1
|
Bai H, Li Z, Weng Y, Cui F, Chen W. Integrated analysis of single-cell RNA-seq and bulk RNA-seq revealed key genes for bone metastasis and chemoresistance in prostate cancer. Genes Genomics 2024:10.1007/s13258-024-01575-x. [PMID: 39395905 DOI: 10.1007/s13258-024-01575-x] [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: 04/06/2024] [Accepted: 09/24/2024] [Indexed: 10/14/2024]
Abstract
BACKGROUND Prostate cancer (PCa) is a serious malignancy. The main causes of PCa aggravation and death are unexplained resistance to chemotherapy and bone metastases. OBJECTIVE This study aimed to investigate the molecular mechanisms associated with the dynamic processes of progression, bone metastasis, and chemoresistance in PCa. METHODS Through comprehensive analysis of single-cell RNA sequencing (scRNA-seq) data, Gene Expression Omnibus (GEO) tumor progression and metastasis-related genes were identified. These genes were subjected to lasso regression modeling using the Cancer Genome Atlas (TCGA) database. Tartrate-resistant acid phosphatase (TRAP) staining and real-time quantitative PCR (RT-qPCR) were used to evaluate osteoclast differentiation. CellMiner was used to confirm the effect of LDHA on chemoresistance. Finally, the relationship between LDHA and chemoresistance was verified using doxorubicin-resistant PCa cell lines. RESULTS 7928 genes were identified as genes related to tumor progression and metastasis. Of these, 7 genes were found to be associated with PCa prognosis. The scRNA-seq and TCGA data showed that the expression of LDHA was higher in tumors and associated with poor prognosis of PCa. In addition, upregulation of LDHA in PCa cells induces osteoclast differentiation. Additionally, high LDHA expression was associated with resistance to Epirubicin, Elliptinium acetate, and doxorubicin. Cellular experiments demonstrated that LDHA knockdown inhibited doxorubicin resistance in PCa cells. CONCLUSIONS LDHA may play a potential contributory role in PCa initiation and development, bone metastasis, and chemoresistance. LDHA is a key target for the treatment of PCa.
Collapse
Affiliation(s)
- Hongai Bai
- Clinical Trial Department, Wenzhou Central Hospital, Wenzhou, People's Republic of China
| | - Zhenyue Li
- Pharmacy Department, Wenzhou Central Hospital, Wenzhou, People's Republic of China
| | - Yueyue Weng
- Pharmacy Department, Wenzhou Central Hospital, Wenzhou, People's Republic of China
| | - Facai Cui
- Department of Clinical Laboratory, Henan provincial people's hospital, The people's hospital of Zhengzhou University, Zhengzhou, People's Republic of China
| | - Wenpu Chen
- Urology Surgery, Jinshan Branch of Shanghai Sixth People's Hospital, Shanghai, People's Republic of China.
| |
Collapse
|
2
|
Fan Y, Meng Y, Hu X, Liu J, Qin X. Uncovering novel mechanisms of chitinase-3-like protein 1 in driving inflammation-associated cancers. Cancer Cell Int 2024; 24:268. [PMID: 39068486 PMCID: PMC11282867 DOI: 10.1186/s12935-024-03425-y] [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: 02/25/2024] [Accepted: 07/01/2024] [Indexed: 07/30/2024] Open
Abstract
Chitinase-3-like protein 1 (CHI3L1) is a secreted glycoprotein that is induced and regulated by multiple factors during inflammation in enteritis, pneumonia, asthma, arthritis, and other diseases. It is associated with the deterioration of the inflammatory environment in tissues with chronic inflammation caused by microbial infection or autoimmune diseases. The expression of CHI3L1 expression is upregulated in several malignant tumors, underscoring the crucial role of chronic inflammation in the initiation and progression of cancer. While the precise mechanism connecting inflammation and cancer is unclear, the involvement of CHI3L1 is involved in chronic inflammation, suggesting its role as a contributing factor to in the link between inflammation and cancer. CHI3L1 can aggravate DNA oxidative damage, induce the cancerous phenotype, promote the development of a tumor inflammatory environment and angiogenesis, inhibit immune cells, and promote cancer cell growth, invasion, and migration. Furthermore, it participates in the initiation of cancer progression and metastasis by binding with transmembrane receptors to mediate intracellular signal transduction. Based on the current research on CHI3L1, we explore introduce the receptors that interact with CHI3L1 along with the signaling pathways that may be triggered during chronic inflammation to enhance tumorigenesis and progression. In the last section of the article, we provide a brief overview of anti-inflammatory therapies that target CHI3L1.
Collapse
Affiliation(s)
- Yan Fan
- Department of Laboratory Medicine, Liaoning Clinical Research Center for Laboratory Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning, 110122, China
- Liaoning Clinical Research Center for Laboratory Medicine, Shenyang, Liaoning Province, China
| | - Yuan Meng
- Department of Laboratory Medicine, Liaoning Clinical Research Center for Laboratory Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning, 110122, China
- Liaoning Clinical Research Center for Laboratory Medicine, Shenyang, Liaoning Province, China
| | - Xingwei Hu
- Department of Laboratory Medicine, Liaoning Clinical Research Center for Laboratory Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning, 110122, China
- Liaoning Clinical Research Center for Laboratory Medicine, Shenyang, Liaoning Province, China
| | - Jianhua Liu
- Department of Laboratory Medicine, Liaoning Clinical Research Center for Laboratory Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning, 110122, China
- Liaoning Clinical Research Center for Laboratory Medicine, Shenyang, Liaoning Province, China
| | - Xiaosong Qin
- Department of Laboratory Medicine, Liaoning Clinical Research Center for Laboratory Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning, 110122, China.
- Liaoning Clinical Research Center for Laboratory Medicine, Shenyang, Liaoning Province, China.
| |
Collapse
|
3
|
Wang Y, Li C, He J, Zhao Q, Zhou Y, Sun H, Zhu H, Ding B, Ren M. Multi-omics analysis and experimental validation of the value of monocyte-associated features in prostate cancer prognosis and immunotherapy. Front Immunol 2024; 15:1426474. [PMID: 38947325 PMCID: PMC11211272 DOI: 10.3389/fimmu.2024.1426474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 05/31/2024] [Indexed: 07/02/2024] Open
Abstract
Background Monocytes play a critical role in tumor initiation and progression, with their impact on prostate adenocarcinoma (PRAD) not yet fully understood. This study aimed to identify key monocyte-related genes and elucidate their mechanisms in PRAD. Method Utilizing the TCGA-PRAD dataset, immune cell infiltration levels were assessed using CIBERSORT, and their correlation with patient prognosis was analyzed. The WGCNA method pinpointed 14 crucial monocyte-related genes. A diagnostic model focused on monocytes was developed using a combination of machine learning algorithms, while a prognostic model was created using the LASSO algorithm, both of which were validated. Random forest and gradient boosting machine singled out CCNA2 as the most significant gene related to prognosis in monocytes, with its function further investigated through gene enrichment analysis. Mendelian randomization analysis of the association of HLA-DR high-expressing monocytes with PRAD. Molecular docking was employed to assess the binding affinity of CCNA2 with targeted drugs for PRAD, and experimental validation confirmed the expression and prognostic value of CCNA2 in PRAD. Result Based on the identification of 14 monocyte-related genes by WGCNA, we developed a diagnostic model for PRAD using a combination of multiple machine learning algorithms. Additionally, we constructed a prognostic model using the LASSO algorithm, both of which demonstrated excellent predictive capabilities. Analysis with random forest and gradient boosting machine algorithms further supported the potential prognostic value of CCNA2 in PRAD. Gene enrichment analysis revealed the association of CCNA2 with the regulation of cell cycle and cellular senescence in PRAD. Mendelian randomization analysis confirmed that monocytes expressing high levels of HLA-DR may promote PRAD. Molecular docking results suggested a strong affinity of CCNA2 for drugs targeting PRAD. Furthermore, immunohistochemistry experiments validated the upregulation of CCNA2 expression in PRAD and its correlation with patient prognosis. Conclusion Our findings offer new insights into monocyte heterogeneity and its role in PRAD. Furthermore, CCNA2 holds potential as a novel targeted drug for PRAD.
Collapse
Affiliation(s)
- YaXuan Wang
- Department of Urology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Chao Li
- Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University Munich, Munich, Germany
| | - JiaXing He
- Department of Urology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - QingYun Zhao
- Department of Urology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yu Zhou
- Department of Urology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - HaoDong Sun
- Department of Urology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - HaiXia Zhu
- Clinical Laboratory, Tumor Hospital Affiliated to Nantong University, Nantong, China
| | - BeiChen Ding
- Department of Urology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - MingHua Ren
- Department of Urology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
4
|
Galván GC, Macias E, Sanders S, Ramirez-Torres A, Stock S, You S, Riera CE, Tamukong P, Smith-Warner SA, Genkinger JM, Luthringer DJ, Freeman MR, Freedland SJ. The effects of glycemic index on prostate cancer progression in a xenograft mouse model. Prostate Cancer Prostatic Dis 2024; 27:348-354. [PMID: 38082056 PMCID: PMC11096094 DOI: 10.1038/s41391-023-00769-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 11/15/2023] [Accepted: 11/24/2023] [Indexed: 05/18/2024]
Abstract
BACKGROUND Previously, we found low-carbohydrate diets slowed prostate cancer (PC) growth and increased survival vs. a Western diet in mice, by inhibiting the insulin/IGF-1 axis. Thus, we tested whether modifying carbohydrate quality to lower glycemic index (GI) without changing quantity results in similar benefits as with reduced quantity. METHODS Male SCID mice injected with LAPC-4 cells were single-housed and randomized when their tumors reached 200 mm3 on average to a LoGI (48% carbohydrate kcal, from Hylon-VII) or HiGI Western diet (48% carbohydrate kcal, from sucrose). Body weight and tumor volume were measured weekly. Body composition was assessed 35 days after randomization. Blood glucose and serum insulin, IGF-1 and IGFBP3 were measured at study end when tumor volumes reached 800 mm3. We analyzed gene expression of mice tumors by RNA-sequencing and human tumors using the Prostate Cancer Transcriptome Atlas. RESULTS There were no significant differences in tumor volume (P > 0.05), tumor proliferation (P = 0.29), and overall survival (P = 0.15) between groups. At 35 days after randomization, the LoGI group had 30% lower body fat (P = 0.007) despite similar body weight (P = 0.58). At sacrifice, LoGI mice had smaller livers (P < 0.001) and lower glucose (P = 0.15), insulin (P = 0.11), IGF-1 (P = 0.07) and IGF-1:IGFBP3 ratio (P = 0.05), and higher IGFBP3 (P = 0.09) vs. HiGI, although none of these metabolic differences reached statistical significance. We observed differential gene expression and pathway enrichment in mice tumors by diet. The most upregulated and downregulated gene in the LoGI group showed expression patterns more closely resembling expression in human benign prostate tissue vs. PC. CONCLUSIONS In this single mouse xenograft model, consuming a low GI diet did not delay PC growth or survival vs. a high GI diet despite suggestions of decreased activation of the insulin/IGF-1 pathway. These data suggest that improving carbohydrate quality alone while consuming a high carbohydrate diet may not effectively slow PC growth.
Collapse
Affiliation(s)
| | - Everardo Macias
- Department of Pathology, Duke University School of Medicine, Durham, NC, USA
| | - Sergio Sanders
- Department of Urology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | | | - Shannon Stock
- Department of Mathematics and Computer Science, College of the Holy Cross, Worcester, MA, USA
- Department of Surgery, Urology Section, Veterans Affairs Health Care System, Durham, NC, USA
| | - Sungyong You
- Department of Urology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Computational Biomedicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Celine E Riera
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Patrick Tamukong
- Department of Urology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Stephanie A Smith-Warner
- Departments of Nutrition and Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Jeanine M Genkinger
- Mailman School of Public Health, Columbia University, New York, NY, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, USA
| | | | - Michael R Freeman
- Department of Urology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Stephen J Freedland
- Department of Urology, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
- Department of Surgery, Urology Section, Veterans Affairs Health Care System, Durham, NC, USA.
| |
Collapse
|
5
|
Perri G, Vilas Boas VG, Nogueira MRS, Mello Júnior EJF, Coelho AL, Posadas EM, Hogaboam C, Cavassani KA, Campanelli AP. Interleukin 33 supports squamous cell carcinoma growth via a dual effect on tumour proliferation, migration and invasion, and T cell activation. Cancer Immunol Immunother 2024; 73:110. [PMID: 38662248 PMCID: PMC11045681 DOI: 10.1007/s00262-024-03676-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 03/14/2024] [Indexed: 04/26/2024]
Abstract
Interleukin (IL)-33 is an important cytokine in the tumour microenvironment; it is known to promote the growth and metastasis of solid cancers, such as gastric, colorectal, ovarian and breast cancer. Our group demonstrated that the IL-33/ST2 pathway enhances the development of squamous cell carcinoma (SCC). Conversely, other researchers have reported that IL-33 inhibits tumour progression. In addition, the crosstalk between IL-33, cancer cells and immune cells in SCC remains unknown. The aim of this study was to investigate the effect of IL-33 on the biology of head and neck SCC lines and to evaluate the impact of IL-33 neutralisation on the T cell response in a preclinical model of SCC. First, we identified epithelial and peritumoural cells as a major local source of IL-33 in human SCC samples. Next, in vitro experiments demonstrated that the addition of IL-33 significantly increased the proliferative index, motility and invasiveness of SCC-25 cells, and downregulated MYC gene expression in SCC cell lines. Finally, IL-33 blockade significantly delayed SCC growth and led to a marked decrease in the severity of skin lesions. Importantly, anti-IL-33 monoclonal antibody therapy increase the percentage of CD4+IFNγ+ T cells and decreased CD4+ and CD8+ T cells secreting IL-4 in tumour-draining lymph nodes. Together, these data suggest that the IL-33/ST2 pathway may be involved in the crosstalk between the tumour and immune cells by modulating the phenotype of head and neck SCC and T cell activity. IL-33 neutralisation may offer a novel therapeutic strategy for SCC.
Collapse
Affiliation(s)
- Graziela Perri
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Al. Dr. Octávio Pinheiro Brisolla, Bauru, SP, 17012-901, Brazil
| | - Vanessa Garcia Vilas Boas
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Al. Dr. Octávio Pinheiro Brisolla, Bauru, SP, 17012-901, Brazil
| | - Maria Renata Sales Nogueira
- Research and Teaching Division, State Department of Health, Instituto Lauro de Souza Lima, Bauru, SP, Brazil
| | | | - Ana Lucia Coelho
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Edwin M Posadas
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Cory Hogaboam
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Karen A Cavassani
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Ana Paula Campanelli
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Al. Dr. Octávio Pinheiro Brisolla, Bauru, SP, 17012-901, Brazil.
| |
Collapse
|
6
|
Zhang S, Tang Z. Prognostic and clinicopathological significance of systemic inflammation response index in patients with hepatocellular carcinoma: a systematic review and meta-analysis. Front Immunol 2024; 15:1291840. [PMID: 38469315 PMCID: PMC10925676 DOI: 10.3389/fimmu.2024.1291840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Accepted: 02/05/2024] [Indexed: 03/13/2024] Open
Abstract
Background It is unclear whether the systemic inflammation response index (SIRI) can predict the prognosis of patients with hepatocellular carcinoma (HCC). Consequently, the present study focused on systematically identifying the relationship between SIRI and the prognosis of patients with HCC through a meta-analysis. Methods Systematic and comprehensive studies were retrieved from PubMed, Web of Science, Embase, and the Cochrane Library from their inception to August 10, 2023. The role of SIRI in predicting overall survival (OS) and progression-free survival (PFS) in HCC was determined using pooled hazard ratios (HRs) and 95% confidence intervals (CIs). Odds ratios (ORs) and 95% CIs were pooled to analyze the correlations between SIRI and the clinicopathological features of HCC. Results Ten articles involving 2,439 patients were included. An elevated SIRI was significantly associated with dismal OS (HR=1.75, 95% CI=1.52-2.01, p<0.001) and inferior PFS (HR=1.66, 95% CI=1.34-2.05, p<0.001) in patients with HCC. Additionally, according to the combined results, the increased SIRI was significantly related to multiple tumor numbers (OR=1.42, 95% CI=1.09-1.85, p=0.009) and maximum tumor diameter >5 cm (OR=3.06, 95% CI=1.76-5.30, p<0.001). However, the SIRI did not show any significant relationship with sex, alpha-fetoprotein content, Child-Pugh class, or hepatitis B virus infection. Conclusion According to our results, elevated SIRI significantly predicted OS and PFS in patients with HCC. Moreover, the SIRI was significantly associated with tumor aggressiveness. Systematic review registration https://inplasy.com/inplasy-2023-9-0003/, identifier INPLASY202390003.
Collapse
Affiliation(s)
| | - Zhining Tang
- Clinical Laboratory, Huzhou Central Hospital, Affiliated Central Hospital of Huzhou University, The Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, Huzhou, Zhejiang, China
| |
Collapse
|
7
|
Chen ML, Yuan TT, Chuang CF, Huang YT, Chung IC, Huang WC. A Novel Enolase-1 Antibody Targets Multiple Interacting Players in the Tumor Microenvironment of Advanced Prostate Cancer. Mol Cancer Ther 2022; 21:1337-1347. [PMID: 35700013 PMCID: PMC9662882 DOI: 10.1158/1535-7163.mct-21-0285] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 08/11/2021] [Accepted: 06/03/2022] [Indexed: 01/07/2023]
Abstract
Prostate cancer is one of the most common causes of cancer death in men worldwide, and the treatment options are limited for patients with advanced stages of prostate cancer. Upon oncogenic or inflammatory stimulation, tumor cells or immune cells express cell surface enolase-1 (ENO1) as plasminogen receptor to facilitate their migration via plasmin activation. Little is known about the roles of ENO1 in prostate cancer, especially in the tumor microenvironment (TME). We hypothesized that targeting surface ENO1 with specific mAbs would exert multifactorial therapeutic potentials against prostate cancer. In vivo, we showed ENO1 mAb (HuL227) reduced the growth of subcutaneous PC-3 xenograft, monocytes recruitment, and intratumoral angiogenesis. In a PC-3 intratibial implantation model, HuL227 reduced tumor growth and osteoclast activation in the bone. To investigate the antitumor mechanism of ENO1 mAb, we found that blocking surface ENO1 significantly reduced VEGF-A-induced tube formation of endothelial cells in vitro. Furthermore, HuL227 inhibited inflammation-enhanced osteoclasts activity and the secretion of invasion-related cytokines CCL2 and TGFβ from osteoclasts. In addition, inflammation-induced migration and chemotaxis of androgen-independent prostate cancer cells were dose-dependently inhibited by HuL227. In summary, we showed that, ENO1 mAb targets multiple TME niches involved in prostate cancer progression and bone metastasis via a plasmin-related mechanism, which may provide a novel immunotherapy approach for men with advanced prostate cancer.
Collapse
Affiliation(s)
| | - Ta-Tung Yuan
- HuniLife Biotechnology Inc., Taiwan.,Corresponding Authors: Wei-Ching Huang, Department of Research and Development, HuniLife Biotechnology Inc., Rm. 1, 6F, No.308, Sec. 1, Neihu Road, Neihu District, Taipei City 114, Taiwan. Phone: 8862-2657-9668; Fax: 8862-2657-9669; E-mail: ; and Ta-Tung Yuan,
| | | | | | | | - Wei-Ching Huang
- HuniLife Biotechnology Inc., Taiwan.,Corresponding Authors: Wei-Ching Huang, Department of Research and Development, HuniLife Biotechnology Inc., Rm. 1, 6F, No.308, Sec. 1, Neihu Road, Neihu District, Taipei City 114, Taiwan. Phone: 8862-2657-9668; Fax: 8862-2657-9669; E-mail: ; and Ta-Tung Yuan,
| |
Collapse
|
8
|
Liang X, Yu G, Zha L, Guo X, Cheng A, Qin C, Zhang H, Wang Z. Identification and Comprehensive Prognostic Analysis of a Novel Chemokine-Related lncRNA Signature and Immune Landscape in Gastric Cancer. Front Cell Dev Biol 2022; 9:797341. [PMID: 35096827 PMCID: PMC8795836 DOI: 10.3389/fcell.2021.797341] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 12/28/2021] [Indexed: 12/19/2022] Open
Abstract
Gastric cancer (GC) is a malignant tumor with poor survival outcomes. Immunotherapy can improve the prognosis of many cancers, including GC. However, in clinical practice, not all cancer patients are sensitive to immunotherapy. Therefore, it is essential to identify effective biomarkers for predicting the prognosis and immunotherapy sensitivity of GC. In recent years, chemokines have been widely reported to regulate the tumor microenvironment, especially the immune landscape. However, whether chemokine-related lncRNAs are associated with the prognosis and immune landscape of GC remains unclear. In this study, we first constructed a novel chemokine-related lncRNA risk model to predict the prognosis and immune landscape of GC patients. By using various algorithms, we identified 10 chemokine-related lncRNAs to construct the risk model. Then, we determined the prognostic efficiency and accuracy of the risk model. The effectiveness and accuracy of the risk model were further validated in the testing set and the entire set. In addition, our risk model exerted a crucial role in predicting the infiltration of immune cells, immune checkpoint genes expression, immunotherapy scores and tumor mutation burden of GC patients. In conclusion, our risk model has preferable prognostic performance and may provide crucial clues to formulate immunotherapy strategies for GC.
Collapse
Affiliation(s)
- Xiaolong Liang
- Department of Gastrointestinal Surgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Gangfeng Yu
- Institute of Life Sciences, Chongqing Medical University, Chongqing, China
| | - Lang Zha
- Department of Gastrointestinal Surgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiong Guo
- Department of Gastrointestinal Surgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Anqi Cheng
- Department of Gastrointestinal Surgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Chuan Qin
- Department of Gastrointestinal Surgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Department of Digestive Oncology, Three Gorges Hospital, Chongqing University, Chongqing, China
| | - Han Zhang
- Department of Digestive Oncology, Three Gorges Hospital, Chongqing University, Chongqing, China
| | - Ziwei Wang
- Department of Gastrointestinal Surgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
9
|
Barreto FS, Ribeiro WLC, Cavalcanti BC, Silva PGDB, Soares CN, Vasconcelos GS, Nunes APN, Moraes Filho MOD, Macedo DS. Early maternal separation enhances melanoma progression in adult female mice by immune mechanisms. Ann N Y Acad Sci 2021; 1502:40-53. [PMID: 34184281 DOI: 10.1111/nyas.14625] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 05/04/2021] [Accepted: 05/25/2021] [Indexed: 01/04/2023]
Abstract
Maternal separation (MS) is a risk factor for major depressive disorder. Both cancer and depression seem to share a common biological link. Here, we evaluated the progression of melanoma and the underlying mechanisms related to this progression, namely cell proliferation and apoptosis, in adult female mice exposed to MS. Female C57BL/6 mice were exposed to MS for 60 min/day during the first 2 postnatal weeks (here called MS mice) or left undisturbed (here called non-MS mice). Melanoma cells were inoculated subcutaneously into the axillary region of adult animals, and tumor progression was evaluated for 25 days. Adult MS mice presented depressive-like behavior and working memory deficits. MS accelerated murine melanoma growth by mechanisms related to decreased apoptosis and increased cell proliferation rate, such as increased expression of IL-6 and mTOR. MS stimulated eukaryotic elongation factor 2 expression and increased the number of circulating monocytes and DNA damage in peripheral blood leukocytes, an effect associated with oxidative DNA damage. In conclusion, MS accelerated the progression of murine melanoma by mechanisms related to tumor proliferation and apoptosis, revealing a relationship between adverse childhood experiences and cancer progression, particularly melanoma.
Collapse
Affiliation(s)
- Francisco Stefânio Barreto
- Laboratory of Experimental Oncology, Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - Wesley Lyeverton Correia Ribeiro
- Laboratory of Experimental Oncology, Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - Bruno Coêlho Cavalcanti
- Laboratory of Experimental Oncology, Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - Paulo Goberlânio de Barros Silva
- Division of Oral Pathology, Department of Dental Clinic, Faculty of Pharmacy, Dentistry and Nursing, Federal University of Ceara, Fortaleza, Ceara, Brazil
| | - Caren Nádia Soares
- Neuropsychopharmacology Laboratory, Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - Germana Silva Vasconcelos
- Neuropsychopharmacology Laboratory, Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - Ana Paula Negreiros Nunes
- Division of Oral Pathology, Department of Dental Clinic, Faculty of Pharmacy, Dentistry and Nursing, Federal University of Ceara, Fortaleza, Ceara, Brazil
| | - Manoel Odorico de Moraes Filho
- Laboratory of Experimental Oncology, Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - Danielle S Macedo
- Neuropsychopharmacology Laboratory, Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceará, Fortaleza, Ceará, Brazil.,National Institute for Translational Medicine (INCT-TM, CNPq), Ribeirão Preto, Brazil
| |
Collapse
|
10
|
Xavier CPR, Castro I, Caires HR, Ferreira D, Cavadas B, Pereira L, Santos LL, Oliveira MJ, Vasconcelos MH. Chitinase 3-like-1 and fibronectin in the cargo of extracellular vesicles shed by human macrophages influence pancreatic cancer cellular response to gemcitabine. Cancer Lett 2021; 501:210-223. [PMID: 33212158 DOI: 10.1016/j.canlet.2020.11.013] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Revised: 10/25/2020] [Accepted: 11/09/2020] [Indexed: 12/13/2022]
Abstract
Tumour-associated macrophages have been implicated in pancreatic ductal adenocarcinoma (PDAC) therapy response and Extracellular vesicles (EVs) shed by macrophages might have a role in this process. Here, we demonstrated that large EVs released by anti-inflammatory human macrophages decreased PDAC cellular sensitivity to gemcitabine. Using proteomic analysis, chitinase 3-like-1 (CHI3L1) and fibronectin (FN1) were identified as two of the most abundant proteins in the cargo of macrophages-derived EVs. Overexpression of CHI3L1 and FN1, using recombinant human proteins, induced PDAC cellular resistance to gemcitabine through ERK (extracellular-signal-regulated kinase) activation. Inhibition of CHI3L1 and FN1 by pentoxifylline and pirfenidone, respectively, partially reverted gemcitabine resistance. In PDAC patient samples, CHI3L1 and FN1 were expressed in the stroma, associated with the high presence of macrophages. The Cancer Genome Atlas analysis revealed an association between CHI3L1 and FN1 gene expression, overall survival of PDAC patients, gemcitabine response, and macrophage infiltration. Altogether, our data identifies CHI3L1 and FN1 as potential targets for pharmacological inhibition in PDAC. Further pre-clinical in vivo work is warranted to study the possibility of repurposing pentoxifylline and pirfenidone as adjuvant therapies for PDAC treatment.
Collapse
Affiliation(s)
- Cristina P R Xavier
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Portugal; Cancer Drug Resistance Group, IPATIMUP - Institute of Molecular Pathology and Immunology, University of Porto, Portugal
| | - Inês Castro
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Portugal; Cancer Drug Resistance Group, IPATIMUP - Institute of Molecular Pathology and Immunology, University of Porto, Portugal
| | - Hugo R Caires
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Portugal; Cancer Drug Resistance Group, IPATIMUP - Institute of Molecular Pathology and Immunology, University of Porto, Portugal
| | - Dylan Ferreira
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Portugal; Experimental Pathology and Therapeutics Group, IPO - Instituto Português de Oncologia, Porto, Portugal
| | - Bruno Cavadas
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Portugal; Genetic Diversity Group, IPATIMUP - Institute of Molecular Pathology and Immunology, University of Porto, Portugal
| | - Luisa Pereira
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Portugal; Genetic Diversity Group, IPATIMUP - Institute of Molecular Pathology and Immunology, University of Porto, Portugal
| | - Lúcio L Santos
- Experimental Pathology and Therapeutics Group, IPO - Instituto Português de Oncologia, Porto, Portugal; ICBAS - Biomedical Sciences Institute Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Maria J Oliveira
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Portugal; FMUP - Faculdade de Medicina da Universidade do Porto, Porto, Portugal; Tumour and Microenvironment Interactions Group, INEB - Instituto Nacional de Engenharia Biomédica, Porto, Portugal
| | - M Helena Vasconcelos
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Portugal; Cancer Drug Resistance Group, IPATIMUP - Institute of Molecular Pathology and Immunology, University of Porto, Portugal; Department of Biological Sciences, FFUP - Faculty of Pharmacy of the University of Porto, Porto, Portugal.
| |
Collapse
|
11
|
Interleukin-6 and Lymphocyte Count Associated and Predicted the Progression of Frailty Syndrome in Prostate Cancer Patients Undergoing Antiandrogen Therapy. Cancers (Basel) 2020; 12:cancers12071716. [PMID: 32610428 PMCID: PMC7408184 DOI: 10.3390/cancers12071716] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 06/24/2020] [Accepted: 06/25/2020] [Indexed: 02/07/2023] Open
Abstract
Frailty syndrome is a functional state that includes a loss of ability to react to stressors, and is associated with poor outcomes, morbidity and premature mortality. The first line treatment in many men with prostate cancer (PCa) consists of an androgen-deprivation therapy (ADT) which can promote or favor frailty syndrome and ADT may therefore favor the progression of frailty over time. Among the pathophysiological bases of frailty, the presence of chronic low-grade inflammation has been associated with its adverse outcomes, but longitudinal studies are needed to validate these biomarkers. In this study, we prospectively evaluate frailty syndrome and blood inflammatory markers (IL1-beta, IL-6, IL-8, TNF alpha, C reactive protein) and leukocytes were measured at baseline and an average of 1 year later in PCa under ADT. Frailty was defined as having three or more of the following components: low lean mass, weakness, self-reported exhaustion, low activity level, and slow walking speed; prefrailty was defined as having one or two of those components. Multinomial regression analysis showed that among the inflammatory biomarkers, those significantly and repeatedly (baseline and follow-up time points) (p < 0.05) associated with frailty syndrome were high IL-6 levels and low lymphocyte counts in blood. Other biomarkers such as IL-8, monocyte counts and C reactive protein were significantly associated with frailty syndrome (p < 0.05) in cross-sectional analyses, but they do not predict frailty progression at 1 year-follow-up. Receiver operating characteristic curve analysis showed that both lymphocyte counts and IL-6 concentration significantly (p < 0.05) (although moderately) discriminate PCa patients that progressed in the severity of frailty syndrome. IL-6 and lymphocytes count are possible biomarkers, useful for identifying frail patients and predicting the progression of frailty in PCa under ADT. Our study suggests the use of these biomarkers to guide clinical decisions on prostate cancer treatment based on a multidisciplinary approach.
Collapse
|
12
|
Griffiths SG, Ezrin A, Jackson E, Dewey L, Doucette AA. A robust strategy for proteomic identification of biomarkers of invasive phenotype complexed with extracellular heat shock proteins. Cell Stress Chaperones 2019; 24:1197-1209. [PMID: 31650515 PMCID: PMC6882979 DOI: 10.1007/s12192-019-01041-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 10/03/2019] [Accepted: 10/09/2019] [Indexed: 12/17/2022] Open
Abstract
As an extension of their orchestration of intracellular pathways, secretion of extracellular heat shock proteins (HSPs) is an emerging paradigm of homeostasis imperative to multicellular organization. Extracellular HSP is axiomatic to the survival of cells during tumorigenesis; proportional representation of specific HSP family members is indicative of invasive potential and prognosis. Further significance has been added by the knowledge that all cancer-derived exosomes have surface-exposed HSPs that reflect the membrane topology of cells that secrete them. Extracellular HSPs are also characteristic of chronic inflammation and sepsis. Accordingly, interrogation of extracellular HSPs secreted from cell culture models may represent a facile means of identifying translational biomarker signatures for targeting in situ. In the current study, we evaluated a simple peptide-based multivalent HSP affinity approach using the Vn96 peptide for low speed pelleting of HSP complexes from bioreactor cultures of cell lines with varying invasive phenotype in xenotransplant models: U87 (glioblastoma multiforme; invasive); HELA (choriocarcinoma; minimally invasive); HEK293T (virally transformed immortalized; embryonic). Proteomic profiling by bottom-up mass spectrometry revealed a comprehensive range of candidate biomarkers including primary HSP ligands. HSP complexes were associated with additional chaperones of prognostic significance such as protein disulfide isomerases, as well as pleiotropic metabolic enzymes, established as proportionally reflective of invasive phenotype. Biomarkers of inflammatory and mechanotransductive phenotype were restricted to the most invasive cell model U87, including chitinase CHI3L1, lamin C, amyloid derivatives, and histone isoforms.
Collapse
Affiliation(s)
| | - Alan Ezrin
- NX Development Corporation, Louisville, KY, USA
| | - Emily Jackson
- David H. Murdock Research Institute, Kannapolis, NC, USA
| | - Lisa Dewey
- David H. Murdock Research Institute, Kannapolis, NC, USA
| | | |
Collapse
|
13
|
Hayashi T, Fujita K, Matsushita M, Nonomura N. Main Inflammatory Cells and Potentials of Anti-Inflammatory Agents in Prostate Cancer. Cancers (Basel) 2019; 11:cancers11081153. [PMID: 31408948 PMCID: PMC6721573 DOI: 10.3390/cancers11081153] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 08/08/2019] [Accepted: 08/09/2019] [Indexed: 02/07/2023] Open
Abstract
Prostate cancer is the most common type of cancer and the leading cause of cancer deaths among men in many countries. Preventing progression is a major concern for prostate cancer patients on active surveillance, patients with recurrence after radical therapies, and patients who acquired resistance to systemic therapies. Inflammation, which is induced by various factors such as infection, microbiome, obesity, and a high-fat diet, is the major etiology in the development of prostate cancer. Inflammatory cells play important roles in tumor progression. Various immune cells including tumor-associated neutrophils, tumor-infiltrating macrophages, myeloid-derived suppressor cells, and mast cells promote prostate cancer via various intercellular signaling. Further basic studies examining the relationship between the inflammatory process and prostate cancer progression are warranted. Interventions by medications and diets to control systemic and/or local inflammation might be effective therapies for prostate cancer progression. Epidemiological investigations and basic research using human immune cells or mouse models have revealed that non-steroidal anti-inflammatory drugs, metformin, statins, soy isoflavones, and other diets are potential interventions for preventing progression of prostate cancer by suppressing inflammation. It is essential to evaluate appropriate indications and doses of each drug and diet.
Collapse
Affiliation(s)
- Takuji Hayashi
- Department of Urology, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Kazutoshi Fujita
- Department of Urology, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan.
| | - Makoto Matsushita
- Department of Urology, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Norio Nonomura
- Department of Urology, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| |
Collapse
|
14
|
Heeke S, Mograbi B, Alix-Panabières C, Hofman P. Never Travel Alone: The Crosstalk of Circulating Tumor Cells and the Blood Microenvironment. Cells 2019; 8:cells8070714. [PMID: 31337010 PMCID: PMC6678604 DOI: 10.3390/cells8070714] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 07/10/2019] [Accepted: 07/11/2019] [Indexed: 12/13/2022] Open
Abstract
Commonly, circulating tumor cells (CTCs) are described as source of metastasis in cancer patients. However, in this process cancer cells of the primary tumor site need to survive the physical and biological challenges in the blood stream before leaving the circulation to become the seed of a new metastatic site in distant parenchyma. Most of the CTCs released in the blood stream will not resist those challenges and will consequently fail to induce metastasis. A few of them, however, interact closely with other blood cells, such as neutrophils, platelets, and/or macrophages to survive in the blood stream. Recent studies demonstrated that the interaction and modulation of the blood microenvironment by CTCs is pivotal for the development of new metastasis, making it an interesting target for potential novel treatment strategies. This review will discuss the recent research on the processes in the blood microenvironment with CTCs and will outline currently investigated treatment strategies.
Collapse
Affiliation(s)
- Simon Heeke
- Université Côte d'Azur, CHU Nice, FHU OncoAge, 06000 Nice, France
- Université Côte d'Azur, CNRS UMR7284, Inserm U1081, Institute for Research on Cancer and Aging, Nice (IRCAN), FHU OncoAge, 06000 Nice, France
| | - Baharia Mograbi
- Université Côte d'Azur, CHU Nice, FHU OncoAge, 06000 Nice, France
- Université Côte d'Azur, CNRS UMR7284, Inserm U1081, Institute for Research on Cancer and Aging, Nice (IRCAN), FHU OncoAge, 06000 Nice, France
| | - Catherine Alix-Panabières
- Laboratory of Rare Human Circulating Cells (LCCRH), University Medical Centre, EA2415, Montpellier University, 34093 Montpellier, France
| | - Paul Hofman
- Université Côte d'Azur, CHU Nice, FHU OncoAge, 06000 Nice, France.
- Université Côte d'Azur, CNRS UMR7284, Inserm U1081, Institute for Research on Cancer and Aging, Nice (IRCAN), FHU OncoAge, 06000 Nice, France.
- Laboratory of Clinical and Experimental Pathology and Biobank BB-0033-00025, Pasteur Hospital, FHU OncoAge, 06000 Nice, France.
| |
Collapse
|
15
|
Peng H, Luo X. Prognostic significance of elevated pretreatment systemic inflammatory markers for patients with prostate cancer: a meta-analysis. Cancer Cell Int 2019; 19:70. [PMID: 30962764 PMCID: PMC6434630 DOI: 10.1186/s12935-019-0785-2] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Accepted: 03/15/2019] [Indexed: 02/06/2023] Open
Abstract
Background Pretreatment inflammatory factors, including neutrophil, lymphocyte, platelet and monocyte counts as well as the ratios between them such as neutrophil–lymphocyte ratio (NLR), platelet–lymphocyte ratio (PLR) and lymphocyte–monocyte ratio (LMR) have been suggested as potential prognostic predictors for patients with prostate cancer (PCa). However, the prognostic effects remain controversial. Therefore, the goal of this study was evaluate the prognostic values of these markers for PCa patients using a meta-analysis. Methods Potentially relevant publications in PubMed and Cochrane Library were searched. Pooled hazard ratio (HR) with 95% confidence interval (CI) for overall survival (OS), cancer-specific survival (CSS), progression-free survival (PFS), recurrence free survival (RFS) and distant metastases-free survival (DMFS) were determined using a fixed or random effects model by STATA 13.0 software. Results Thirty-two studies involving 21,949 participants were included. Our pooled results demonstrated that a high pretreatment NLR (HR = 1.55, 95% CI 1.37–1.76), PLR (HR = 1.72; 95% CI 1.36–2.18), neutrophil (HR = 1.10; 95% CI 1.03–1.18 and monocyte counts (HR = 2.25; 95% CI 1.67–3.05) predicted inferior OS, while elevated pretreatment LMR (HR = 2.27; 95% CI 1.76–2.94) was correlated with favorable OS. Furthermore, the higher NLR (HR = 1.62; 95% CI 1.29–2.04) and monocyte counts (HR = 1.75; 95% CI 1.36–2.25), but lower LMR predicted worse PFS (HR = 2.18; 95% CI 1.58–3.02); poor RFS was only associated with NLR (HR = 1.12; 95% CI 1.04–1.20). The subgroup analysis showed that the higher NLR may be a predictive factor for OS only in patients with mCRPC and undergoing chemotherapy; while the higher PLR was only significantly associated with OS in localized PCa regardless of treatment. Conclusion This meta-analysis reveals that pretreatment NLR, PLR, LMR, neutrophil, and monocyte counts may be effective predictive biomarkers for prognosis in patients with PCa.
Collapse
Affiliation(s)
- Hao Peng
- Department of Urological Surgery, Zhoukou Central Hospital of Henan Province, No. 26 Renmin East Road, Chuanhui District, Zhoukou, 466000 China
| | - Xiaogang Luo
- 2State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Materials Science and Engineering, Donghua University, Shanghai, 201620 China
| |
Collapse
|
16
|
Cavassani KA, Meza RJ, Habiel DM, Chen J, Montes A, Tripathi M, Martins GA, Crother TR, You S, Hogaboam CM, Bhowmick N, Posadas EM. Circulating monocytes from prostate cancer patients promote invasion and motility of epithelial cells. Cancer Med 2018; 7:4639-4649. [PMID: 30094958 PMCID: PMC6143932 DOI: 10.1002/cam4.1695] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 06/28/2018] [Accepted: 06/29/2018] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Recruited myeloid cells are known to promote cancer initiation, malignant progression, metastasis, and resistance to therapy in the tumor niche. We tested the hypothesis that circulating blood monocytes from advanced prostate cancer (PCa) patients exhibit a protumor phenotype and directly influence the tumor microenvironment in response to tumor-derived signals. METHODS Blood monocytes from advanced and stable PCa patients were cultured, and the conditioned media (CM) were collected and analyzed using standard invasion and wound closure assays to measure effects on invasion and motility of PCa tumor cells. We then identified the proteome profile of these monocytes using proteome array and ELISA. RESULTS Conditioned media from circulating monocytes in patients with metastatic prostate cancer (PCa-M) increased invasion of epithelial PCa cells in vitro. Proteome Profiler Analysis revealed that monocyte-derived CM from metastatic castration-resistant (mCRPC) patients presented high levels of chitinase-3-like 1 (CHI3L1, YKL-40) when compared to patients with stable disease (PCa-N) and healthy control individuals (HC). The only described receptor for CHI3L1, interleukin-13 receptor α2 (IL-13Rα2), was significantly up-regulated in the human metastatic PCa cell line, ARCaPM . Accordingly, we observed that the activation of IL-13Rα2 from PCa-M CM increased the invasiveness of ARCaPM cells while siRNA directed against this receptor significantly reduced invasiveness of these cells in the presence of CM from PCa-M patients. CONCLUSIONS Thus, we show that circulating monocytes from metastatic PCa patients exert a tumor-promoting role via the secretion of CHI3L1, and CHI3L1 demands further exploration as a possible therapeutic target in advanced PCa.
Collapse
Affiliation(s)
- Karen A. Cavassani
- Urologic Oncology Program/Uro‐Oncology Research LaboratoriesSamuel Oschin Comprehensive Cancer InstituteCedars‐Sinai Medical CenterLos AngelesCA90048USA
| | - Rebecca J. Meza
- Urologic Oncology Program/Uro‐Oncology Research LaboratoriesSamuel Oschin Comprehensive Cancer InstituteCedars‐Sinai Medical CenterLos AngelesCA90048USA
| | - David M. Habiel
- Division of Pulmonary and Critical Care MedicineDepartment of Medicine & Women's Guild Lung InstituteCedars‐Sinai Medical CenterLos AngelesCA90048USA
| | - Jie‐Fu Chen
- Urologic Oncology Program/Uro‐Oncology Research LaboratoriesSamuel Oschin Comprehensive Cancer InstituteCedars‐Sinai Medical CenterLos AngelesCA90048USA
| | - Alexander Montes
- Urologic Oncology Program/Uro‐Oncology Research LaboratoriesSamuel Oschin Comprehensive Cancer InstituteCedars‐Sinai Medical CenterLos AngelesCA90048USA
| | - Manisha Tripathi
- Department of MedicineCedars‐Sinai Medical CenterLos AngelesCA90048USA
| | - Gislâine A. Martins
- F. Widjaja Foundation Inflammatory Bowel and Immunobiology Research InstituteCedars‐Sinai Medical CenterLos AngelesCA90048USA
| | - Timothy R. Crother
- Department of Pediatric, Infectious diseases and ImmunologyCedars‐Sinai Medical CenterLos AngelesCA90048USA
| | - Sungyong You
- Urologic Oncology Program/Uro‐Oncology Research LaboratoriesSamuel Oschin Comprehensive Cancer InstituteCedars‐Sinai Medical CenterLos AngelesCA90048USA
- Division of Hematology/OncologyDepartment of MedicineCedars‐Sinai Medical CenterLos AngelesCA90048USA
| | - Cory M. Hogaboam
- Division of Pulmonary and Critical Care MedicineDepartment of Medicine & Women's Guild Lung InstituteCedars‐Sinai Medical CenterLos AngelesCA90048USA
| | - Neil Bhowmick
- Department of MedicineCedars‐Sinai Medical CenterLos AngelesCA90048USA
| | - Edwin M. Posadas
- Urologic Oncology Program/Uro‐Oncology Research LaboratoriesSamuel Oschin Comprehensive Cancer InstituteCedars‐Sinai Medical CenterLos AngelesCA90048USA
- Division of Hematology/OncologyDepartment of MedicineCedars‐Sinai Medical CenterLos AngelesCA90048USA
- Translational Oncology ProgramSamuel Oschin Comprehensive Cancer InstituteLos AngelesCA90048USA
| |
Collapse
|