1
|
Kzhyshkowska J, Shen J, Larionova I. Targeting of TAMs: can we be more clever than cancer cells? Cell Mol Immunol 2024; 21:1376-1409. [PMID: 39516356 PMCID: PMC11607358 DOI: 10.1038/s41423-024-01232-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 10/12/2024] [Indexed: 11/16/2024] Open
Abstract
АBSTRACT: With increasing incidence and geography, cancer is one of the leading causes of death, reduced quality of life and disability worldwide. Principal progress in the development of new anticancer therapies, in improving the efficiency of immunotherapeutic tools, and in the personification of conventional therapies needs to consider cancer-specific and patient-specific programming of innate immunity. Intratumoral TAMs and their precursors, resident macrophages and monocytes, are principal regulators of tumor progression and therapy resistance. Our review summarizes the accumulated evidence for the subpopulations of TAMs and their increasing number of biomarkers, indicating their predictive value for the clinical parameters of carcinogenesis and therapy resistance, with a focus on solid cancers of non-infectious etiology. We present the state-of-the-art knowledge about the tumor-supporting functions of TAMs at all stages of tumor progression and highlight biomarkers, recently identified by single-cell and spatial analytical methods, that discriminate between tumor-promoting and tumor-inhibiting TAMs, where both subtypes express a combination of prototype M1 and M2 genes. Our review focuses on novel mechanisms involved in the crosstalk among epigenetic, signaling, transcriptional and metabolic pathways in TAMs. Particular attention has been given to the recently identified link between cancer cell metabolism and the epigenetic programming of TAMs by histone lactylation, which can be responsible for the unlimited protumoral programming of TAMs. Finally, we explain how TAMs interfere with currently used anticancer therapeutics and summarize the most advanced data from clinical trials, which we divide into four categories: inhibition of TAM survival and differentiation, inhibition of monocyte/TAM recruitment into tumors, functional reprogramming of TAMs, and genetic enhancement of macrophages.
Collapse
Affiliation(s)
- Julia Kzhyshkowska
- Department of Innate Immunity and Tolerance, Institute of Transfusion Medicine and Immunology, Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, University of Heidelberg, Theodor-Kutzer-Ufer, 1-3, 68167, Mannheim, Germany.
- German Red Cross Blood Service Baden-Württemberg - Hessen, Friedrich-Ebert Str. 107, 68167, Mannheim, Germany.
- Laboratory of Translational Cellular and Molecular Biomedicine, National Research Tomsk State University, 634050, Lenina av.36, Tomsk, Russia.
- Bashkir State Medical University of the Ministry of Health of Russia, 450000, Teatralnaya Street, 2a, Ufa, Russia.
| | - Jiaxin Shen
- Department of Innate Immunity and Tolerance, Institute of Transfusion Medicine and Immunology, Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, University of Heidelberg, Theodor-Kutzer-Ufer, 1-3, 68167, Mannheim, Germany
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Irina Larionova
- Laboratory of Translational Cellular and Molecular Biomedicine, National Research Tomsk State University, 634050, Lenina av.36, Tomsk, Russia
- Bashkir State Medical University of the Ministry of Health of Russia, 450000, Teatralnaya Street, 2a, Ufa, Russia
- Laboratory of Molecular Therapy of Cancer, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, 634009, Kooperativnyi st, Tomsk, Russia
| |
Collapse
|
2
|
Liu H, Lv Z, Zhang G, Yan Z, Bai S, Dong D, Wang K. Molecular understanding and clinical aspects of tumor-associated macrophages in the immunotherapy of renal cell carcinoma. J Exp Clin Cancer Res 2024; 43:242. [PMID: 39169402 PMCID: PMC11340075 DOI: 10.1186/s13046-024-03164-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: 07/06/2024] [Accepted: 08/12/2024] [Indexed: 08/23/2024] Open
Abstract
Renal cell carcinoma (RCC) is one of the most common tumors that afflicts the urinary system, accounting for 90-95% of kidney cancer cases. Although its incidence has increased over the past decades, its pathogenesis is still unclear. Tumor-associated macrophages (TAMs) are the most prominent immune cells in the tumor microenvironment (TME), comprising more than 50% of the tumor volume. By interacting with cancer cells, TAMs can be polarized into two distinct phenotypes, M1-type and M2-type TAMs. In the TME, M2-type TAMs, which are known to promote tumorigenesis, are more abundant than M1-type TAMs, which are known to suppress tumor growth. This ratio of M1 to M2 TAMs can create an immunosuppressive environment that contributes to tumor cell progression and survival. This review focused on the role of TAMs in RCC, including their polarization, impacts on tumor proliferation, angiogenesis, invasion, migration, drug resistance, and immunosuppression. In addition, we discussed the potential of targeting TAMs for clinical therapy in RCC. A deeper understanding of the molecular biology of TAMs is essential for exploring innovative therapeutic strategies for the treatment of RCC.
Collapse
Affiliation(s)
- Han Liu
- Department of Urology, Shengjing Hospital of China Medical University, #36 Sanhao Street, Shenyang, Liaoning, 110004, China
| | - Zongwei Lv
- Department of Urology, Shengjing Hospital of China Medical University, #36 Sanhao Street, Shenyang, Liaoning, 110004, China
| | - Gong Zhang
- Department of Urology, Shengjing Hospital of China Medical University, #36 Sanhao Street, Shenyang, Liaoning, 110004, China
| | - Zhenhong Yan
- Department of Urology, Shengjing Hospital of China Medical University, #36 Sanhao Street, Shenyang, Liaoning, 110004, China
| | - Song Bai
- Department of Urology, Shengjing Hospital of China Medical University, #36 Sanhao Street, Shenyang, Liaoning, 110004, China.
| | - Dan Dong
- College of Basic Medical Science, China Medical University, #77 Puhe Road, Shenyang, Liaoning, 110122, China.
| | - Kefeng Wang
- Department of Urology, Shengjing Hospital of China Medical University, #36 Sanhao Street, Shenyang, Liaoning, 110004, China.
| |
Collapse
|
3
|
Yang X, Li X, Xu H, Du S, Wang C, He H. Predicting CTLA4 expression and prognosis in clear cell renal cell carcinoma using a pathomics signature of histopathological images and machine learning. Heliyon 2024; 10:e34877. [PMID: 39145002 PMCID: PMC11320204 DOI: 10.1016/j.heliyon.2024.e34877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 07/15/2024] [Accepted: 07/17/2024] [Indexed: 08/16/2024] Open
Abstract
Background CTLA4, an immune checkpoint, plays an important role in tumor immunotherapy. The purpose of this study was to develop a pathomics signature to evaluate CTLA4 expression and predict clinical outcomes in clear cell renal cell carcinoma (ccRCC) patients. Methods A total of 354 patients from the TCGA-KIRC dataset were enrolled in this study. The patients were stratified into two groups based on the level of CTLA4 expression, and overall survival rates were analyzed between groups. Pathological features were identified using machine learning algorithms, and a gradient boosting machine (GBM) was employed to construct the pathomics signatures for predicting prognosis and CTLA4 expression. The predictive performance of the model was subsequently assessed. Enrichment analysis was performed on diferentially expressed genes related to the pathomics score (PS). Additionally, correlations between PS and TMB, as well as immune infiltration profiles associated with different PS values, were explored. In vitro experiments, CTLA4 knockdown was performed to investigate its impact on cell proliferation, migration, invasion, TGF-β signaling pathway, and macrophage polarization. Results High expression of CTLA4 was associated with an unfavorable prognosis in ccRCC patients. The pathomics signature displayed good performance in the validation set (AUC = 0.737; P < 0.001 in the log-rank test). The PS was positively correlated with CTLA4 expression. We next explored the underlying mechanism and found the associations between the pathomics signature and TGF-β signaling pathways, TMB, and Tregs. Further in vitro experiments demonstrated that CTLA4 knockdown inhibited cell proliferation, migration, invasion, TGF-β expression, and macrophage M2 polarization. Conclusion High expression of CTLA4 was found to correlate with poor prognosis in ccRCC patients. The pathomics signature established by our group using machine learning effectively predicted both patient prognosis and CTLA4 expression levels in ccRCC cases.
Collapse
Affiliation(s)
- Xiaoqun Yang
- Department of Pathology, Shanghai Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Xiangyun Li
- Department of Pathology, Shanghai Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Haimin Xu
- Department of Pathology, Shanghai Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Silin Du
- University Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Chaofu Wang
- Department of Pathology, Shanghai Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Hongchao He
- Department of Urology, Shanghai Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| |
Collapse
|
4
|
Guan B, Li M, Cui D, Shen C, Hao Z, Li X. Single-cell transcriptomic analysis in clear cell renal cell carcinoma: Deciphering the role of APP within the tumour microenvironment. J Cell Mol Med 2024; 28:e18186. [PMID: 38445803 PMCID: PMC10915830 DOI: 10.1111/jcmm.18186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 01/14/2024] [Accepted: 01/31/2024] [Indexed: 03/07/2024] Open
Abstract
Clear cell renal cell carcinoma (ccRCC) represents a significant challenge in oncology, primarily due to its resistance to conventional therapies. Understanding the tumour microenvironment (TME) is crucial for developing new treatment strategies. This study focuses on the role of amyloid precursor protein (APP) in tumour-associated macrophages (TAMs) within the ccRCC TME, exploring its potential as a prognostic biomarker. Basing TAM-related genes, the prognostic model was important to constructed. Employing advanced single-cell transcriptomic analysis, this research dissects the TME of ccRCC at an unprecedented cellular resolution. By isolating and examining the gene expression profiles of individual cells, particularly focusing on TAMs, the study investigates the expression levels of APP and their association with the clinical outcomes of ccRCC patients. The analysis reveals a significant correlation between the expression of APP in TAMs and patient prognosis in ccRCC. Patients with higher APP expression in TAMs showed differing clinical outcomes compared to those with lower expression. This finding suggests that APP could serve as a novel prognostic biomarker for ccRCC, providing insights into the disease progression and potential therapeutic targets. This study underscores the importance of single-cell transcriptomics in understanding the complex dynamics of the TME in ccRCC. The correlation between APP expression in TAMs and patient prognosis highlights APP as a potential prognostic biomarker. However, further research is needed to validate these findings and explore the regulatory mechanisms and therapeutic implications of APP in ccRCC.
Collapse
Affiliation(s)
- Bo Guan
- Department of UrologyFuyang People's Hospital of Anhui Medical UniversityFuyangChina
| | - Ming Li
- Department of UrologyFuyang People's Hospital of Anhui Medical UniversityFuyangChina
| | - Di Cui
- Fuyang Medical CollegeFuyang Normal UniversityFuyangChina
| | - Chen Shen
- Department of UrologyFuyang People's Hospital of Anhui Medical UniversityFuyangChina
- Department of UrologyRenji HospitalShanghaiChina
| | - Zongyao Hao
- Department of UrologyFuyang People's Hospital of Anhui Medical UniversityFuyangChina
- Department of Urologythe First Affiliated Hospital of Anhui Medical UniversityAnhuiChina
- Institute of UrologyAnhui Medical UniversityAnhuiChina
| | - Xiaowei Li
- Department of NephrologyFuyang People's Hospital of Anhui Medical UniversityFuyangChina
| |
Collapse
|
5
|
Cornice J, Verzella D, Arboretto P, Vecchiotti D, Capece D, Zazzeroni F, Franzoso G. NF-κB: Governing Macrophages in Cancer. Genes (Basel) 2024; 15:197. [PMID: 38397187 PMCID: PMC10888451 DOI: 10.3390/genes15020197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 01/26/2024] [Accepted: 01/27/2024] [Indexed: 02/25/2024] Open
Abstract
Tumor-associated macrophages (TAMs) are the major component of the tumor microenvironment (TME), where they sustain tumor progression and or-tumor immunity. Due to their plasticity, macrophages can exhibit anti- or pro-tumor functions through the expression of different gene sets leading to distinct macrophage phenotypes: M1-like or pro-inflammatory and M2-like or anti-inflammatory. NF-κB transcription factors are central regulators of TAMs in cancers, where they often drive macrophage polarization toward an M2-like phenotype. Therefore, the NF-κB pathway is an attractive therapeutic target for cancer immunotherapy in a wide range of human tumors. Hence, targeting NF-κB pathway in the myeloid compartment is a potential clinical strategy to overcome microenvironment-induced immunosuppression and increase anti-tumor immunity. In this review, we discuss the role of NF-κB as a key driver of macrophage functions in tumors as well as the principal strategies to overcome tumor immunosuppression by targeting the NF-κB pathway.
Collapse
Affiliation(s)
- Jessica Cornice
- Department of Immunology and Inflammation, Imperial College London, London W12 0NN, UK; (J.C.); (P.A.)
| | - Daniela Verzella
- Department of Biotechnological and Applied Clinical Sciences (DISCAB), University of L’Aquila, 67100 L’Aquila, Italy; (D.V.); (D.C.); (F.Z.)
| | - Paola Arboretto
- Department of Immunology and Inflammation, Imperial College London, London W12 0NN, UK; (J.C.); (P.A.)
| | - Davide Vecchiotti
- Department of Biotechnological and Applied Clinical Sciences (DISCAB), University of L’Aquila, 67100 L’Aquila, Italy; (D.V.); (D.C.); (F.Z.)
| | - Daria Capece
- Department of Biotechnological and Applied Clinical Sciences (DISCAB), University of L’Aquila, 67100 L’Aquila, Italy; (D.V.); (D.C.); (F.Z.)
| | - Francesca Zazzeroni
- Department of Biotechnological and Applied Clinical Sciences (DISCAB), University of L’Aquila, 67100 L’Aquila, Italy; (D.V.); (D.C.); (F.Z.)
| | - Guido Franzoso
- Department of Immunology and Inflammation, Imperial College London, London W12 0NN, UK; (J.C.); (P.A.)
| |
Collapse
|
6
|
Chen S, Pan X, Zhang L, Cui X, Ye J. FOXK1 upregulation is correlated with tumor progression and tumor associated macrophages infiltration in renal cell carcinoma. Mol Carcinog 2024; 63:136-144. [PMID: 37818826 DOI: 10.1002/mc.23641] [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: 08/11/2023] [Revised: 09/13/2023] [Accepted: 09/15/2023] [Indexed: 10/13/2023]
Abstract
Renal cell carcinoma (RCC) is one of the most common urological cancers in adults. Forkhead box k1 (FOXK1) is a transcription factor involved in the progression of various malignant tumors. In this study, we aimed to investigate the expression and roles of FOXK1 in RCC development. Our findings revealed increased expression of FOXK1 in RCC tumor tissues and cell lines compared with normal controls. Functional assays demonstrated that knockdown of FOXK1 significantly inhibited proliferation, migration, invasion, and promoted apoptosis in RCC cells. Furthermore, FOXK1 knockdown suppressed epithelial-mesenchymal transition and Wnt signaling in RCC cells. Additionally, we observed a correlation between FOXK1 upregulation and tumor associated macrophages infiltration in RCC. These results suggest that FOXK1 acts as an oncogene in RCC and may serve as a potential therapeutic target for RCC treatment.
Collapse
Affiliation(s)
- Shaojun Chen
- Department of Urology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiuwu Pan
- Department of Urology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Liang Zhang
- Department of Urology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xingang Cui
- Department of Urology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jianqing Ye
- Department of Urology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
7
|
Hekmatirad S, Moloudizargari M, Fallah M, Rahimi A, Poortahmasebi V, Asghari MH. Cancer-associated immune cells and their modulation by melatonin. Immunopharmacol Immunotoxicol 2023; 45:788-801. [PMID: 37489565 DOI: 10.1080/08923973.2023.2239489] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 07/17/2023] [Indexed: 07/26/2023]
Abstract
OBJECTIVES Rapidly growing evidence suggests that immune cells play a key role in determining tumor progression. Tumor cells are surrounded by a microenvironment composed of different cell populations including immune cells. The cross talk between tumor cells and the neighboring microenvironment is an important factor to take into account while designing tumor therapies. Despite significant advances in immunotherapy strategies, a relatively small proportion of patients have successfully responded to them. Therefore, the search for safe and efficient drugs, which could be used alongside conventional therapies to boost the immune system against tumors, is an ongoing need. In the present work, the modulatory effects of melatonin on different components of tumor immune microenvironment are reviewed. METHODS A thorough literature review was performed in PubMed, Scopus, and Web of Science databases. All published papers in English on tumor immune microenvironment and the relevant modulatory effects of melatonin were scrutinized. RESULTS Melatonin modulates macrophage polarization and prevents M2 induction. Moreover, it prevents the conversion of fibroblasts into cancer-associated fibroblasts (CAFs) and prevents cancer cell stemness. In addition, it can affect the payload composition of tumor-derived exosomes (TEXs) and their secretion levels to favor a more effective anti-tumor immune response. Melatonin is a safe molecule that affects almost all components of the tumor immune microenvironment and prevents them from being negatively affected by the tumor. CONCLUSION Based on the effects of melatonin on normal cells, tumor cells and microenvironment components, it could be an efficient compound to be used in combination with conventional immune-targeted therapies to increase their efficacy.
Collapse
Affiliation(s)
- Shirin Hekmatirad
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Marjan Fallah
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Medicinal Plant Research Centre, Islamic Azad University, Amol, Iran
| | - Atena Rahimi
- Department of Pharmacology and Toxicology, School of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Vahdat Poortahmasebi
- Department of Bacteriology and Virology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Hossein Asghari
- Department of Pharmacology and Toxicology, School of Medicine, Babol University of Medical Sciences, Babol, Iran
| |
Collapse
|
8
|
Nakahata S, Enriquez-Vera D, Jahan MI, Sugata K, Satou Y. Understanding the Immunopathology of HTLV-1-Associated Adult T-Cell Leukemia/Lymphoma: A Comprehensive Review. Biomolecules 2023; 13:1543. [PMID: 37892225 PMCID: PMC10605031 DOI: 10.3390/biom13101543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 10/13/2023] [Accepted: 10/16/2023] [Indexed: 10/29/2023] Open
Abstract
Human T-cell leukemia virus type-1 (HTLV-1) causes adult T-cell leukemia/lymphoma (ATL). HTLV-1 carriers have a lifelong asymptomatic balance between infected cells and host antiviral immunity; however, 5-10% of carriers lose this balance and develop ATL. Coinfection with Strongyloides promotes ATL development, suggesting that the immunological status of infected individuals is a determinant of HTLV-1 pathogenicity. As CD4+ T cells play a central role in host immunity, the deregulation of their function and differentiation via HTLV-1 promotes the immune evasion of infected T cells. During ATL development, the accumulation of genetic and epigenetic alterations in key host immunity-related genes further disturbs the immunological balance. Various approaches are available for treating these abnormalities; however, hematopoietic stem cell transplantation is currently the only treatment with the potential to cure ATL. The patient's immune state may contribute to the treatment outcome. Additionally, the activity of the anti-CC chemokine receptor 4 antibody, mogamulizumab, depends on immune function, including antibody-dependent cytotoxicity. In this comprehensive review, we summarize the immunopathogenesis of HTLV-1 infection in ATL and discuss the clinical findings that should be considered when developing treatment strategies for ATL.
Collapse
Affiliation(s)
- Shingo Nakahata
- Division of HTLV-1/ATL Carcinogenesis and Therapeutics, Joint Research Center for Human Retrovirus Infection, Kagoshima University, Kagoshima 890-8544, Japan
| | - Daniel Enriquez-Vera
- Division of HTLV-1/ATL Carcinogenesis and Therapeutics, Joint Research Center for Human Retrovirus Infection, Kagoshima University, Kagoshima 890-8544, Japan
| | - M. Ishrat Jahan
- Division of Genomics and Transcriptomics, Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto 860-8556, Japan
| | - Kenji Sugata
- Division of Genomics and Transcriptomics, Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto 860-8556, Japan
| | - Yorifumi Satou
- Division of Genomics and Transcriptomics, Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto 860-8556, Japan
| |
Collapse
|
9
|
Xu Y, Li L, Yang W, Zhang K, Zhang Z, Yu C, Qiu J, Cai L, Gong Y, Zhang Z, Zhou J, Gong K. TRAF2 promotes M2-polarized tumor-associated macrophage infiltration, angiogenesis and cancer progression by inhibiting autophagy in clear cell renal cell carcinoma. J Exp Clin Cancer Res 2023; 42:159. [PMID: 37415241 DOI: 10.1186/s13046-023-02742-w] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 06/26/2023] [Indexed: 07/08/2023] Open
Abstract
BACKGROUND The management of advanced clear cell renal cell carcinoma (ccRCC) remains a major challenge in clinical practice, and the construction of more reliable prognostic prediction models and the further elucidation of key molecular mechanisms of tumor progression are topics in urgent need of in-depth investigation. METHODS We used CIBERSORT to estimate the proportion of 22 tumor-infiltrating immune cell types in the TCGA-KIRC cohort. Weighted gene co-expression network analysis, least absolute shrinkage and selection operator regression analysis were used to build risk prediction models. Expression patterns and clinical significance of TRAF2 were determined through bioinformatics analysis, real-time qPCR, Western Blot, immunohistochemistry. GSEA analysis, transmission electron microscopy, 2D/3D colony formation assay, cell migration and invasion assay, and tube-formation assay were used to investigate the underlying function and mechanism of the TRAF2/M2 macrophage/autophagy axis. RESULTS We constructed a novel prognostic prediction model based on M2 macrophage-related genes, which was identified as an accurate, independent and specific prognostic risk model for ccRCC patients. A reliable nomogram was constructed to predict 1-, 3-, and 5-year overall survival for patients with ccRCC. As one of the constituent genes of the risk model, TRAF2 was determined to be upregulated in ccRCC and associated with poor clinical prognosis. We found that TRAF2 promotes malignant progression of ccRCC by regulating macrophage polarization, migration and angiogenesis. Mechanistically, we found that TRAF2 promotes the polarization of M2 macrophages, and this chemotaxis is achieved in an autophagy-dependent pathway. Orthotopic tumor growth assay results revealed that TRAF2 plays a key role as a promotor of ccRCC growth and metastasis. CONCLUSIONS In conclusion, this risk model is highly predictive of prognostic in ccRCC patients, which is expected to promote improved treatment evaluation and comprehensive management of ccRCC. Moreover, our findings reveal that the TRAF2/M2 macrophage/autophagy axis plays a key regulatory role in the malignant progression of ccRCC, and suggest that TRAF2 is a potential novel therapeutic target for advanced ccRCC.
Collapse
Affiliation(s)
- Yawei Xu
- Department of Urology, Peking University First Hospital; Institute of Urology, Peking University; Beijing Key Laboratory of Urogenital Diseases (Male) Molecular Diagnosis and Treatment Center, National Urological Cancer Center, Beijing, 100034, China
| | - Lei Li
- Department of Urology, Peking University First Hospital; Institute of Urology, Peking University; Beijing Key Laboratory of Urogenital Diseases (Male) Molecular Diagnosis and Treatment Center, National Urological Cancer Center, Beijing, 100034, China
| | - Wuping Yang
- Department of Urology, Peking University First Hospital; Institute of Urology, Peking University; Beijing Key Laboratory of Urogenital Diseases (Male) Molecular Diagnosis and Treatment Center, National Urological Cancer Center, Beijing, 100034, China
| | - Kenan Zhang
- Department of Urology, Peking University First Hospital; Institute of Urology, Peking University; Beijing Key Laboratory of Urogenital Diseases (Male) Molecular Diagnosis and Treatment Center, National Urological Cancer Center, Beijing, 100034, China
| | - Zedan Zhang
- Department of Urology, Peking University First Hospital; Institute of Urology, Peking University; Beijing Key Laboratory of Urogenital Diseases (Male) Molecular Diagnosis and Treatment Center, National Urological Cancer Center, Beijing, 100034, China
| | - Chaojian Yu
- Department of Urology, Peking University First Hospital; Institute of Urology, Peking University; Beijing Key Laboratory of Urogenital Diseases (Male) Molecular Diagnosis and Treatment Center, National Urological Cancer Center, Beijing, 100034, China
| | - Jianhui Qiu
- Department of Urology, Peking University First Hospital; Institute of Urology, Peking University; Beijing Key Laboratory of Urogenital Diseases (Male) Molecular Diagnosis and Treatment Center, National Urological Cancer Center, Beijing, 100034, China
| | - Lin Cai
- Department of Urology, Peking University First Hospital; Institute of Urology, Peking University; Beijing Key Laboratory of Urogenital Diseases (Male) Molecular Diagnosis and Treatment Center, National Urological Cancer Center, Beijing, 100034, China
| | - Yanqing Gong
- Department of Urology, Peking University First Hospital; Institute of Urology, Peking University; Beijing Key Laboratory of Urogenital Diseases (Male) Molecular Diagnosis and Treatment Center, National Urological Cancer Center, Beijing, 100034, China
| | - Zheng Zhang
- Department of Urology, Peking University First Hospital; Institute of Urology, Peking University; Beijing Key Laboratory of Urogenital Diseases (Male) Molecular Diagnosis and Treatment Center, National Urological Cancer Center, Beijing, 100034, China
| | - Jingcheng Zhou
- Department of Urology, Peking University First Hospital; Institute of Urology, Peking University; Beijing Key Laboratory of Urogenital Diseases (Male) Molecular Diagnosis and Treatment Center, National Urological Cancer Center, Beijing, 100034, China.
| | - Kan Gong
- Department of Urology, Peking University First Hospital; Institute of Urology, Peking University; Beijing Key Laboratory of Urogenital Diseases (Male) Molecular Diagnosis and Treatment Center, National Urological Cancer Center, Beijing, 100034, China.
| |
Collapse
|
10
|
Ascenti V, Arico FM, Trimarchi R, Cicero G, Ieni A, Rossanese M, Ascenti G. Minimal Fat Content in Papillary Renal Cell Carcinoma Diagnosed with Dual-Layer Dual-Energy CT. Diagnostics (Basel) 2023; 13:diagnostics13101742. [PMID: 37238225 DOI: 10.3390/diagnostics13101742] [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: 04/18/2023] [Revised: 05/07/2023] [Accepted: 05/10/2023] [Indexed: 05/28/2023] Open
Abstract
A 56-year-old man with a previous right nephrectomy for multiple papillary renal cell carcinomas (pRCC) underwent a follow-up CT scan. Using a dual-layer dual-energy CT (dlDECT), we demonstrated the presence of a small amount of fat in a 2.5 cm pRCC that mimicked the diagnosis of angiomyolipoma (AML). Histological examination demonstrated the absence of macroscopic intratumoral adipose tissue, showing a fair amount of enlarged foam macrophages loaded with intracytoplasmic lipids. The presence of fat density in an RCC is an extremely rare occurrence in the literature. To our knowledge, this is the first description using dlDECT of a minimal amount of fat tissue in a small RCC due to the presence of tumor-associated foam macrophages. Radiologists should be aware of this possibility when characterizing a renal mass with DECT. The option of RCCs must be considered, especially in the case of masses with an aggressive character or a positive history of RCC.
Collapse
Affiliation(s)
- Velio Ascenti
- Postgraduate School of Radiodiagnostics, Policlinico Universitario, University of Milan, 20133 Milano, Italy
| | - Francesco M Arico
- Diagnostic and Interventional Radiology Unit, BIOMORF Department, University Hospital "Policlinico G. Martino", 98124 Messina, Italy
| | - Renato Trimarchi
- Diagnostic and Interventional Radiology Unit, BIOMORF Department, University Hospital "Policlinico G. Martino", 98124 Messina, Italy
| | - Giuseppe Cicero
- Diagnostic and Interventional Radiology Unit, BIOMORF Department, University Hospital "Policlinico G. Martino", 98124 Messina, Italy
| | - Antonio Ieni
- Department of Human Pathology of Adult and Evolutive Age "Gaetano Barresi"-Section of Pathological Anatomy, University of Messina, Viale Gazzi, 98125 Messina, Italy
| | - Marta Rossanese
- Gaetano Barresi Department of Human and Paediatric Pathology, Urologic Section, University of Messina, 98166 Messina, Italy
| | - Giorgio Ascenti
- Diagnostic and Interventional Radiology Unit, BIOMORF Department, University Hospital "Policlinico G. Martino", 98124 Messina, Italy
| |
Collapse
|
11
|
Shapiro DD, Dolan B, Laklouk IA, Rassi S, Lozar T, Emamekhoo H, Wentland AL, Lubner MG, Abel EJ. Understanding the Tumor Immune Microenvironment in Renal Cell Carcinoma. Cancers (Basel) 2023; 15:cancers15092500. [PMID: 37173966 PMCID: PMC10177515 DOI: 10.3390/cancers15092500] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 04/24/2023] [Accepted: 04/24/2023] [Indexed: 05/15/2023] Open
Abstract
Scientific understanding of how the immune microenvironment interacts with renal cell carcinoma (RCC) has substantially increased over the last decade as a result of research investigations and applying immunotherapies, which modulate how the immune system targets and eliminates RCC tumor cells. Clinically, immune checkpoint inhibitor therapy (ICI) has revolutionized the treatment of advanced clear cell RCC because of improved outcomes compared to targeted molecular therapies. From an immunologic perspective, RCC is particularly interesting because tumors are known to be highly inflamed, but the mechanisms underlying the inflammation of the tumor immune microenvironment are atypical and not well described. While technological advances in gene sequencing and cellular imaging have enabled precise characterization of RCC immune cell phenotypes, multiple theories have been suggested regarding the functional significance of immune infiltration in RCC progression. The purpose of this review is to describe the general concepts of the anti-tumor immune response and to provide a detailed summary of the current understanding of the immune response to RCC tumor development and progression. This article describes immune cell phenotypes that have been reported in the RCC microenvironment and discusses the application of RCC immunophenotyping to predict response to ICI therapy and patient survival.
Collapse
Affiliation(s)
- Daniel D Shapiro
- Department of Urology, The University of Wisconsin School of Medicine and Public Health, Madison, WI 53726, USA
- Division of Urology, William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA
| | - Brendan Dolan
- Department of Urology, The University of Wisconsin School of Medicine and Public Health, Madison, WI 53726, USA
| | - Israa A Laklouk
- Department of Pathology, The University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| | - Sahar Rassi
- Department of Urology, The University of Wisconsin School of Medicine and Public Health, Madison, WI 53726, USA
| | - Taja Lozar
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53726, USA
| | - Hamid Emamekhoo
- Department of Medical Oncology, The University of Wisconsin School of Medicine and Public Health, Madison, WI 53726, USA
| | - Andrew L Wentland
- Department of Radiology, The University of Wisconsin School of Medicine and Public Health, Madison, WI 53726, USA
| | - Meghan G Lubner
- Department of Radiology, The University of Wisconsin School of Medicine and Public Health, Madison, WI 53726, USA
| | - Edwin Jason Abel
- Department of Urology, The University of Wisconsin School of Medicine and Public Health, Madison, WI 53726, USA
| |
Collapse
|
12
|
Barry ST, Gabrilovich DI, Sansom OJ, Campbell AD, Morton JP. Therapeutic targeting of tumour myeloid cells. Nat Rev Cancer 2023; 23:216-237. [PMID: 36747021 DOI: 10.1038/s41568-022-00546-2] [Citation(s) in RCA: 130] [Impact Index Per Article: 65.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/21/2022] [Indexed: 02/08/2023]
Abstract
Myeloid cells are pivotal within the immunosuppressive tumour microenvironment. The accumulation of tumour-modified myeloid cells derived from monocytes or neutrophils - termed 'myeloid-derived suppressor cells' - and tumour-associated macrophages is associated with poor outcome and resistance to treatments such as chemotherapy and immune checkpoint inhibitors. Unfortunately, there has been little success in large-scale clinical trials of myeloid cell modulators, and only a few distinct strategies have been used to target suppressive myeloid cells clinically so far. Preclinical and translational studies have now elucidated specific functions for different myeloid cell subpopulations within the tumour microenvironment, revealing context-specific roles of different myeloid cell populations in disease progression and influencing response to therapy. To improve the success of myeloid cell-targeted therapies, it will be important to target tumour types and patient subsets in which myeloid cells represent the dominant driver of therapy resistance, as well as to determine the most efficacious treatment regimens and combination partners. This Review discusses what we can learn from work with the first generation of myeloid modulators and highlights recent developments in modelling context-specific roles for different myeloid cell subtypes, which can ultimately inform how to drive more successful clinical trials.
Collapse
Affiliation(s)
- Simon T Barry
- Bioscience, Early Oncology, AstraZeneca, Cambridge, UK.
| | | | - Owen J Sansom
- Cancer Research UK Beatson Institute, Glasgow, UK
- School of Cancer Sciences, University of Glasgow, Glasgow, UK
| | | | - Jennifer P Morton
- Cancer Research UK Beatson Institute, Glasgow, UK
- School of Cancer Sciences, University of Glasgow, Glasgow, UK
| |
Collapse
|
13
|
Lee JH, Hwang S, Jee B, Kim JH, Lee J, Chung JH, Song W, Sung HH, Jeon HG, Jeong BC, Seo SI, Jeon SS, Lee HM, Park SH, Kwon GY, Kang M. Fat Loss in Patients with Metastatic Clear Cell Renal Cell Carcinoma Treated with Immune Checkpoint Inhibitors. Int J Mol Sci 2023; 24:ijms24043994. [PMID: 36835404 PMCID: PMC9967473 DOI: 10.3390/ijms24043994] [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: 01/18/2023] [Revised: 02/11/2023] [Accepted: 02/14/2023] [Indexed: 02/18/2023] Open
Abstract
The purpose of this study was to determine the prognostic impact of fat loss after immune checkpoint inhibitor (ICI) treatment in patients with metastatic clear cell renal cell carcinoma (ccRCC). Data from 60 patients treated with ICI therapy for metastatic ccRCC were retrospectively analyzed. Changes in cross-sectional areas of subcutaneous fat (SF) between the pre-treatment and post-treatment abdominal computed tomography (CT) images were expressed as percentages and were divided by the interval between the CT scans to calculate ΔSF (%/month). SF loss was defined as ΔSF < -5%/month. Survival analyses for overall survival (OS) and progression-free survival (PFS) were performed. Patients with SF loss had shorter OS (median, 9.5 months vs. not reached; p < 0.001) and PFS (median, 2.6 months vs. 33.5 months; p < 0.001) than patients without SF loss. ΔSF was independently associated with OS (adjusted hazard ratio (HR), 1.49; 95% confidence interval (CI), 1.07-2.07; p = 0.020) and PFS (adjusted HR, 1.57; 95% CI, 1.17-2.12; p = 0.003), with a 5%/month decrease in SF increasing the risk of death and progression by 49% and 57%, respectively. In conclusion, Loss of SF after treatment initiation is a significant and independent poor prognostic factor for OS and PFS in patients with metastatic ccRCC who receive ICI therapy.
Collapse
Affiliation(s)
- Ji Hyun Lee
- Department of Radiology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Republic of Korea
| | - Soohyun Hwang
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Republic of Korea
| | - ByulA Jee
- Department of Urology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul 06351, Republic of Korea
| | - Jae-Hun Kim
- Department of Radiology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Republic of Korea
| | - Jihwan Lee
- Department of Urology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul 06351, Republic of Korea
| | - Jae Hoon Chung
- Department of Urology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul 06351, Republic of Korea
| | - Wan Song
- Department of Urology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul 06351, Republic of Korea
| | - Hyun Hwan Sung
- Department of Urology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul 06351, Republic of Korea
| | - Hwang Gyun Jeon
- Department of Urology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul 06351, Republic of Korea
| | - Byong Chang Jeong
- Department of Urology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul 06351, Republic of Korea
| | - Seong Il Seo
- Department of Urology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul 06351, Republic of Korea
| | - Seong Soo Jeon
- Department of Urology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul 06351, Republic of Korea
| | - Hyun Moo Lee
- Department of Urology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul 06351, Republic of Korea
| | - Se Hoon Park
- Division of Hematology-Oncology, Department of Internal Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Republic of Korea
| | - Ghee Young Kwon
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Republic of Korea
| | - Minyong Kang
- Department of Urology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul 06351, Republic of Korea
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul 06351, Republic of Korea
- Samsung Genome Institute, Samsung Medical Center, Seoul 06351, Republic of Korea
- Correspondence: ; Tel.: +82-2-3410-1138; Fax: +82-2-3410-6992
| |
Collapse
|
14
|
Dias AS, Almeida CR, Helguero LA, Duarte IF. Metabolic Reprogramming of Breast Tumor-Educated Macrophages Revealed by NMR Metabolomics. Cancers (Basel) 2023; 15:cancers15041211. [PMID: 36831553 PMCID: PMC9954003 DOI: 10.3390/cancers15041211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 02/03/2023] [Accepted: 02/09/2023] [Indexed: 02/17/2023] Open
Abstract
The metabolic crosstalk between tumor cells and tumor-associated macrophages (TAMs) has emerged as a critical contributor to tumor development and progression. In breast cancer (BC), the abundance of immune-suppressive TAMs positively correlates with poor prognosis. However, little is known about how TAMs reprogram their metabolism in the BC microenvironment. In this work, we have assessed the metabolic and phenotypic impact of incubating THP-1-derived macrophages in conditioned media (CM) from two BC cell lines cultured in normoxia/hypoxia: MDA-MB-231 cells (highly metastatic, triple-negative BC), and MCF-7 cells (less aggressive, luminal BC). The resulting tumor-educated macrophages (TEM) displayed prominent differences in their metabolic activity and composition, compared to control cells (M0), as assessed by exo- and endometabolomics. In particular, TEM turned to the utilization of extracellular pyruvate, alanine, and branched chain keto acids (BCKA), while exhibiting alterations in metabolites associated with several intracellular pathways, including polyamines catabolism (MDA-TEM), collagen degradation (mainly MCF-TEM), adenosine accumulation (mainly MDA-TEM) and lipid metabolism. Interestingly, following a second-stage incubation in fresh RPMI medium, TEM still displayed several metabolic differences compared to M0, indicating persistent reprogramming. Overall, this work provided new insights into the metabolic plasticity of TEM, revealing potentially important nutritional exchanges and immunoregulatory metabolites in the BC TME.
Collapse
Affiliation(s)
- Ana S. Dias
- CICECO—Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal
- iBiMED—Institute of Biomedicine, Department of Medical Sciences, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Catarina R. Almeida
- iBiMED—Institute of Biomedicine, Department of Medical Sciences, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Luisa A. Helguero
- iBiMED—Institute of Biomedicine, Department of Medical Sciences, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Iola F. Duarte
- CICECO—Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal
- Correspondence: ; Tel.: +351-234-401-418
| |
Collapse
|
15
|
Lauridsen KM, Hokland M, Al-Karradi S, Møller HJ, Donskov F, Andersen MN. Soluble CD163: a novel independent prognostic biomarker in patients with metastatic renal cell carcinoma. Cancer Immunol Immunother 2023; 72:461-473. [PMID: 35953682 PMCID: PMC10992473 DOI: 10.1007/s00262-022-03266-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 07/28/2022] [Indexed: 01/26/2023]
Abstract
The hemoglobin-haptoglobin scavenger receptor CD163 is present in both a membrane-bound form on monocytes and macrophages (mCD163) and a shed soluble circulating form (sCD163). CD163 is a well-described marker of M2-like tumor-associated macrophages, but in patients with metastatic renal cell carcinoma (mRCC), monocyte mCD163 and serum sCD163 levels have not previously been investigated and associated with patient overall survival (OS). Here, we report mCD163 expression on peripheral blood monocytes, as well as sCD163 serum levels, in samples from 89 patients newly diagnosed with mRCC and 20 healthy controls. We found that in mRCC patients, compared to healthy controls, monocyte mCD163 levels were reduced (P < 0.001) whereas serum sCD163 levels were increased (P = 0.004). Moreover, an inverse correlation between mCD163 and sCD163 levels (P = 0.04) was shown. In survival analyses, intermediary levels of monocyte mCD163 were associated with longest OS, compared to both lower and higher mCD163 levels, which were both associated with worse outcomes (P < 0.01). Further, higher levels of sCD163 at diagnosis were associated with poor OS in both univariate (P < 0.001) and multivariate analysis (HR = 1.28; 95%CI 1.09-1.50, P = 0.002). Importantly, stratification by low vs. high sCD163 was able to separate patients with International Metastatic RCC Database Consortium (IMDC) intermediate risk (IMDCINT) into two subgroups with different OS (P = 0.03): IMDCINT-sCD163LOW showed survival similar to IMDCFAV patients, and IMDCINT-sCD163HIGH showed survival similar to IMDCPOOR patients. Thus, baseline sCD163 is a novel independent biomarker of OS in mRCC, and using sCD163 as an add-on biomarker may improve prognostic value for patients in the heterogenous IMDC intermediate group.
Collapse
Affiliation(s)
- Kasper Munch Lauridsen
- Department of Biomedicine, Aarhus University, Høegh-Guldbergs Gade 10. Building 1115, 224, 8000, Aarhus C, Denmark
- Department of Clinical Biochemistry, Aarhus University Hospital, Aarhus, Denmark
| | - Marianne Hokland
- Department of Biomedicine, Aarhus University, Høegh-Guldbergs Gade 10. Building 1115, 224, 8000, Aarhus C, Denmark.
| | - Sinan Al-Karradi
- Department of Biomedicine, Aarhus University, Høegh-Guldbergs Gade 10. Building 1115, 224, 8000, Aarhus C, Denmark
| | - Holger Jon Møller
- Department of Clinical Biochemistry, Aarhus University Hospital, Aarhus, Denmark
| | - Frede Donskov
- Department of Oncology, Aarhus University Hospital, Aarhus, Denmark
- Department of Oncology, University Hospital of Southern Denmark, Esbjerg, Denmark
| | - Morten Nørgaard Andersen
- Department of Biomedicine, Aarhus University, Høegh-Guldbergs Gade 10. Building 1115, 224, 8000, Aarhus C, Denmark
- Department of Clinical Biochemistry, Aarhus University Hospital, Aarhus, Denmark
- Department of Hematology, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
16
|
Ding G, Wang T, Liu S, Zhou Z, Ma J, Wu J. Wiskott-Aldrich syndrome gene as a prognostic biomarker correlated with immune infiltrates in clear cell renal cell carcinoma. Front Immunol 2023; 14:1102824. [PMID: 37122750 PMCID: PMC10130519 DOI: 10.3389/fimmu.2023.1102824] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Accepted: 03/20/2023] [Indexed: 05/02/2023] Open
Abstract
Introduction The abnormal expression of the Wiskott-Aldrich syndrome protein (WASP) encoded by the Wiskott-Aldrich syndrome (WAS) gene has been implicated in tumor invasion and immune regulation. However, prognostic implications of WAS and its correlation tumor infiltrating in renal clear cell carcinoma (ccRCC) is not clear cut. Methods The correlation between WAS expression, clinicopathological variables and clinical outcomes were evaluated using The Cancer Genome Atlas (TCGA), Gene Expression Omnibus (GEO), Tumor Immune Estimation Resource (TIMER), UALCAN, Gene Expression Profiling Interaction Analysis (GEPIA), Kaplan-Meier (KM) plotter and other databases. Furthermore, we assessed the transcription expression of WAS in renal cancer tissues, various renal carcinoma cell lines and human renal tubular cells (HK2) using quantitative polymerase chain reaction (qPCR). A comprehensive analysis of multiple databases including TIMER, GEPIA, TISIDB, ESTIMATE algorithm, and CIBERSORT algorithm were performed to determine the correlation between WAS and tumor infiltrating immune cells in ccRCC. Results The results displayed an increase in WAS mRNA level in ccRCC compared to normal tissue. WAS protein level was found highly expressed in cancer tissues, particularly within renal tumor cells via the human protein atlas (HPA). Interestingly, we found that elevated WAS expression was significantly positively correlated with the infiltration of CD8+ T cells, B cells, Monocytes, Neutrophils, Macrophages, T cell regulation, NK cells, and Dendritic cells in ccRCC. Bioinformatics demonstrated a strong correlation between WAS expression and 42 immune checkpoints, including the T cell exhaustion gene PD-1, which is critical for exploring immunotherapy for ccRCC. We revealed that patients with high WAS expression were less sensitive to immunotherapy medications. Conclusion In conclusion, our study identified that WAS was a prognostic biomarker and correlated with immune infiltrates in ccRCC.
Collapse
Affiliation(s)
- Guixin Ding
- Department of Urology, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong, China
| | - Tianqi Wang
- Department of Urology, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong, China
| | - Shangjing Liu
- Department of Urology, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong, China
| | - Zhongbao Zhou
- Department of Urology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Jian Ma
- Department of Urology, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong, China
- *Correspondence: Jitao Wu, ; Jian Ma,
| | - Jitao Wu
- Department of Urology, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong, China
- *Correspondence: Jitao Wu, ; Jian Ma,
| |
Collapse
|
17
|
Najm R, Hachim MY, Kandasamy RK. Divulging a Pleiotropic Role of Succinate Receptor SUCNR1 in Renal Cell Carcinoma Microenvironment. Cancers (Basel) 2022; 14:cancers14246064. [PMID: 36551549 PMCID: PMC9776839 DOI: 10.3390/cancers14246064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 11/29/2022] [Accepted: 12/05/2022] [Indexed: 12/13/2022] Open
Abstract
The succinate receptor, SUCNR1, has been attributed to tumor progression, metastasis, and immune response modulation upon its activation via the oncometabolite succinate. Nonetheless, little is known about the prognostic relevance of SUCNR1 and its association with tumor immune infiltrates and microbiota in renal cell carcinoma (RCC). Herein, publicly available platforms including Human Protein Atlas, cBioPortal, TIMER2.0, and TISIDB were utilized to depict a divergent implication of SUCNR1 in the immune microenvironment of clear cell RCC (KIRC) and papillary RCC (KIRP); the two major subtypes of RCC. Our results showed that the SUCNR1 expression level was augmented in RCC compared to other solid cancers, yet with opposite survival rate predictions in RCC subtypes. Consequently, a higher expression level of SUCNR1 was associated with a good disease-specific survival rate (p = 5.797 × 10-5) in KIRC patients albeit a poor prognostic prediction in KIRP patients (p = 1.9282 × 10-3). Intriguingly, SUCNR1 was mainly correlated to immunomodulators and diverse immune infiltrates in KIRP. Additionally, the SUCNR1 was mostly associated with a repertoire of microbes including beneficial bacteria that likely influenced a better disease-specific survival rate in KIRC. Our findings illustrate a significant novel subtype-specific role of SUCNR1 in RCC which potentially modulates tumor immune infiltration and microbiome signature, hence altering the prognosis of cancer patients.
Collapse
Affiliation(s)
- Rania Najm
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, United Arab Emirates
| | - Mahmood Yaseen Hachim
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, United Arab Emirates
| | - Richard K. Kandasamy
- Centre of Molecular Inflammation Research (CEMIR), Department of Clinical and Molecular Medicine (IKOM), Norwegian University of Science and Technology, 7491 Trondheim, Norway
- Department of Laboratory Medicine and Pathology, Center for Individualized Medicine, Mayo Clinic, Rochester, MN 55905, USA
- Correspondence: or
| |
Collapse
|
18
|
Kerneur C, Cano CE, Olive D. Major pathways involved in macrophage polarization in cancer. Front Immunol 2022; 13:1026954. [PMID: 36325334 PMCID: PMC9618889 DOI: 10.3389/fimmu.2022.1026954] [Citation(s) in RCA: 125] [Impact Index Per Article: 41.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 09/23/2022] [Indexed: 11/16/2022] Open
Abstract
Macrophages play an important role in tissue homeostasis, tissue remodeling, immune response, and progression of cancer. Consequently, macrophages exhibit significant plasticity and change their transcriptional profile and function in response to environmental, tissue, and inflammatory stimuli resulting in pro- and anti-tumor effects. Furthermore, the categorization of tissue macrophages in inflammatory situations remains difficult; however, there is an agreement that macrophages are predominantly polarized into two different subtypes with pro- and anti-inflammatory properties, the so-called M1-like and M2-like macrophages, respectively. These two macrophage classes can be considered as the extreme borders of a continuum of many intermediate subsets. On one end, M1 are pro-inflammatory macrophages that initiate an immunological response, damage tissue integrity, and dampen tumor progression by fostering robust T and natural killer (NK) cell anti-tumoral responses. On the other end, M2 are anti-inflammatory macrophages involved in tissue remodeling and tumor growth, that promote cancer cell proliferation, invasion, tumor metastasis, angiogenesis and that participate to immune suppression. These decisive roles in tumor progression occur through the secretion of cytokines, chemokines, growth factors, and matrix metalloproteases, as well as by the expression of immune checkpoint receptors in the case of M2 macrophages. Moreover, macrophage plasticity is supported by stimuli from the Tumor Microenvironment (TME) that are relayed to the nucleus through membrane receptors and signaling pathways that result in gene expression reprogramming in macrophages, thus giving rise to different macrophage polarization outcomes. In this review, we will focus on the main signaling pathways involved in macrophage polarization that are activated upon ligand-receptor recognition and in the presence of other immunomodulatory molecules in cancer.
Collapse
Affiliation(s)
- Clément Kerneur
- ImCheck Therapeutics, Marseille, France
- Team Immunity and Cancer, Centre de Recherche en Cancérologie de Marseille (CRCM), Inserm U1068, CNRS UMR7258, Institut Paoli Calmettes, Marseille, France
- *Correspondence: Clément Kerneur, ; Carla E. Cano, ; Daniel Olive,
| | - Carla E. Cano
- ImCheck Therapeutics, Marseille, France
- *Correspondence: Clément Kerneur, ; Carla E. Cano, ; Daniel Olive,
| | - Daniel Olive
- Team Immunity and Cancer, Centre de Recherche en Cancérologie de Marseille (CRCM), Inserm U1068, CNRS UMR7258, Institut Paoli Calmettes, Marseille, France
- *Correspondence: Clément Kerneur, ; Carla E. Cano, ; Daniel Olive,
| |
Collapse
|
19
|
Li X, Dong H, Chen L, Wang Y, Hao Z, Zhang Y, Jiao Y, Zhao Z, Peng X, Zhan X. Identification of N7-methylguanosine related subtypes and construction of prognostic model in gastric cancer. Front Immunol 2022; 13:984149. [PMID: 36300121 PMCID: PMC9589367 DOI: 10.3389/fimmu.2022.984149] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 09/26/2022] [Indexed: 12/24/2022] Open
Abstract
Background N7-methylguanosine (m7G), one of the most common post-transcriptional modifications, can be present in tRNA, mRNA, and miRNA to mediate the progression of various tumors. However, the possible role of m7G in gastric cancer (GC) is still unknown. Materials and Methods In this study, SNVs (single nucleotide variations), CNVs (copy number variations), and methylation of m7G-related genes (m7GRGs) were analyzed. The relationship between them and the expression of m7GRGs and prognosis of GC patients was explored. Based on 13 prognostic-related m7GRGs, 567 GC samples were classified into three subtypes using the ConsensusClusterPlus package. we compared survival status, clinical traits, immune cell infiltration, immune checkpoints, tumor microenvironment (TME), tumor immune dysfunction and exclusion (TIDE), and potential biological pathways among the three subtypes. Then, patients were again grouped into different genetic subtypes based on the DEGs among the three subtypes. In addition, a prognostic m7GRG_Score was constructed using five risk genes applicable to patients of any age, gender and stage. We also assessed tumor mutational burden (TMB), microsatellite instability (MSI), cancer stem cell (CSC) index, sensitivity of antineoplastic drugs, efficacy of anti-PD-1 and anti-CTLA4 immunotherapy between high and low m7GRG_Score groups. Finally, we established a nomogram based on m7GRG_Score and tumor stage to enhance the clinical application of the model. miRNAs and lncRNAs that could regulate expression of risk genes were searched. Results SNVs, CNVs, and methylation of m7GRGs were associated with m7GRGs expression. However, they did not significantly affect the survival of GC patients. Our results also confirmed that patients in subtypes B and C and low m7GRG_Score groups had longer survival time, better clinical stage, more immune cell infiltration, fewer immune escape and dysfunction compared to subtype A and high m7GRG_Score groups. A low m7GRG_score was featured with increased microsatellite instability-high (MSI-H), TMB, and efficacy of immunotherapy. Conclusion The m7GRG_Score model may become a beneficial tool for predicting prognosis and guiding personalized treatment in GC patients. These findings will improve our knowledge of m7G in GC and provide new methods for more effective treatment strategies.
Collapse
Affiliation(s)
- Xiaoxiao Li
- Department of Oncology, Changhai Hospital, Naval Military Medical University, Shanghai, China
| | - Hao Dong
- Department of Gastrointestinal Nutrition and Hernia Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Ling Chen
- Department of Oncology, Changhai Hospital, Naval Military Medical University, Shanghai, China
| | - Yujie Wang
- Department of Oncology, Changhai Hospital, Naval Military Medical University, Shanghai, China
| | - Zhibin Hao
- Department of Oncology, Changhai Hospital, Naval Military Medical University, Shanghai, China
| | - Yingyi Zhang
- Department of Oncology, Changhai Hospital, Naval Military Medical University, Shanghai, China
| | - Yuan Jiao
- Department of Oncology, Changhai Hospital, Naval Military Medical University, Shanghai, China
| | - Zhiyue Zhao
- Department of Oncology, Changhai Hospital, Naval Military Medical University, Shanghai, China
| | - Xiaobo Peng
- Department of Oncology, Changhai Hospital, Naval Military Medical University, Shanghai, China
- *Correspondence: Xiaobo Peng, ; Xianbao Zhan,
| | - Xianbao Zhan
- Department of Oncology, Changhai Hospital, Naval Military Medical University, Shanghai, China
- *Correspondence: Xiaobo Peng, ; Xianbao Zhan,
| |
Collapse
|
20
|
Kim JH, Kim GH, Ryu YM, Kim SY, Kim HD, Yoon SK, Cho Y, Lee JL. Clinical implications of the tumor microenvironment using multiplexed immunohistochemistry in patients with advanced or metastatic renal cell carcinoma treated with nivolumab plus ipilimumab. Front Oncol 2022; 12:969569. [PMID: 36237314 PMCID: PMC9552830 DOI: 10.3389/fonc.2022.969569] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 09/13/2022] [Indexed: 11/13/2022] Open
Abstract
Purpose Immune checkpoint inhibitors (ICIs) such as nivolumab and ipilimumab (N/I) are important treatment options for advanced renal cell carcinoma (RCC). The tumor microenvironment (TME) in these ICI-treated patients is largely unknown. Methods Twenty-four patients treated with N/I between July 2015 and June 2020 were analyzed. Multiplexed immunohistochemistry (mIHC) was conducted to define the TME, including various T cell subsets, B cells, macrophages, and dendritic cells. Results The median age of the study patients was 61 years (range, 39-80) and 75.0% of these cases were men. The objective response rate with N/I was 50.0%. The densities of the CD8+ cytotoxic T cells (P=0.005), specifically CD137+ CD8+ T cells (P=0.017), Foxp3- CD4+ helper T cells (P=0.003), Foxp3+ CD4+ regulatory T cells (P=0.045), CD68+ CD206- M1 macrophages (P=0.008), and CD68+ CD206+ M2 macrophages (P=0.021) were significantly higher in the treatment responders. At a median follow-up duration of 24.7 months, the median progression-free survival (PFS) was 11.6 months. The high densities (≥median) of Foxp3- CD4+ helper T cells (P=0.016) and CD68+ CD206- M1 macrophages (P=0.008) were significantly associated with better PFS, and the density of CD137+ CD8+ cytotoxic T cells (P=0.079) was marginally associated with better PFS. After multivariate analysis, the higher density of Foxp3- CD4+ helper T cells was independently associated with better PFS (hazard ratio 0.19; P=0.016). Conclusion The properties and clinical implications of the TME properties in RCC indicate that Foxp3- CD4+ helper T cells, M1 macrophages, and CD137+ CD8+ T cells are potential predictive biomarkers and treatment targets.
Collapse
Affiliation(s)
- Jwa Hoon Kim
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
- Division of Oncology, Department of Internal Medicine, Korea University Anam Hospital, Korea University College of Medicine, Seoul, South Korea
| | - Gi Hwan Kim
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Yeon-Mi Ryu
- Department of Convergence Medicine, University of Ulsan College of Medicine, Asan Medical Center, Seoul, South Korea
| | - Sang-Yeob Kim
- Department of Convergence Medicine, University of Ulsan College of Medicine, Asan Medical Center, Seoul, South Korea
| | - Hyung-Don Kim
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Shin Kyo Yoon
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Yong Mee Cho
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Jae Lyun Lee
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| |
Collapse
|
21
|
Nguyen TN, Nguyen-Tran HH, Chen CY, Hsu T. IL6 and CCL18 Mediate Cross-talk between VHL-Deficient Kidney Cells and Macrophages during Development of Renal Cell Carcinoma. Cancer Res 2022; 82:2716-2733. [PMID: 35666812 PMCID: PMC9662868 DOI: 10.1158/0008-5472.can-21-3749] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 04/18/2022] [Accepted: 05/25/2022] [Indexed: 01/07/2023]
Abstract
Loss of the von Hippel-Lindau (VHL) tumor suppressor gene function accounts for 70% to 80% of all clear-cell renal cell carcinoma (ccRCC) cases, the most prevalent form of RCC. Accumulating evidence has indicated that ccRCC arises from sites of chronic inflammation, yet how ccRCC tumor cells interact with immune components of the microenvironment has not been fully elucidated. In this study, we used unbiased proteomic and genomic analyses on components of the tumor microenvironment under different conditions, identifying the molecular and cellular mechanisms that underlie the cross-talk between VHL-deficient kidney tubule cells and macrophages. In vitro and in a Vhlh conditional knockout mouse model, VHL-deficient noncancerous kidney epithelial cells, representing the early stage of ccRCC initiation, secreted IL6 that induced macrophage infiltration and polarization toward the protumorigenic M2 phenotype. Activated human macrophages secreted CCL18 and TGFβ1 to stimulate epithelial-to-mesenchymal transition (EMT) of the kidney tubule cells. Treatment with IL6-neutralizing antibody rescued inflammatory, proliferative, and EMT phenotypes of kidney epithelial cells in Vhlh conditional knockout mice. Furthermore, in a human ccRCC xenograft model, exogenous human primary or cultured macrophages significantly promoted primary tumor growth and metastasis in a CCL18-dependent manner. These findings identify specific factors involved in reciprocal cross-talk between tumor cells and immune components in the microenvironment, thus providing an avenue for early intervention in ccRCC. SIGNIFICANCE The identification of VHL-deficient kidney tubule cell cross-talk with macrophages regulated by IL6 and CCL18 reveals potential targets for the prevention and treatment of ccRCC.
Collapse
Affiliation(s)
- Thi-Ngoc Nguyen
- Department of Biomedical Sciences and Engineering, National Central University, Taoyuan City, Taiwan, Republic of China.,Graduate Institute of Biomedical Sciences, China Medical University-Taiwan, Taichung, Taiwan, Republic of China
| | - Hieu-Huy Nguyen-Tran
- Department of Biomedical Sciences and Engineering, National Central University, Taoyuan City, Taiwan, Republic of China.,Graduate Institute of Biomedical Sciences, China Medical University-Taiwan, Taichung, Taiwan, Republic of China
| | - Chen-Yun Chen
- Department of Biomedical Sciences and Engineering, National Central University, Taoyuan City, Taiwan, Republic of China
| | - Tien Hsu
- Department of Biomedical Sciences and Engineering, National Central University, Taoyuan City, Taiwan, Republic of China.,Graduate Institute of Biomedical Sciences, China Medical University-Taiwan, Taichung, Taiwan, Republic of China.,Corresponding Author: Tien Hsu, Graduate Institute of Biomedical Sciences, China Medical University-Taiwan, No. 91, Hsueh-Shih Rd., Taichung, Taiwan 40402, Republic of China. Phone: 220-53366-8210; E-mail:
| |
Collapse
|
22
|
Li X, Chen L, Peng X, Zhan X. Progress of tumor-associated macrophages in the epithelial-mesenchymal transition of tumor. Front Oncol 2022; 12:911410. [PMID: 35965509 PMCID: PMC9366252 DOI: 10.3389/fonc.2022.911410] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Accepted: 07/07/2022] [Indexed: 11/29/2022] Open
Abstract
As a significant public health problem with high morbidity and mortality worldwide, tumor is one of the major diseases endangering human life. Moreover, metastasis is the most important contributor to the death of tumor patients. Epithelial-mesenchymal transition (EMT) is an essential biological process in developing primary tumors to metastasis. It underlies tumor progression and metastasis by inducing a series of alterations in tumor cells that confer the ability to move and migrate. Tumor-associated macrophages (TAMs) are one of the primary infiltrating immune cells in the tumor microenvironment, and they play an indispensable role in the EMT process of tumor cells by interacting with tumor cells. With the increasing clarity of the relationship between TAMs and EMT and tumor metastasis, targeting TAMs and EMT processes is emerging as a promising target for developing new cancer therapies. Therefore, this paper reviews the recent research progress of tumor-associated macrophages in tumor epithelial-mesenchymal transition and briefly discusses the current anti-tumor therapies targeting TAMs and EMT processes.
Collapse
Affiliation(s)
| | | | - Xiaobo Peng
- *Correspondence: Xiaobo Peng, ; Xianbao Zhan,
| | | |
Collapse
|
23
|
Ferician AM, Ferician OC, Cumpanas AD, Berzava PL, Nesiu A, Barmayoun A, Cimpean AM. Heterogeneity of Platelet Derived Growth Factor Pathway Gene Expression Profile Defines Three Distinct Subgroups of Renal Cell Carcinomas. Cancer Genomics Proteomics 2022; 19:477-489. [PMID: 35732321 PMCID: PMC9247877 DOI: 10.21873/cgp.20334] [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: 03/18/2022] [Revised: 04/18/2022] [Accepted: 05/11/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND/AIM We previously described four different vascular patterns (reticular, diffuse, fasciculate, and trabecular) in renal cell carcinoma (RCC) suggesting an early and heterogeneous acquisition of perivascular cells most probably due to a particular PDGF pathway gene expression profile. The aim of the study was to study PDGF pathway gene expression profiles, separately for each vascular pattern. MATERIALS AND METHODS TaqMan assay for the PDGF pathway was performed on twelve cases of ccRCC previously evaluated by histopathology, immunohistochemistry, and RNAscope. Gene expression profile was correlated with grade, invasion, vascular patterns, and VEGF. RESULTS PIK3C3 and SLC9A3 genes were overexpressed in all vascular patterns, but they were significantly correlated with high VEGF mRNA in the reticular and diffuse pattern. STAT1, JAK2, SHC2, SRF and CHUK (IKK) were exclusively overexpressed in cases with diffuse vascular pattern. SLC9A3, CHUK and STAT3 were overexpressed in G2 tumors. CONCLUSION Three ccRCC subgroups were defined: 1) PIK3C3 (VSP34)/SLC9A3 which may be proper for anti PIK3C3 inhibitors; 2) VEGFhigh subgroup where association of anti VEGF may be a benefit and 3) JAK2/STAT1 subgroup, potentially being eligible for anti JAK/STAT therapy associated with IKK inhibitors.
Collapse
Affiliation(s)
- Adela Maria Ferician
- Department of Orthopedy and Traumatology/Urology, Victor Babes University of Medicine and Pharmacy, Timisoara, Romania
| | - Ovidiu Catalin Ferician
- Department of Orthopedy and Traumatology/Urology, Victor Babes University of Medicine and Pharmacy, Timisoara, Romania;
| | - Andrei Dragos Cumpanas
- Department of Microscopic Morphology/Histology, Victor Babes University of Medicine and Pharmacy, Timisoara, Romania
- Angiogenesis Research Center, Victor Babes University of Medicine and Pharmacy, Timisoara, Romania
| | - Patricia Lorena Berzava
- Department of Microscopic Morphology/Histology, Victor Babes University of Medicine and Pharmacy, Timisoara, Romania
- Angiogenesis Research Center, Victor Babes University of Medicine and Pharmacy, Timisoara, Romania
| | - Alexandru Nesiu
- Department of Urology, Faculty of Medicine "Vasile Goldiş" Western University, Arad, Romania
| | - Ariana Barmayoun
- Klinik fur Psychiatrie, Psychosomatik und Psychotherapie, Uniklinikum Frankfurt, Frankfurt, Germany
| | - Anca Maria Cimpean
- Department of Microscopic Morphology/Histology, Victor Babes University of Medicine and Pharmacy, Timisoara, Romania
- Angiogenesis Research Center, Victor Babes University of Medicine and Pharmacy, Timisoara, Romania
- Center of Expertise for Rare Vascular Disease in Children, Emergency Hospital for Children Louis Turcanu, Timisoara, Romania
| |
Collapse
|
24
|
Lee MG, Lee YK, Huang SC, Chang CL, Ko CY, Lee WC, Chen TY, Tzou SJ, Huang CY, Tai MH, Lin YW, Kung ML, Tsai MC, Chen YL, Chang YC, Wen ZH, Huang CC, Chu TH. DLK2 Acts as a Potential Prognostic Biomarker for Clear Cell Renal Cell Carcinoma Based on Bioinformatics Analysis. Genes (Basel) 2022; 13:genes13040629. [PMID: 35456435 PMCID: PMC9030291 DOI: 10.3390/genes13040629] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 03/26/2022] [Accepted: 03/28/2022] [Indexed: 02/07/2023] Open
Abstract
Clear cell renal cell carcinoma (ccRCC) is the most common RCC subtype with a high mortality. It has been reported that delta-like 1 homologue (DLK1) participates in the tumor microenvironmental remodeling of ccRCC, but the relationship between delta-like 2 homologue (DLK2, a DLK1 homologue) and ccRCC is still unclear. Thus, this study aims to investigate the role of DLK2 in the biological function and disease prognosis of ccRCC using bioinformatics analysis. The TNMplot database showed that DLK2 was upregulated in ccRCC tissues. From the UALCAN analysis, the overexpression of DLK2 was associated with advanced stage and high grade in ccRCC. Moreover, the Kaplan-Meier plotter (KM Plotter) database showed that DLK2 upregulation was associated with poor survival outcome in ccRCC. By the LinkedOmics analysis, DLK2 signaling may participated in the modulation of ccRCC extracellular matrix (ECM), cell metabolism, ribosome biogenesis, TGF-β signaling and Notch pathway. Besides, Tumor Immune Estimation Resource (TIMER) analysis showed that the macrophage and CD8+ T cell infiltrations were associated with good prognosis in ccRCC patients. Finally, DLK2 overexpression was associated with the reduced macrophage recruitments and the M1–M2 polarization of macrophage in ccRCC tissues. Together, DLK2 may acts as a novel biomarker, even therapeutic target in ccRCC. However, this study lacks experimental validation, and further studies are required to support this viewpoint.
Collapse
Affiliation(s)
- Man-Gang Lee
- Department of Surgery, Division of Urology, Kaohsiung Armed Forces General Hospital, Kaohsiung 80284, Taiwan;
- Department of Surgery, Division of Urology, Zuoying Branch of Kaohsiung Armed Forces General Hospital, Kaohsiung 81342, Taiwan
| | - Yung-Kuo Lee
- Medical Laboratory, Medical Education and Research Center, Kaohsiung Armed Forces General Hospital, Kaohsiung 80284, Taiwan;
| | - Shih-Chung Huang
- Department of Internal Medicine, Division of Cardiology, Kaohsiung Armed Forces General Hospital, Kaohsiung 80284, Taiwan;
- Department of Internal Medicine, Division of Cardiology, Tri-Service General Hospital, National Defense Medical Center, Taipei 11490, Taiwan
- Institute of Medical Science and Technology, National Sun Yat-sen University, Kaohsiung 80424, Taiwan; (C.-L.C.); (C.-Y.K.); (S.-J.T.)
| | - Chen-Lin Chang
- Institute of Medical Science and Technology, National Sun Yat-sen University, Kaohsiung 80424, Taiwan; (C.-L.C.); (C.-Y.K.); (S.-J.T.)
- Department of Psychiatry, Kaohsiung Armed Forces General Hospital, Kaohsiung 80284, Taiwan
| | - Chou-Yuan Ko
- Institute of Medical Science and Technology, National Sun Yat-sen University, Kaohsiung 80424, Taiwan; (C.-L.C.); (C.-Y.K.); (S.-J.T.)
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, Kaohsiung Armed Forces General Hospital, Kaohsiung 80284, Taiwan
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, Tri-Service General Hospital, National Defense Medical Center, Taipei 11490, Taiwan
| | - Wen-Chin Lee
- Department of Internal Medicine, Division of Nephrology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan;
| | - Tung-Yuan Chen
- Department of Surgery, Division of Colorectal Surgery, Kaohsiung Armed Forces General Hospital, Kaohsiung 80284, Taiwan;
| | - Shiow-Jyu Tzou
- Institute of Medical Science and Technology, National Sun Yat-sen University, Kaohsiung 80424, Taiwan; (C.-L.C.); (C.-Y.K.); (S.-J.T.)
- Department of Nursing, Kaohsiung Armed Forces General Hospital, Kaohsiung 80284, Taiwan
| | - Cheng-Yi Huang
- Institute of Biomedical Sciences, National Sun Yat-sen University, Kaohsiung 80424, Taiwan; (C.-Y.H.); (M.-H.T.)
- Department of Pathology, Kaohsiung Armed Forces General Hospital, Kaohsiung 80284, Taiwan
| | - Ming-Hong Tai
- Institute of Biomedical Sciences, National Sun Yat-sen University, Kaohsiung 80424, Taiwan; (C.-Y.H.); (M.-H.T.)
- Center for Neuroscience, National Sun Yat-sen University, Kaohsiung 80424, Taiwan
| | - Yu-Wei Lin
- Department of Radiation Oncology, Kaohsiung Veterans General Hospital, Kaohsiung 813414, Taiwan;
| | - Mei-Lang Kung
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung 813414, Taiwan;
| | - Ming-Chao Tsai
- Department of Internal Medicine, Division of Hepato-Gastroenterology, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan;
| | - Yung-Lung Chen
- Section of Cardiology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan;
| | - Yi-Chen Chang
- Doctoral Degree Program in Marine Biotechnology, National Sun Yat-sen University and Academia Sinica, Kaohsiung 80424, Taiwan;
| | - Zhi-Hong Wen
- Department of Marine Biotechnology and Resources, Asia-Pacific Ocean Research Center, National Sun Yat-sen University, Kaohsiung 80424, Taiwan;
| | - Chao-Cheng Huang
- Department of Pathology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan
- Biobank and Tissue Bank, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan
- Correspondence: (C.-C.H.); (T.-H.C.); Tel.: +886-7-731-7123 (ext. 2557) (C.-C.H.); +886-7-749-6751 (ext. 726201) (T.-H.C.)
| | - Tian-Huei Chu
- Medical Laboratory, Medical Education and Research Center, Kaohsiung Armed Forces General Hospital, Kaohsiung 80284, Taiwan;
- Correspondence: (C.-C.H.); (T.-H.C.); Tel.: +886-7-731-7123 (ext. 2557) (C.-C.H.); +886-7-749-6751 (ext. 726201) (T.-H.C.)
| |
Collapse
|
25
|
Song C, Chen J, Zhang C, Dong D. An Integrated Pan-Cancer Analysis of ADAMTS12 and Its Potential Implications in Pancreatic Adenocarcinoma. Front Oncol 2022; 12:849717. [PMID: 35280819 PMCID: PMC8904364 DOI: 10.3389/fonc.2022.849717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 01/31/2022] [Indexed: 11/23/2022] Open
Abstract
Background A Disintegrin and Metallopeptidase with Thrombospondin Type 1 Motif 12 (ADAMTS12), a member of the ADAMTS family of multidomain extracellular protease enzymes, is involved in the progression of many tumors. However, a pan-cancer analysis of this gene has not yet been performed. Its role in pancreatic adenocarcinoma (PAAD) also remains unclear. Methods The Cancer Genome Atlas (TCGA) and Genotype-Tissue Expression data (GTEx) databases were used to analyze ADAMTS12 expression in pan-cancer. We assessed the expression, clinical characteristics, prognostic significance, copy number alteration, methylation, and mutation of ADAMTS12 and its correlation with the tumor immune microenvironment. qRT-PCR and immunohistochemistry assays were also performed to validate the expression of ADAMTS12 in PAAD. Results Through bioinformatics analysis and preliminary experimental verification, ADAMTS12 was found to be substantially overexpressed in PAAD. High expression level of ADAMTS12 was correlated with worse survival rates in patients with PAAD and high infiltration levels of tumor-associated macrophages, cancer-associated fibroblasts, immune checkpoint proteins, and immunosuppressive genes. Conclusion Our findings suggest ADAMTS12 as a potential prognostic biomarker in PAAD. Elevated ADAMTS12 expression may also indicate an immunosuppressive microenvironment.
Collapse
Affiliation(s)
- Caiyun Song
- Department of Gastroenterology, Wenzhou People's Hospital, Wenzhou, China
| | - Jionghuang Chen
- Zhejiang Engineering Research Center of Cognitive Healthcare, School of Medicine, Zhejiang University, Hangzhou, China
| | - Chaolei Zhang
- Zhejiang Engineering Research Center of Cognitive Healthcare, School of Medicine, Zhejiang University, Hangzhou, China
| | - Dapeng Dong
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
26
|
Xie Y, Chen Z, Zhong Q, Zheng Z, Chen Y, Shangguan W, Zhang Y, Yang J, Zhu D, Xie W. M2 macrophages secrete CXCL13 to promote renal cell carcinoma migration, invasion, and EMT. Cancer Cell Int 2021; 21:677. [PMID: 34922542 PMCID: PMC8684162 DOI: 10.1186/s12935-021-02381-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 11/30/2021] [Indexed: 12/24/2022] Open
Abstract
Objective M2 macrophages are associated with a poor prognosis in a variety of malignancies. There are, however, few relevant investigations in clear cell renal cell carcinoma (ccRCC). Methods The expression of M2 macrophages in ccRCC tissues was first discovered using immunohistochemistry in this study. Then, M2 macrophages were created in vitro to see how they affected the proliferation, migration, invasion, and EMT of ccRCC cells. Using qPCR and prognostic analysis identifies important chemokine. Antibody neutralization tests confirmed the chemokine’s involvement and function. Pathway inhibitors confirmed the main pathway of M2 macrophages in ccRCC. Finally, qPCR and IHC were used to confirm the expression of chemokine receptors in ccRCC tissues. Results The presence of M2 macrophages was linked to a poor outcome in ccRCC. M2 macrophages enhanced the proliferation, migration, invasion, and EMT of ccRCC lines in vitro. CXCL13 was identified as the main chemokine by prognostic analysis and qPCR tests. CXCL13 neutralizing antibodies can inhibit the stimulation of M2 macrophages in ccRCC lines’ proliferation, migration, invasion, and EMT. M2 macrophages and CXCL13 may activate the Akt pathway in ccRCC lines, and Akt inhibitors decrease ccRCC lines proliferation, migration, invasion, and EMT. CXCR5 expression is a poor prognostic factor for renal cell carcinoma, according to qPCR and immunohistochemistry. In vivo experiments further proved that CXCL13 secreted by M2 macrophages can promote tumor proliferation. Conclusions M2 macrophages in the immunological milieu secrete CXCL13, which promotes ccRCC proliferation, migration, invasion, and EMT. Our findings contribute to a better understanding of the function of the tumor microenvironment in the incidence and progression of ccRCC, and they may point to novel therapeutic targets for ccRCC. Supplementary Information The online version contains supplementary material available at 10.1186/s12935-021-02381-1.
Collapse
|