TIME-related genes might have important clinical implications when making decisions for immunotherapy in PRAD.
Collapse
Affiliation(s)
- Shi Zong
- Department of Urology, Union Hospital of Jilin University, No.126, Xian Tai Road, Chang Chun, 130021, China
| | - Ji Gao
- Department of Urology, Union Hospital of Jilin University, No.126, Xian Tai Road, Chang Chun, 130021, China.
| |
Collapse
|
14
|
Lazennec G, Rajarathnam K, Richmond A. CXCR2 chemokine receptor - a master regulator in cancer and physiology. Trends Mol Med 2024; 30:37-55. [PMID: 37872025 PMCID: PMC10841707 DOI: 10.1016/j.molmed.2023.09.003] [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: 08/18/2023] [Revised: 09/14/2023] [Accepted: 09/18/2023] [Indexed: 10/25/2023]
Abstract
Recent findings have modified our understanding of the roles of chemokine receptor CXCR2 and its ligands in cancer, inflammation, and immunity. Studies in Cxcr2 tissue-specific knockout mice show that this receptor is involved in, among other things, cancer, central nervous system (CNS) function, metabolism, reproduction, COVID-19, and the response to circadian cycles. Moreover, CXCR2 involvement in neutrophil function has been revisited not only in physiology but also for its major contribution to cancers. The recent unfolding of the role of CXCR2 in numerous cancers has led to extensive evaluation of multiple CXCR2 antagonists in preclinical and clinical studies. In this review we discuss the potential of targeting CXCR2 for cancer treatment.
Collapse
Affiliation(s)
- Gwendal Lazennec
- Centre National de la Recherche Scientifique (CNRS), Sys2Diag-ALCEDIAG, Cap Delta, Montpellier, France; CNRS Groupement de Recherche (GDR) 3697 'Microenvironment of Tumor Niches', Micronit, France.
| | - Krishna Rajarathnam
- Department of Biochemistry and Molecular Biology, Department of Microbiology and Immunology, Sealy Center for Structural Biology and Molecular Biophysics, University of Texas Medical Branch, Galveston, TX, USA
| | - Ann Richmond
- Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, TN, USA; Vanderbilt University School of Medicine, Department of Pharmacology, Nashville, TN, USA; Vanderbilt Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
15
|
Abstract
Prostate cancer is a leading cause of death in men worldwide. For over 30 years, growing interest has focused on the development of vaccines as treatments for prostate cancer, with the goal of using vaccines to activate immune cells capable of targeting prostate cancer to either eradicate recurrent disease or at least delay disease progression. This interest has been prompted by the prevalence and long natural history of the disease and by the fact that the prostate is an expendable organ. Thus, an immune response elicited by vaccination might not need to target the tumour uniquely but could theoretically target any prostate tissue. To date, different vaccine approaches and targets for prostate cancer have been evaluated in clinical trials. Overall, five approaches have been assessed in randomized phase III trials and sipuleucel-T was approved as a treatment for metastatic castration-resistant prostate cancer, being the only vaccine approved to date by the FDA as a treatment for cancer. Most vaccine approaches showed safety and some evidence of immunological activity but had poor clinical activity when used as monotherapies. However, increased activity has been observed when these vaccines were used in combination with other immune-modulating therapies. This evidence suggests that, in the future, prostate cancer vaccines might be used to activate and expand tumour-specific T cells as part of combination approaches with agents that target tumour-associated immune mechanisms of resistance.
Collapse
Affiliation(s)
- Ichwaku Rastogi
- University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, WI, USA
| | - Anusha Muralidhar
- University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, WI, USA
| | - Douglas G McNeel
- University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, WI, USA.
| |
Collapse
|
16
|
Li D, Xu W, Chang Y, Xiao Y, He Y, Ren S. Advances in landscape and related therapeutic targets of the prostate tumor microenvironment. Acta Biochim Biophys Sin (Shanghai) 2023. [PMID: 37294106 DOI: 10.3724/abbs.2023092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/10/2023] Open
Abstract
The distinct tumor microenvironment (TME) of prostate cancer (PCa), which promotes tumor proliferation and progression, consists of various stromal cells, immune cells, and a dense extracellular matrix (ECM). The understanding of the prostate TME extends to tertiary lymphoid structures (TLSs) and metastasis niches to provide a more concise comprehension of tumor metastasis. These constituents collectively structure the hallmarks of the pro-tumor TME, including immunosuppressive, acidic, and hypoxic niches, neuronal innervation, and metabolic rewiring. In combination with the knowledge of the tumor microenvironment and the advancement of emerging therapeutic technologies, several therapeutic strategies have been developed, and some of them have been tested in clinical trials. This review elaborates on PCa TME components, summarizes various TME-targeted therapies, and provides insights into PCa carcinogenesis, progression, and therapeutic strategies.
Collapse
Affiliation(s)
- Duocai Li
- Department of Urology, Shanghai Changzheng Hospital, Naval Medical University, Shanghai 200003, China
| | - Weidong Xu
- Department of Urology, Shanghai Changzheng Hospital, Naval Medical University, Shanghai 200003, China
| | - Yifan Chang
- Department of Urology, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Yutian Xiao
- Department of Urology, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Yundong He
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai 200062, China
| | - Shancheng Ren
- Department of Urology, Shanghai Changzheng Hospital, Naval Medical University, Shanghai 200003, China
| |
Collapse
|
17
|
Kobelyatskaya AA, Pudova EA, Katunina IV, Snezhkina AV, Fedorova MS, Pavlov VS, Kotelnikova AO, Nyushko KM, Alekseev BY, Krasnov GS, Kudryavtseva AV. Transcriptome Profiling of Prostate Cancer, Considering Risk Groups and the TMPRSS2-ERG Molecular Subtype. Int J Mol Sci 2023; 24:ijms24119282. [PMID: 37298233 DOI: 10.3390/ijms24119282] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 05/12/2023] [Accepted: 05/24/2023] [Indexed: 06/12/2023] Open
Abstract
Molecular heterogeneity in prostate cancer (PCa) is one of the key reasons underlying the differing likelihoods of recurrence after surgical treatment in individual patients of the same clinical category. In this study, we performed RNA-Seq profiling of 58 localized PCa and 43 locally advanced PCa tissue samples obtained as a result of radical prostatectomy on a cohort of Russian patients. Based on bioinformatics analysis, we examined features of the transcriptome profiles within the high-risk group, including within the most commonly represented molecular subtype, TMPRSS2-ERG. The most significantly affected biological processes in the samples were also identified, so that they may be further studied in the search for new potential therapeutic targets for the categories of PCa under consideration. The highest predictive potential was found with the EEF1A1P5, RPLP0P6, ZNF483, CIBAR1, HECTD2, OGN, and CLIC4 genes. We also reviewed the main transcriptome changes in the groups at intermediate risk of PCa-Gleason Score 7 (groups 2 and 3 according to the ISUP classification)-on the basis of which the LPL, MYC, and TWIST1 genes were identified as promising additional prognostic markers, the statistical significance of which was confirmed using qPCR validation.
Collapse
Affiliation(s)
| | - Elena A Pudova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Irina V Katunina
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Anastasiya V Snezhkina
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Maria S Fedorova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Vladislav S Pavlov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | | | - Kirill M Nyushko
- National Medical Research Radiological Center, Ministry of Health of the Russian Federation, 125284 Moscow, Russia
| | - Boris Y Alekseev
- National Medical Research Radiological Center, Ministry of Health of the Russian Federation, 125284 Moscow, Russia
| | - George S Krasnov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Anna V Kudryavtseva
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| |
Collapse
|
18
|
Peng Z, Tong Z, Ren Z, Ye M, Hu K. Cancer-associated fibroblasts and its derived exosomes: a new perspective for reshaping the tumor microenvironment. Mol Med 2023; 29:66. [PMID: 37217855 DOI: 10.1186/s10020-023-00665-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Accepted: 05/14/2023] [Indexed: 05/24/2023] Open
Abstract
Cancer-associated fibroblasts (CAFs) are the most abundant stromal cells within the tumor microenvironment (TME). They extensively communicate with the other cells. Exosome-packed bioactive molecules derived from CAFs can reshape the TME by interacting with other cells and the extracellular matrix, which adds a new perspective for their clinical application in tumor targeted therapy. An in-depth understanding of the biological characteristics of CAF-derived exosomes (CDEs) is critical for depicting the detailed landscape of the TME and developing tailored therapeutic strategies for cancer treatment. In this review, we have summarized the functional roles of CAFs in the TME, particularly focusing on the extensive communication mediated by CDEs that contain biological molecules such as miRNAs, proteins, metabolites, and other components. In addition, we have also highlighted the prospects for diagnostic and therapeutic applications based on CDEs, which could guide the future development of exosome-targeted anti-tumor drugs.
Collapse
Affiliation(s)
- Zhiwei Peng
- Department of General Surgery, First Affiliated Hospital of Anhui Medical University, Anhui, Hefei, 230022, China
| | - Zhiwei Tong
- Department of General Surgery, First Affiliated Hospital of Anhui Medical University, Anhui, Hefei, 230022, China
| | - Zihao Ren
- Department of General Surgery, First Affiliated Hospital of Anhui Medical University, Anhui, Hefei, 230022, China
| | - Manping Ye
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Anhui, Hefei, 230032, China
| | - Kongwang Hu
- Department of General Surgery, First Affiliated Hospital of Anhui Medical University, Anhui, Hefei, 230022, China.
- Department of General Surgery, Fuyang Affiliated Hospital of Anhui Medical University, Anhui, Fuyang, 236000, China.
| |
Collapse
|
19
|
Bone Metastasis of Breast Cancer: Molecular Mechanisms and Therapeutic Strategies. Cancers (Basel) 2022; 14:cancers14235727. [PMID: 36497209 PMCID: PMC9738274 DOI: 10.3390/cancers14235727] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 11/07/2022] [Accepted: 11/17/2022] [Indexed: 11/24/2022] Open
Abstract
Bone metastasis is a common complication of many types of advanced cancer, including breast cancer. Bone metastasis may cause severe pain, fractures, and hypercalcemia, rendering clinical management challenging and substantially reducing the quality of life and overall survival (OS) time of breast cancer patients. Studies have revealed that bone metastasis is related to interactions between tumor cells and the bone microenvironment, and involves complex molecular biological mechanisms, including colonization, osteolytic destruction, and an immunosuppressive bone microenvironment. Agents inhibiting bone metastasis (such as bisphosphate and denosumab) alleviate bone destruction and improve the quality of life of breast cancer patients with bone metastasis. However, the prognosis of these patients remains poor, and the specific biological mechanism of bone metastasis is incompletely understood. Additional basic and clinical studies are urgently needed, to further explore the mechanism of bone metastasis and develop new therapeutic drugs. This review presents a summary of the molecular mechanisms and therapeutic strategies of bone metastasis of breast cancer, aiming to improve the quality of life and prognosis of breast cancer patients and provide a reference for future research directions.
Collapse
|