1
|
Jian J, Wang X, Zhang J, Zhou C, Hou X, Huang Y, Hou J, Lin Y, Wei X. Molecular landscape for risk prediction and personalized therapeutics of castration-resistant prostate cancer: at a glance. Front Endocrinol (Lausanne) 2024; 15:1360430. [PMID: 38887275 PMCID: PMC11180744 DOI: 10.3389/fendo.2024.1360430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Accepted: 05/20/2024] [Indexed: 06/20/2024] Open
Abstract
Prostate cancer (PCa) is commonly occurred with high incidence in men worldwide, and many patients will be eventually suffered from the dilemma of castration-resistance with the time of disease progression. Castration-resistant PCa (CRPC) is an advanced subtype of PCa with heterogeneous carcinogenesis, resulting in poor prognosis and difficulties in therapy. Currently, disorders in androgen receptor (AR)-related signaling are widely acknowledged as the leading cause of CRPC development, and some non-AR-based strategies are also proposed for CRPC clinical analyses. The initiation of CRPC is a consequence of abnormal interaction and regulation among molecules and pathways at multi-biological levels. In this study, CRPC-associated genes, RNAs, proteins, and metabolites were manually collected and integrated by a comprehensive literature review, and they were functionally classified and compared based on the role during CRPC evolution, i.e., drivers, suppressors, and biomarkers, etc. Finally, translational perspectives for data-driven and artificial intelligence-powered CRPC systems biology analysis were discussed to highlight the significance of novel molecule-based approaches for CRPC precision medicine and holistic healthcare.
Collapse
Affiliation(s)
- Jingang Jian
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Department of Urology, The Fourth Affiliated Hospital of Soochow University, Suzhou, China
| | - Xin’an Wang
- Department of Urology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jun Zhang
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Chenchao Zhou
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Xiaorui Hou
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Department of Urology, The Fourth Affiliated Hospital of Soochow University, Suzhou, China
| | - Yuhua Huang
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Jianquan Hou
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Department of Urology, The Fourth Affiliated Hospital of Soochow University, Suzhou, China
| | - Yuxin Lin
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Center for Systems Biology, Department of Bioinformatics, School of Biology and Basic Medical Sciences, Soochow University, Suzhou, China
| | - Xuedong Wei
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| |
Collapse
|
2
|
Lumahan LEV, Arif M, Whitener AE, Yi P. Regulating Androgen Receptor Function in Prostate Cancer: Exploring the Diversity of Post-Translational Modifications. Cells 2024; 13:191. [PMID: 38275816 PMCID: PMC10814774 DOI: 10.3390/cells13020191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 01/05/2024] [Accepted: 01/09/2024] [Indexed: 01/27/2024] Open
Abstract
Androgen receptor (AR) transcriptional activity significantly influences prostate cancer (PCa) progression. In addition to ligand stimulation, AR transcriptional activity is also influenced by a variety of post-translational modifications (PTMs). A number of oncogenes and tumor suppressors have been observed leveraging PTMs to influence AR activity. Subjectively targeting these post-translational modifiers based on their impact on PCa cell proliferation is a rapidly developing area of research. This review elucidates the modifiers, contextualizes the effects of these PTMs on AR activity, and connects these cellular interactions to the progression of PCa.
Collapse
Affiliation(s)
- Lance Edward V. Lumahan
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77204, USA
| | - Mazia Arif
- Center for Nuclear Receptors and Cell Signaling, Department of Biology and Biochemistry, University of Houston, Houston, TX 77205, USA
| | - Amy E. Whitener
- Center for Nuclear Receptors and Cell Signaling, Department of Biology and Biochemistry, University of Houston, Houston, TX 77205, USA
| | - Ping Yi
- Center for Nuclear Receptors and Cell Signaling, Department of Biology and Biochemistry, University of Houston, Houston, TX 77205, USA
| |
Collapse
|
3
|
Weng H, Xiong KP, Wang W, Qian KY, Yuan S, Wang G, Yu F, Luo J, Lu MX, Yang ZH, Liu T, Huang X, Zheng H, Wang XH. Aspartoacylase suppresses prostate cancer progression by blocking LYN activation. Mil Med Res 2023; 10:25. [PMID: 37271807 PMCID: PMC10240701 DOI: 10.1186/s40779-023-00460-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 05/14/2023] [Indexed: 06/06/2023] Open
Abstract
BACKGROUND Globally, despite prostate cancer (PCa) representing second most prevalent malignancy in male, the precise molecular mechanisms implicated in its pathogenesis remain unclear. Consequently, elucidating the key molecular regulators that govern disease progression could substantially contribute to the establishment of novel therapeutic strategies, ultimately advancing the management of PCa. METHODS A total of 49 PCa tissues and 43 adjacent normal tissues were collected from January 2017 to December 2021 at Zhongnan Hospital of Wuhan University. The advanced transcriptomic methodologies were employed to identify differentially expressed mRNAs in PCa. The expression of aspartoacylase (ASPA) in PCa was thoroughly evaluated using quantitative real-time PCR and Western blotting techniques. To elucidate the inhibitory role of ASPA in PCa cell proliferation and metastasis, a comprehensive set of in vitro and in vivo assays were conducted, including orthotopic and tumor-bearing mouse models (n = 8 for each group). A combination of experimental approaches, such as Western blotting, luciferase assays, immunoprecipitation assays, mass spectrometry, glutathione S-transferase pull-down experiments, and rescue studies, were employed to investigate the underlying molecular mechanisms of ASPA's action in PCa. The Student's t-test was employed to assess the statistical significance between two distinct groups, while one-way analysis of variance was utilized for comparisons involving more than two groups. A two-sided P value of less than 0.05 was deemed to indicate statistical significance. RESULTS ASPA was identified as a novel inhibitor of PCa progression. The expression of ASPA was found to be significantly down-regulated in PCa tissue samples, and its decreased expression was independently associated with patients' prognosis (HR = 0.60, 95% CI 0.40-0.92, P = 0.018). Our experiments demonstrated that modulation of ASPA activity, either through gain- or loss-of-function, led to the suppression or enhancement of PCa cell proliferation, migration, and invasion, respectively. The inhibitory role of ASPA in PCa was further confirmed using orthotopic and tumor-bearing mouse models. Mechanistically, ASPA was shown to directly interact with the LYN and inhibit the phosphorylation of LYN as well as its downstream targets, JNK1/2 and C-Jun, in both PCa cells and mouse models, in an enzyme-independent manner. Importantly, the inhibition of LYN activation by bafetinib abrogated the promoting effect of ASPA knockdown on PCa progression in both in vitro and in vivo models. Moreover, we observed an inverse relationship between ASPA expression and LYN activity in clinical PCa samples, suggesting a potential regulatory role of ASPA in modulating LYN signaling. CONCLUSION Our findings provide novel insights into the tumor-suppressive function of ASPA in PCa and highlight its potential as a prognostic biomarker and therapeutic target for the management of this malignancy.
Collapse
Affiliation(s)
- Hong Weng
- Department of Urology, Zhongnan Hospital of Wuhan University, No. 169 Donghu Road, Wuchang District, Wuhan, 430071 China
- Center for Evidence-Based and Translational Medicine, Zhongnan Hospital of Wuhan University, Wuhan, 430071 China
- Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan, 430071 China
- Hubei Key Laboratory of Tumor Biological Behaviors, Wuhan, 430071 China
| | - Kang-Ping Xiong
- Department of Urology, Zhongnan Hospital of Wuhan University, No. 169 Donghu Road, Wuchang District, Wuhan, 430071 China
| | - Wang Wang
- Department of Urology, Zhongnan Hospital of Wuhan University, No. 169 Donghu Road, Wuchang District, Wuhan, 430071 China
| | - Kai-Yu Qian
- Department of Urology, Zhongnan Hospital of Wuhan University, No. 169 Donghu Road, Wuchang District, Wuhan, 430071 China
- Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan, 430071 China
- Hubei Key Laboratory of Tumor Biological Behaviors, Wuhan, 430071 China
| | - Shuai Yuan
- Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan, 430071 China
| | - Gang Wang
- Department of Urology, Zhongnan Hospital of Wuhan University, No. 169 Donghu Road, Wuchang District, Wuhan, 430071 China
- Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan, 430071 China
| | - Fang Yu
- Department of Pathology, Zhongnan Hospital of Wuhan University, Wuhan, 430071 China
- Center for Pathology and Molecular Diagnostics, Wuhan University, Wuhan, 430071 China
| | - Jun Luo
- Department of Pathology, Zhongnan Hospital of Wuhan University, Wuhan, 430071 China
- Center for Pathology and Molecular Diagnostics, Wuhan University, Wuhan, 430071 China
| | - Meng-Xin Lu
- Department of Urology, Zhongnan Hospital of Wuhan University, No. 169 Donghu Road, Wuchang District, Wuhan, 430071 China
| | - Zhong-Hua Yang
- Department of Urology, Zhongnan Hospital of Wuhan University, No. 169 Donghu Road, Wuchang District, Wuhan, 430071 China
| | - Tao Liu
- Department of Urology, Zhongnan Hospital of Wuhan University, No. 169 Donghu Road, Wuchang District, Wuhan, 430071 China
| | - Xing Huang
- Department of Urology, Zhongnan Hospital of Wuhan University, No. 169 Donghu Road, Wuchang District, Wuhan, 430071 China
| | - Hang Zheng
- Department of Urology, Zhongnan Hospital of Wuhan University, No. 169 Donghu Road, Wuchang District, Wuhan, 430071 China
| | - Xing-Huan Wang
- Department of Urology, Zhongnan Hospital of Wuhan University, No. 169 Donghu Road, Wuchang District, Wuhan, 430071 China
- Center for Evidence-Based and Translational Medicine, Zhongnan Hospital of Wuhan University, Wuhan, 430071 China
- Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan, 430071 China
- Hubei Key Laboratory of Tumor Biological Behaviors, Wuhan, 430071 China
- Wuhan Research Center for Infectious Diseases and Cancer, Chinese Academy of Medical Sciences, Wuhan, 430071 China
| |
Collapse
|
4
|
Seven Shades of Grey: A Follow-Up Study on the Molecular Basis of Coat Colour in Indicine Grey Cattle Using Genome-Wide SNP Data. Genes (Basel) 2022; 13:genes13091601. [PMID: 36140768 PMCID: PMC9498432 DOI: 10.3390/genes13091601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 08/31/2022] [Accepted: 08/31/2022] [Indexed: 11/17/2022] Open
Abstract
Shades of grey and brown are a dominant component in mammal coat colours, representing a fundamental trait involved in a great number of processes including cryptism, sexual selection and signalling. The genetic mechanisms of the grey colouration in mammals are very complex and controlled by hundreds of genes whose effects and interactions are still largely unclear. In this study, we adopted a robust multi-cohort Fst outlier approach based on pairwise contrasts between seven grey indicine cattle breeds and both taurine and indicine non-grey cattle breeds in order to find genomic regions potentially related to the grey colouration. On the basis of three main drawn settings, built in order to control both the effect of the sample size and the genetic structure, we have identified some signals common to those obtained in a previous work employing only taurine cattle. In particular, using the top 1% Fst approach, we detected a candidate region (22.6–23.8 megabases) on chromosome 14 in which genes related to pigmentation have been already documented. In addition, when we constructed a phylogenetic tree using the significant markers identified in this study and including also the genotyping data at these loci of both the grey taurine and the extinct wild auroch, we found a topological repartition consistent with breed colour pattern rather than with the known bovine evolutionary history. Thus, on the basis of this evidence, together with the geographical distribution of the current taurine grey cattle, an ancestral indicine origin for the grey phenotype would seem to be a conceivable interpretation. In this context, a higher thermo-tolerance and less UV-induced damage of the grey phenotype might have favoured the retention of advantageous genes into the taurine genome during the post-Neolithic human-mediated cattle expansions.
Collapse
|
5
|
Nisar A, Kayani MA, Nasir W, Mehmood A, Ahmed MW, Parvez A, Mahjabeen I. Fyn and Lyn gene polymorphisms impact the risk of thyroid cancer. Mol Genet Genomics 2022; 297:1649-1659. [PMID: 36058999 DOI: 10.1007/s00438-022-01946-7] [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: 06/27/2021] [Accepted: 08/11/2022] [Indexed: 10/14/2022]
Abstract
Thyroid cancer is the most common malignancy of the endocrine glands, and during last couple of decades, its incidence has risen alarmingly, across the globe. Etiology of thyroid cancer is still debatable. There are a few worth mentioning risk factors which contribute to initiation of abnormalities in thyroid gland leading to cancer. Genetic instability is major risk factors in thyroid carcinogenesis. Among the genetic factors, the Src family of genes (Src, Yes1, Fyn and Lyn) have been implicated in many cancers but there is little data regarding the association of these (Src, Yes1, Fyn and Lyn) genes with thyroid carcinogenesis. Fyn and Lyn genes of Src family found engaged in proliferation, migration, invasion, angiogenesis, and metastasis in different cancers. This study was planned to examine the effect of Fyn and Lyn SNPs on thyroid cancer risk in Pakistani population in 500 patients and 500 controls. Three polymorphisms of Fyn gene (rs6916861, rs2182644 and rs12910) and three polymorphisms of Lyn gene (rs2668011, rs45587541 and rs45489500) were analyzed using Tetra-primer ARMS-PCR followed by DNA sequencing. SNP rs6916861 of Fyn gene mutant genotype (CC) showed statistically significant threefold increased risk of thyroid cancer (P < 0.0001). In case of rs2182644 of Fyn gene, mutant genotype (AA) indicated statistically significant 17-fold increased risk of thyroid cancer (P < 0.0001). Statistically significant threefold increased risk of thyroid cancer was observed in genotype AC (P < 0.0001) of Fyn gene polymorphism rs12910. In SNP rs2668011 of Lyn gene, TT genotype showed statistically significant threefold increased risk of thyroid cancer (P < 0.0001). In case of rs45587541 of Lyn gene, GA genotypes showed statistically significant 11-fold increased risk in thyroid cancer (P < 0.0001). Haplotype analysis revealed that AAATAG*, AGACAG*, AGCCAA*, AGCCAG*, CAATAG*, CGCCAG* and CGCCGA* haplotypes of Fyn and Lyn polymorphisms are associated with increased thyroid cancer risk. These results showed that genotypes and allele distribution of Fyn and Lyn are significantly linked with increased thyroid cancer risk and could be genetic adjuster for said disease.
Collapse
Affiliation(s)
- Asif Nisar
- Cancer Genetics and Epigenetics Lab, Department of Biosciences, COMSATS University Islamabad, Park Road Tarlai Kalan, Islamabad, Pakistan
| | - Mahmood Akhtar Kayani
- Cancer Genetics and Epigenetics Lab, Department of Biosciences, COMSATS University Islamabad, Park Road Tarlai Kalan, Islamabad, Pakistan
| | - Wajiha Nasir
- Department of Radiation, Nuclear Oncology Radiation Institute, Islamabad, Pakistan
| | - Azhar Mehmood
- Cancer Genetics and Epigenetics Lab, Department of Biosciences, COMSATS University Islamabad, Park Road Tarlai Kalan, Islamabad, Pakistan
| | - Malik Waqar Ahmed
- Cancer Genetics and Epigenetics Lab, Department of Biosciences, COMSATS University Islamabad, Park Road Tarlai Kalan, Islamabad, Pakistan.,Pakistan Institute of Rehabilitation Sciences (PIRS), Isra University Islamabad Campus, Islamabad, Pakistan
| | - Aamir Parvez
- Cancer Genetics and Epigenetics Lab, Department of Biosciences, COMSATS University Islamabad, Park Road Tarlai Kalan, Islamabad, Pakistan
| | - Ishrat Mahjabeen
- Cancer Genetics and Epigenetics Lab, Department of Biosciences, COMSATS University Islamabad, Park Road Tarlai Kalan, Islamabad, Pakistan.
| |
Collapse
|
6
|
Wei W, Ban X, Yang F, Li J, Cheng X, Zhang R, Huang X, Huang Y, Li Q, Qiu Y, Zheng M, Zhu X, Li J. Phase II trial of efficacy, safety and biomarker analysis of sintilimab plus anlotinib for patients with recurrent or advanced endometrial cancer. J Immunother Cancer 2022; 10:e004338. [PMID: 35623659 PMCID: PMC9150151 DOI: 10.1136/jitc-2021-004338] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/09/2022] [Indexed: 12/02/2022] Open
Abstract
BACKGROUND Although co-inhibition of the angiogenesis and programmed death 1 (PD-1) pathways is proposed as an effective anticancer strategy, studies in Chinese patients with endometrial cancer are sufficient. Anlotinib is an oral multi-targeted tyrosine kinase inhibitor affecting tumor angiogenesis and proliferation; sintilimab is an anti-PD-1 monoclonal antibody. METHODS This was a phase II trial using Simon's two-stage design. This study enrolled patients with endometrial cancer who had progressed after platinum-based chemotherapy. Sintilimab 200 mg was administered intravenously on day 1 every 3 weeks, and anlotinib 12 mg was administered on days 1-14 in a 21-day cycle. The primary endpoint was the objective response rate (ORR) using the immune-related Response Evaluation Criteria in Solid Tumors criteria. Immunohistochemistry and whole-exome sequencing were used as correlative investigations. RESULTS Between November 2019 and September 2020, 23 eligible patients were enrolled. The ORR and disease control rates were 73.9% (95% CI, 51.6 to 89.8) and 91.3% (95% CI, 72.0 to 98.9), respectively, with 4 complete and 12 partial responses. With a median follow-up of 15.4 months (95% CI, 12.6 to 18.3), the median progression-free survival was not reached, and the probability of PFS >12 months was 57.1% (95% CI, 33.6 to 75.0). Exploratory analysis revealed that mutations in the homologous repair pathway showed a trend for higher ORR (100% vs 0%, p=0.07). Treatment-related grade 3/4 adverse events were observed in 50.0% of the patients. CONCLUSIONS Sintilimab plus anlotinib demonstrated robust therapeutic benefits with tolerable toxicity in endometrial cancer. TRIAL REGISTRATION NUMBER NCT04157491.
Collapse
Affiliation(s)
- Wei Wei
- Department of Gynecologic Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Xiaohua Ban
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
- Department of Radiology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Fan Yang
- Department of Gynecologic Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Jibin Li
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
- Department of Clinical Research, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xiaqin Cheng
- Department of Gynecologic Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Rong Zhang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
- Department of Radiology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xin Huang
- Department of Gynecologic Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Yongwen Huang
- Department of Gynecologic Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Qiaqia Li
- Department of Gynecologic Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Ya Qiu
- Department of Gynecologic Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Min Zheng
- Department of Gynecologic Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Xiaofeng Zhu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
- Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Jundong Li
- Department of Gynecologic Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| |
Collapse
|
7
|
Klose K, Packeiser EM, Granados-Soler JL, Hewicker-Trautwein M, Murua Escobar H, Nolte I. Evaluation of the therapeutic potential of masitinib and expression of its specific targets c-Kit, PDGFR-α, PDGFR-β, and Lyn in canine prostate cancer cell lines. Vet Comp Oncol 2022; 20:641-652. [PMID: 35384248 DOI: 10.1111/vco.12817] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 03/31/2022] [Accepted: 04/01/2022] [Indexed: 11/28/2022]
Abstract
Canine prostate cancer is classified into adenocarcinoma, transitional cell carcinoma with prostatic involvement, and mixed forms. Early metastatic spread leads to poor prognosis and limited treatment options. Masitinib is approved for the treatment of canine mast cell tumours and inhibits tyrosine kinase c-Kit, tyrosine-protein kinase Lyn (Lyn), and platelet-derived growth factor receptors alpha and beta (PDGFR-α, PDGFR-β), which are known to be expressed in canine prostate cancer. The aim of this study was to evaluate masitinib in an in vitro model consisting of cell lines from primary prostate adenocarcinoma, the associated lymph node metastasis of the same patient, and transitional cell carcinoma. To assess the suitability of the model system, the targets of masitinib were investigated by immunocytochemistry in the cell lines and by immunohistochemistry in the respective formalin-fixed, paraffin-embedded (FFPE) original neoplastic tissue. After exposure to masitinib, cell viability, cell count, apoptosis induction, and protein expression of c-Kit, Lyn, PDGFR-α, and PDGFR-β were assessed. To hedge the efficacy, two application protocols of masitinib (single application or 12-h double-dose regimen) were compared. Immunocytochemical and immunohistochemical analysis revealed increased Lyn, PDGFR-α, and PDGFR-β expression in cell lines and FFPE original neoplastic tissue compared to healthy prostate tissue. Masitinib exposure increased apoptosis, while the cell counts and cell viability decreased in a dose- and application interval-dependent manner, with increased impact in the 12-h double-dose regimen. These in vitro effects of masitinib in canine prostate cancer and associated metastasis support further in vivo research and modifications of the clinical treatment protocol in future studies.
Collapse
Affiliation(s)
- Katharina Klose
- Small Animal Clinic, University of Veterinary Medicine Hannover, Foundation, Hannover, Germany
| | - Eva-Maria Packeiser
- Small Animal Clinic, University of Veterinary Medicine Hannover, Foundation, Hannover, Germany
| | | | | | - Hugo Murua Escobar
- Division of Medicine Clinic III, Hematology, Oncology and Palliative Medicine, University of Rostock, Rostock, Germany
| | - Ingo Nolte
- Small Animal Clinic, University of Veterinary Medicine Hannover, Foundation, Hannover, Germany
| |
Collapse
|
8
|
Zhong S, Peng S, Chen Z, Chen Z, Luo JL. Choosing Kinase Inhibitors for Androgen Deprivation Therapy-Resistant Prostate Cancer. Pharmaceutics 2022; 14:498. [PMID: 35335873 PMCID: PMC8950316 DOI: 10.3390/pharmaceutics14030498] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 01/26/2022] [Accepted: 02/22/2022] [Indexed: 11/25/2022] Open
Abstract
Androgen deprivation therapy (ADT) is a systemic therapy for advanced prostate cancer (PCa). Although most patients initially respond to ADT, almost all cancers eventually develop castration resistance. Castration-resistant PCa (CRPC) is associated with a very poor prognosis, and the treatment of which is a serious clinical challenge. Accumulating evidence suggests that abnormal expression and activation of various kinases are associated with the emergence and maintenance of CRPC. Many efforts have been made to develop small molecule inhibitors to target the key kinases in CRPC. These inhibitors are designed to suppress the kinase activity or interrupt kinase-mediated signal pathways that are associated with PCa androgen-independent (AI) growth and CRPC development. In this review, we briefly summarize the roles of the kinases that are abnormally expressed and/or activated in CRPC and the recent advances in the development of small molecule inhibitors that target kinases for the treatment of CRPC.
Collapse
Affiliation(s)
- Shangwei Zhong
- Department of General Surgery, Xiangya Hospital, Central South University, Hunan 410008, China; (S.Z.); (S.P.); (Z.C.)
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, FL 33459, USA
| | - Shoujiao Peng
- Department of General Surgery, Xiangya Hospital, Central South University, Hunan 410008, China; (S.Z.); (S.P.); (Z.C.)
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, FL 33459, USA
| | - Zihua Chen
- Department of General Surgery, Xiangya Hospital, Central South University, Hunan 410008, China; (S.Z.); (S.P.); (Z.C.)
| | - Zhikang Chen
- Department of General Surgery, Xiangya Hospital, Central South University, Hunan 410008, China; (S.Z.); (S.P.); (Z.C.)
| | - Jun-Li Luo
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, FL 33459, USA
| |
Collapse
|
9
|
Fu X, Zhang Z, Liu M, Li J, A J, Fu L, Huang C, Dong JT. AR imposes different effects on ZFHX3 transcription depending on androgen status in prostate cancer cells. J Cell Mol Med 2021; 26:800-812. [PMID: 34953044 PMCID: PMC8817138 DOI: 10.1111/jcmm.17125] [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: 09/09/2021] [Revised: 11/11/2021] [Accepted: 11/29/2021] [Indexed: 12/24/2022] Open
Abstract
Both androgen receptor (AR) and the ZFHX3 transcription factor modulate prostate development. While AR drives prostatic carcinogenesis, ZFHX3 is a tumour suppressor whose loss activates the PI3K/AKT signalling in advanced prostate cancer (PCa). However, it is unknown whether ZFHX3 and AR are functionally related in PCa cells and, if so, how. Here, we report that in AR-positive LNCaP and C4-2B PCa cells, androgen upregulates ZFHX3 transcription via androgen-induced AR binding to the androgen-responsive elements (AREs) of the ZFHX3 promoter. Androgen also upregulated ZFHX3 transcription in vivo, as castration dramatically reduced Zfhx3 mRNA and protein levels in mouse prostates, and ZFHX3 mRNA levels correlated with AR activities in human PCa. Interestingly, the binding of AR to one ARE occurred in the absence of androgen, and the binding repressed ZFHX3 transcription as this repressive binding was interrupted by androgen treatment. The enzalutamide antiandrogen prevented androgen from inducing ZFHX3 transcription and caused excess ZFHX3 protein degradation. In human PCa, ZFHX3 was downregulated and the downregulation correlated with worse patient survival. These findings establish a regulatory relationship between AR and ZFHX3, suggest a role of ZFHX3 in AR function and implicate ZFHX3 loss in the antiandrogen therapies of PCa.
Collapse
Affiliation(s)
- Xing Fu
- Department of Genetics and Cell Biology, College of Life Sciences, Nankai University, Tianjin, China.,Department of Human Cell Biology and Genetics, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Zhiqian Zhang
- Department of Human Cell Biology and Genetics, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Mingcheng Liu
- Department of Human Cell Biology and Genetics, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Juan Li
- Department of Human Cell Biology and Genetics, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Jun A
- Department of Human Cell Biology and Genetics, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Liya Fu
- Department of Genetics and Cell Biology, College of Life Sciences, Nankai University, Tianjin, China
| | - Chenyang Huang
- Department of Human Cell Biology and Genetics, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Jin-Tang Dong
- Department of Human Cell Biology and Genetics, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| |
Collapse
|
10
|
Abstract
Effective regulation of immune-cell activation is critical for ensuring that the immune response, and inflammation generated for the purpose of pathogen elimination, are limited in space and time to minimize tissue damage. Autoimmune disease can occur when immunoreceptor signaling is dysregulated, leading to unrestrained inflammation and organ damage. Conversely, tumors can coopt the tissue healing and immunosuppressive functions of hematopoietic cells to promote metastasis and evade therapy. The Src-family kinase Lyn is an essential regulator of immunoreceptor signaling, initiating both proinflammatory and suppressive signaling pathways in myeloid immune cells (eg, neutrophils, dendritic cells, monocytes, macrophages) and in B lymphocytes. Defects in Lyn signaling are implicated in autoimmune disease, but mechanisms by which Lyn, expressed along with a battery of other Src-family kinases, may uniquely direct both positive and negative signaling remain incompletely defined. This review describes our current understanding of the activating and inhibitory contributions of Lyn to immunoreceptor signaling and how these processes contribute to myeloid and B-cell function. We also highlight recent work suggesting that the 2 proteins generated by alternative splicing of lyn, LynA and LynB, differentially regulate both immune and cancer-cell signaling. These principles may also extend to other Lyn-expressing cells, such as neuronal and endocrine cells. Unraveling the common and cell-specific aspects of Lyn function could lead to new approaches to therapeutically target dysregulated pathways in pathologies ranging from autoimmune and neurogenerative disease to cancer.
Collapse
Affiliation(s)
- Ben F Brian
- Department of Pharmacology, University of Minnesota, Minneapolis, MN, USA
- Current Affiliation: Current affiliation for B.F.B.: Division of Immunology & Pathogenesis, Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Tanya S Freedman
- Department of Pharmacology, University of Minnesota, Minneapolis, MN, USA
- Center for Immunology, University of Minnesota, Minneapolis, MN, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
- Center for Autoimmune Diseases Research, University of Minnesota, Minneapolis, MN, USA
- Correspondence: Tanya S. Freedman, PhD, University of Minnesota Twin Cities Campus: University of Minnesota, 6-120 Jackson Hall, 321 Church St. S.E., Minneapolis, MN 55455, USA. E-mail:
| |
Collapse
|
11
|
Dai Y, Siemann D. c-Src is required for hypoxia-induced metastasis-associated functions in prostate cancer cells. Onco Targets Ther 2019; 12:3519-3529. [PMID: 31190858 PMCID: PMC6512571 DOI: 10.2147/ott.s201320] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2019] [Accepted: 03/20/2019] [Indexed: 12/14/2022] Open
Abstract
Background: Metastasis is the major cause of therapeutic failure in prostate cancer patients, and hypoxia has been shown to promote metastatic functions. However, whether Src family kinases (SFKs) can be upregulated under hypoxia is unclear. Materials and methods: In the current study, we evaluated the effects of hypoxia on cellular functions and activities of different SFK members (c-Src, Lyn, Fyn) in prostate cancer cells. Prostate cancer cell functions were determined in vitro including migration (wound-healing assay), invasion (Matrigel-based transwell assay) and clonogenic cell survival (colony formation assay). Protein expression was detected by Western blotting and gene knockdown was accomplished by siRNA transfection. Results:SRC, but not LYN and FYN, is associated with overall survival in prostate cancer patients, while all three phosphorylated proteins are highly expressed in tumors compared to normal tissues. Short-term hypoxic exposure significantly enhances cell migration, invasion, clonogenic survival, and consistently, c-Src phosphorylation in both PC-3ML and C4-2B cells. Knockdown of SRC, but not LYN or FYN, abolished hypoxia-induced functions. Finally, small molecule Src inhibitors strongly inhibited cell behaviors and c-Src activation under hypoxic conditions. Conclusion: Our data show that hypoxia is able to enhance metastatic-associated cell functions by activating c-Src in prostate cancer cells. Importantly, SFK inhibition by small molecule inhibitors was able to impair hypoxia-induced metastasis associated cell functions, suggesting a possible role of SFK inhibitors for prostate cancer treatment.
Collapse
Affiliation(s)
- Yao Dai
- Department of Radiation Oncology, University of Florida, Gainesville, FL 32608, USA
| | - Dietmar Siemann
- Department of Radiation Oncology, University of Florida, Gainesville, FL 32608, USA
| |
Collapse
|
12
|
Zhang Q, Meng X, Qin G, Xue X, Dang N. Lyn Kinase Promotes the Proliferation of Malignant Melanoma Cells through Inhibition of Apoptosis and Autophagy via the PI3K/Akt Signaling Pathway. J Cancer 2019; 10:1197-1208. [PMID: 30854129 PMCID: PMC6400685 DOI: 10.7150/jca.28908] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 01/05/2019] [Indexed: 01/02/2023] Open
Abstract
Melanoma is a malignant tumor of cutaneous melanocytes that is characterized by high grade malignancy, rapid progression and high mortality. Thus far, its specific etiological mechanism has been unclear. In this study, we discovered that Lyn kinase expression was up-regulated in melanoma tissues and cells. The function of Lyn was determined by knocking down its expression with a lentivirus containing Lyn shRNA and upregulating its expression with pcDNA3.1-Lyn in the melanoma cell lines M14 and A375. The results showed that Lyn knockdown could significantly inhibit the proliferation, migration and invasiveness through its inhibition of apoptosis and autophagy via the PI3K/Akt pathway in melanoma cell lines. This was further confirmed by treatment with PI3K inhibitor BEZ235. Up-regulation of Lyn promoted the expression of p-Akt and Cyclin D1. Additionally, we investigated the effects of Lyn inhibitor Bafetinib on melanoma cells and the results were consistent with Lyn knockdown. Collectively, our results indicated that Lyn plays a carcinogenic role in multiple cellular functions during melanoma development through regulating apoptosis and autophagy via the PI3K/Akt pathway and may be a valuable potential target for the clinical treatment of melanoma.
Collapse
Affiliation(s)
- Qianqian Zhang
- Department of Dermatology, Jinan Central Hospital affiliated to Shandong University, Jinan, Shandong Province, China
- Taishan Medical University, Taian, Shandong Province, China
- Department of Dermatology, No. 960 Hospital of The Chinese People's Liberation Army, Taian, Shandong Province, China
| | - Xianguang Meng
- Department of Dermatology, Jinan Central Hospital affiliated to Shandong University, Jinan, Shandong Province, China
| | - Guojing Qin
- Department of Dermatology, Jinan Central Hospital affiliated to Shandong University, Jinan, Shandong Province, China
| | - Xiaotong Xue
- Department of Dermatology, Jinan Central Hospital affiliated to Shandong University, Jinan, Shandong Province, China
- School of Medicine, Shandong University, Jinan, Shandong Province, China
| | - Ningning Dang
- Department of Dermatology, Jinan Central Hospital affiliated to Shandong University, Jinan, Shandong Province, China
| |
Collapse
|
13
|
Naboulsi I, Aboulmouhajir A, Kouisni L, Bekkaoui F, Yasri A. Combining a QSAR Approach and Structural Analysis to Derive an SAR Map of Lyn Kinase Inhibition. Molecules 2018; 23:E3271. [PMID: 30544914 PMCID: PMC6320833 DOI: 10.3390/molecules23123271] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 11/15/2018] [Accepted: 11/22/2018] [Indexed: 01/11/2023] Open
Abstract
Lyn kinase, a member of the Src family of protein tyrosine kinases, is mainly expressed by various hematopoietic cells, neural and adipose tissues. Abnormal Lyn kinase regulation causes various diseases such as cancers. Thus, Lyn represents, a potential target to develop new antitumor drugs. In the present study, using 176 molecules (123 training set molecules and 53 test set molecules) known by their inhibitory activities (IC50) against Lyn kinase, we constructed predictive models by linking their physico-chemical parameters (descriptors) to their biological activity. The models were derived using two different methods: the generalized linear model (GLM) and the artificial neural network (ANN). The ANN Model provided the best prediction precisions with a Square Correlation coefficient R² = 0.92 and a Root of the Mean Square Error RMSE = 0.29. It was able to extrapolate to the test set successfully (R² = 0.91 and RMSE = 0.33). In a second step, we have analyzed the used descriptors within the models as well as the structural features of the molecules in the training set. This analysis resulted in a transparent and informative SAR map that can be very useful for medicinal chemists to design new Lyn kinase inhibitors.
Collapse
Affiliation(s)
- Imane Naboulsi
- AgroBioSciences Research Division, Mohammed VI Polytechnic University, Lot 660⁻Hay Moulay Rachid, 43150 Ben-Guerir, Morocco.
- Organic Synthesis, Extraction and Valorization Laboratory, Faculty of Sciences Ain Chock, Hassan II University, Km 8 El Jadida Road, 20100 Casablanca, Morocco.
| | - Aziz Aboulmouhajir
- Organic Synthesis, Extraction and Valorization Laboratory, Faculty of Sciences Ain Chock, Hassan II University, Km 8 El Jadida Road, 20100 Casablanca, Morocco.
- Team of Molecular Modeling and Spectroscopy, Faculty of Sciences, Chouaib Doukkali University, 24000 El Jadida, Morocco.
| | - Lamfeddal Kouisni
- AgroBioSciences Research Division, Mohammed VI Polytechnic University, Lot 660⁻Hay Moulay Rachid, 43150 Ben-Guerir, Morocco.
| | - Faouzi Bekkaoui
- AgroBioSciences Research Division, Mohammed VI Polytechnic University, Lot 660⁻Hay Moulay Rachid, 43150 Ben-Guerir, Morocco.
- School of Agriculture, Fertilizer and Environment Sciences, Mohammed VI Polytechnic University, Lot 660 Hay Moulay Rachid, 43150 Ben Guerir, Morocco.
| | - Abdelaziz Yasri
- AgroBioSciences Research Division, Mohammed VI Polytechnic University, Lot 660⁻Hay Moulay Rachid, 43150 Ben-Guerir, Morocco.
| |
Collapse
|
14
|
Xu Y, He Q, Lu Y, Tao F, Zhao L, Ou R. MicroRNA-218-5p inhibits cell growth and metastasis in cervical cancer via LYN/NF-κB signaling pathway. Cancer Cell Int 2018; 18:198. [PMID: 30524205 PMCID: PMC6278036 DOI: 10.1186/s12935-018-0673-1] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2018] [Accepted: 10/31/2018] [Indexed: 01/09/2023] Open
Abstract
Background We are committed to investigate miR-218-5 effects on the progression of cervical cancer (CC) cell and find out the molecular mechanism. Methods GSE9750 was obtained from GEO database and R Limma package was applied to filter out dysregulated genes. The pathways were enriched by GSEA software, ClusterProfiler and enrichplot packages to predict the function of DEGs. The binding sites of LYN were detected by miRanda and TargetScan. The miR2Disease database was used to find miRNAs related with CC. The expression of miR-218-5p and LYN were quantified by qRT-PCR and that of LYN protein was measured by western blot. The targeted relationships between miR-218-5p and LYN were verified by dual-luciferase reporter assay. Colony formation assays, wound healing, transwell invasion assay and flow cytometer analysis were performed to investigate the roles that miR-218-5p and LYN played in migration, invasion and death of cervical carcinoma. Xenografts established in nude mice were used to assess tumor growth in vivo. Results The highly expressed mRNA LYN was selected by microarray analysis in GSE9750. NF-κB signaling pathway was enriched base on GSEA results. The expression of miR-218-5p was lower but LYN was higher in CC primary tumors compared with normal control. In addition, miR-218-5p could regulate the expression of LYN in HeLa cells negatively. Overexpression of LYN could promote cell migration and invasion, but inhibit cell death in vitro, and also promote tumor formation in vivo via activating NF-κB signaling pathway which could be reversed by miR-218-5p. Conclusions MiR-218-5p suppressed the progression of CC via LYN/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Yunsheng Xu
- 1Laboratory for Advanced Interdisciplinary Research, Institutes of Translational Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000 Zhejiang China.,2Department of Dermatovenereology, The Seventh Affiliated Hospital, Sun Yat-sen University, No. 628 Zhenyuan Road, Guangming District, Shenzhen, 518107 Guangdong China
| | - Qin He
- 3Department of Dermatovenereology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000 Zhejiang China
| | - Yiyi Lu
- 3Department of Dermatovenereology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000 Zhejiang China
| | - Fengxing Tao
- 3Department of Dermatovenereology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000 Zhejiang China
| | - Liang Zhao
- 1Laboratory for Advanced Interdisciplinary Research, Institutes of Translational Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000 Zhejiang China
| | - Rongying Ou
- 4Department of Gynaecology and Obstetrics, The First Affiliated Hospital of Wenzhou Medical University, Nanbaixiang Street, Ouhai District, Wenzhou, 325000 Zhejiang China
| |
Collapse
|
15
|
Liu S, Hao X, Ouyang X, Dong X, Yang Y, Yu T, Hu J, Hu L. Tyrosine kinase LYN is an oncotarget in human cervical cancer: A quantitative proteomic based study. Oncotarget 2018; 7:75468-75481. [PMID: 27690342 PMCID: PMC5342753 DOI: 10.18632/oncotarget.12258] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2016] [Accepted: 09/02/2016] [Indexed: 01/05/2023] Open
Abstract
Cervical cancer is one of the most common malignant tumor in women. The mechanisms of cervical cancer are intricate and have not been fully understood. Therefore, we employed iTRAQ to obtain novel proteins profile which participates in the tumor oncogenesis of cervical cancer. 3300 proteins were identified aberrantly expressed in cervical cancer, and western bolt was performed to validate the results of iTRAQ. Then, we selected LYN for further study. Immunohistochemistry identified that LYN expression was significantly increased in cervical cancer tissues than that in cancer adjacent normal cervical tissues and normal cervical tissues. The increased LYN expression was significantly correlated with cancer differentiation and FIGO stage. Silencing LYN inhibited cell proliferation, migration and invasion, conversely, overexpression LYN promoted cell proliferation, migration and invasion. In terms of mechanism, LYN could also promote cervical cancer cells metastasis through activating IL-6/STAT3 pathway. In vivo study, overexpression LYN promoted tumor growth, meanwhile knockdown LYN inhibited tumor growth. These results indicate that LYN tyrosine kinase is an oncogenic gene and can serve as a novel target for cervical cancer research and therapy.
Collapse
Affiliation(s)
- Shuaibin Liu
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, PR China
| | - Xiaoming Hao
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, PR China
| | - Xiaolan Ouyang
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, PR China
| | - Xiaojing Dong
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, PR China
| | - Yixuan Yang
- Department of Infectious Diseases, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, PR China
| | - Tinghe Yu
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, PR China
| | - Jianguo Hu
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, PR China
| | - Lina Hu
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, PR China
| |
Collapse
|
16
|
Hu J, Wang G, Sun T. Dissecting the roles of the androgen receptor in prostate cancer from molecular perspectives. Tumour Biol 2017; 39:1010428317692259. [PMID: 28475016 DOI: 10.1177/1010428317692259] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Androgen receptor plays a pivotal role in prostate cancer progression, and androgen deprivation therapy to intercept androgen receptor signal pathway is an indispensable treatment for most advanced prostate cancer patients to delay cancer progression. However, the emerging of castration-resistant prostate cancer reminds us the alteration of androgen receptor, which includes androgen receptor mutation, the formation of androgen receptor variants, and androgen receptor distribution in cancer cells. In this review, we introduce the process of androgen receptor and also its variants' formation, translocation, and function alteration by protein modification or interaction with other pathways. We dissect the roles of androgen receptor in prostate cancer from molecular perspective to provide clues for battling prostate cancer, especially castration-resistant prostate cancer.
Collapse
Affiliation(s)
- Jieping Hu
- Department of Urology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Gongxian Wang
- Department of Urology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Ting Sun
- Department of Urology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
17
|
Thaper D, Vahid S, Nip KM, Moskalev I, Shan X, Frees S, Roberts ME, Ketola K, Harder KW, Gregory-Evans C, Bishop JL, Zoubeidi A. Targeting Lyn regulates Snail family shuttling and inhibits metastasis. Oncogene 2017; 36:3964-3975. [PMID: 28288135 DOI: 10.1038/onc.2017.5] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Revised: 11/27/2016] [Accepted: 12/27/2016] [Indexed: 02/06/2023]
Abstract
The acquisition of an invasive phenotype by epithelial cells occurs through a loss of cellular adhesion and polarity, heralding a multistep process that leads to metastatic dissemination. Since its characterization in 1995, epithelial-mesenchymal transition (EMT) has been closely linked to the metastatic process. As a defining aspect of EMT, loss of cell adhesion through downregulation of E-cadherin is carried out by several transcriptional repressors; key among them the SNAI family of transcription factors. Here we identify for the first time that Lyn kinase functions as a key modulator of SNAI family protein localization and stability through control of the Vav-Rac1-PAK1 (Vav-Rac1-p21-activated kinase) pathway. Accordingly, targeting Lyn in vitro reduces EMT and in vivo reduces metastasis of primary tumors. We also demonstrate the clinical relevance of targeting Lyn as a key player controlling EMT; patient samples across many cancers revealed a strong negative correlation between Lyn and E-cadherin, and high Lyn expression in metastatic tumors as well as metastasis-prone primary tumors. This work reveals a novel pancancer mechanism of Lyn-dependent control of EMT and further underscores the role of this kinase in tumor progression.
Collapse
Affiliation(s)
- D Thaper
- Department of Urology, Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, Canada.,Faculty of Medicine, Department of Urologic Science, University of British Columbia, Vancouver, BC, Canada
| | - S Vahid
- Department of Urology, Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, Canada.,Faculty of Medicine, Department of Urologic Science, University of British Columbia, Vancouver, BC, Canada
| | - K M Nip
- Department of Urology, Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, Canada.,Faculty of Medicine, Department of Urologic Science, University of British Columbia, Vancouver, BC, Canada
| | - I Moskalev
- Department of Urology, Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, Canada
| | - X Shan
- Faculty of Medicine, Department Ophthalmology and Visual Sciences, University of British Columbia, Vancouver, BC, Canada
| | - S Frees
- Department of Urology, Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, Canada
| | - M E Roberts
- Faculty of Science, Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC, Canada
| | - K Ketola
- Department of Urology, Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, Canada
| | - K W Harder
- Faculty of Science, Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC, Canada
| | - C Gregory-Evans
- Faculty of Medicine, Department Ophthalmology and Visual Sciences, University of British Columbia, Vancouver, BC, Canada
| | - J L Bishop
- Department of Urology, Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, Canada
| | - A Zoubeidi
- Department of Urology, Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, Canada.,Faculty of Medicine, Department of Urologic Science, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
18
|
Leung JK, Sadar MD. Non-Genomic Actions of the Androgen Receptor in Prostate Cancer. Front Endocrinol (Lausanne) 2017; 8:2. [PMID: 28144231 PMCID: PMC5239799 DOI: 10.3389/fendo.2017.00002] [Citation(s) in RCA: 94] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Accepted: 01/05/2017] [Indexed: 12/15/2022] Open
Abstract
Androgen receptor (AR) is a validated drug target for prostate cancer based on its role in proliferation, survival, and metastases of prostate cancer cells. Unfortunately, despite recent improvements to androgen deprivation therapy and the advent of better antiandrogens with a superior affinity for the AR ligand-binding domain (LBD), most patients with recurrent disease will eventually develop lethal metastatic castration-resistant prostate cancer (CRPC). Expression of constitutively active AR splice variants that lack the LBD contribute toward therapeutic resistance by bypassing androgen blockade and antiandrogens. In the canonical pathway, binding of androgen to AR LBD triggers the release of AR from molecular chaperones which enable conformational changes and protein-protein interactions to facilitate its nuclear translocation where it regulates the expression of target genes. However, preceding AR function in the nucleus, initial binding of androgen to AR LBD in the cytoplasm may already initiate signal transduction pathways to modulate cellular proliferation and migration. In this article, we review the significance of signal transduction pathways activated by rapid, non-genomic signaling of the AR during the progression to metastatic CRPC and put into perspective the implications for current and novel therapies that target different domains of AR.
Collapse
Affiliation(s)
- Jacky K. Leung
- Department of Genome Sciences Centre, British Columbia Cancer Agency, Vancouver, BC, Canada
| | - Marianne D. Sadar
- Department of Genome Sciences Centre, British Columbia Cancer Agency, Vancouver, BC, Canada
- *Correspondence: Marianne D. Sadar,
| |
Collapse
|
19
|
Ning P, Zhong JG, Jiang F, Zhang Y, Zhao J, Tian F, Li W. Role of protein S in castration-resistant prostate cancer-like cells. Endocr Relat Cancer 2016; 23:595-607. [PMID: 27342144 DOI: 10.1530/erc-16-0126] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Accepted: 06/24/2016] [Indexed: 01/13/2023]
Abstract
Understanding how castration-resistant prostate cancer (CRPC) cells survive the androgen-deprivation condition is crucial for treatment of this advanced prostate cancer (PCa). Here, we reported for the first time the up-regulation of protein S (PROS), an anticoagulant plasma glycoprotein with multiple biological functions, in androgen-insensitive PCa cells and in experimentally induced castration-resistant PCa cells. Overexpression of exogenous PROS in LNCaP cells reduced androgen deprivation-induced apoptosis and enhanced anchorage-dependent clonogenic ability under androgen deprivation condition. Reciprocally, PROS1 knockdown inhibited cell invasiveness and migration, caused the growth inhibition of castration-resistant tumor xenograft under androgen-depleted conditions, and potentiated Taxol (a widely prescribed anti-neoplastic agent)-mediated cell death in PC3 cells. Furthermore, PROS overexpression significantly stimulated AKT activation but failed to evoke oxidative stress in LNCaP cells under normal condition, suggesting that the malignance-promoting effects of the above-mentioned pathway may occur in the order of oxidative stress/PROS/AKT. The potential mechanism may be due to control of oxidative stress-elicited activation of PI3K-AKT-mTOR pathway. Taken together, our gain-of-function, loss-of-function analyses suggest that PROS may facilitate cell proliferation and promote castration resistance in human castration-resistant PCa-like cells via its apoptosis-regulating property. Future study emphasizing on delineating how PROS regulate cellular processes controlling transformation during the development of castration resistance should open new doors for the development of novel therapeutic targets for CRPC.
Collapse
Affiliation(s)
- Peng Ning
- Department of Histology and EmbryologyFourth Military Medical University, Xi'an, China Department of Tumor Radiotherapy3rd Hospital of PLA, Bao Ji, China
| | - Jia-Guo Zhong
- Section 2 of Department of Surgery42nd Hospital of PLA, Jiajiang County Leshan City, Sichuan, China
| | - Fan Jiang
- Department of Tumor Radiotherapy3rd Hospital of PLA, Bao Ji, China
| | - Yi Zhang
- Department of Tumor Radiotherapy3rd Hospital of PLA, Bao Ji, China
| | - Jie Zhao
- Department of Histology and EmbryologyFourth Military Medical University, Xi'an, China
| | - Feng Tian
- Department of Thoracic SurgeryTangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Wei Li
- Department of Histology and EmbryologyFourth Military Medical University, Xi'an, China
| |
Collapse
|
20
|
Toren P, Kim S, Johnson F, Zoubeidi A. Combined AKT and MEK Pathway Blockade in Pre-Clinical Models of Enzalutamide-Resistant Prostate Cancer. PLoS One 2016; 11:e0152861. [PMID: 27046225 PMCID: PMC4821639 DOI: 10.1371/journal.pone.0152861] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Accepted: 03/20/2016] [Indexed: 11/25/2022] Open
Abstract
Despite recent improvements in patient outcomes using newer androgen receptor (AR) pathway inhibitors, treatment resistance in castrate resistant prostate cancer (CRPC) continues to remain a clinical problem. Co-targeting alternate resistance pathways are of significant interest to treat CRPC and delay the onset of resistance. Both the AKT and MEK signaling pathways become activated as prostate cancer develops resistance to AR-targeted therapies. This pre-clinical study explores co-targeting these pathways in AR-positive prostate cancer models. Using various in vitro models of prostate cancer disease states including androgen dependent (LNCaP), CRPC (V16D and 22RV1) and ENZ-resistant prostate cancer (MR49C and MR49F), we evaluate the relevance of targeting both AKT and MEK pathways. Our data reveal that AKT inhibition induces apoptosis and inhibits cell growth in PTEN null cell lines independently of their sensitivity to hormone therapy; however, AKT inhibition had no effect on the PTEN positive 22RV1 cell line. Interestingly, we found that MEK inhibition had greater effect on 22RV1 cells compared to LNCaP, V16D or ENZ-resistant cells MR49C and MR49F cells. In vitro, combination AKT and MEK blockade had evidence of synergy observed in some cell lines and assays, but this was not consistent across all results. In vivo, the combination of AKT and MEK inhibition resulted in more consistent tumor growth inhibition of MR49F xenografts and longer disease specific survival compared to AKT inhibitor monotherapy. As in our in vitro study, 22RV1 xenografts were more resistant to AKT inhibition while they were more sensitive to MEK inhibition. Our results suggest that targeting AKT and MEK in combination may be a valuable strategy in prostate cancer when both pathways are activated and further support the importance of characterizing the dominant oncogenic pathway in each patient's tumor in order to select optimal therapy.
Collapse
Affiliation(s)
- Paul Toren
- The Vancouver Prostate Centre, Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Soojin Kim
- The Vancouver Prostate Centre, Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Fraser Johnson
- The Vancouver Prostate Centre, Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Amina Zoubeidi
- The Vancouver Prostate Centre, Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
21
|
Chatterji T, Varkaris AS, Parikh NU, Song JH, Cheng CJ, Schweppe RE, Alexander S, Davis JW, Troncoso P, Friedl P, Kuang J, Lin SH, Gallick GE. Yes-mediated phosphorylation of focal adhesion kinase at tyrosine 861 increases metastatic potential of prostate cancer cells. Oncotarget 2016; 6:10175-94. [PMID: 25868388 PMCID: PMC4496348 DOI: 10.18632/oncotarget.3391] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Accepted: 02/16/2015] [Indexed: 01/15/2023] Open
Abstract
To study the role of FAK signaling complexes in promoting metastatic properties of prostate cancer (PCa) cells, we selected stable, highly migratory variants, termed PC3 Mig-3 and DU145 Mig-3, from two well-characterized PCa cell lines, PC3 and DU145. These variants were not only increased migration and invasion in vitro, but were also more metastatic to lymph nodes following intraprostatic injection into nude mice. Both PC3 Mig-3 and DU145 Mig-3 were specifically increased in phosphorylation of FAK Y861. We therefore examined potential alterations in Src family kinases responsible for FAK phosphorylation and determined only Yes expression was increased. Overexpression of Yes in PC3 parental cells and src-/-fyn-/-yes-/- fibroblasts selectively increased FAK Y861 phosphorylation, and increased migration. Knockdown of Yes in PC3 Mig-3 cells decreased migration and decreased lymph node metastasis following orthotopic implantation of into nude mice. In human specimens, Yes expression was increased in lymph node metastases relative to paired primary tumors from the same patient, and increased pFAK Y861 expression in lymph node metastases correlated with poor prognosis. These results demonstrate a unique role for Yes in phosphorylation of FAK and in promoting PCa metastasis. Therefore, phosphorylated FAK Y861 and increased Yes expression may be predictive markers for PCa metastasis.
Collapse
Affiliation(s)
- Tanushree Chatterji
- Department of Genitourinary Medical Oncology, The David Koch Center for Applied Research in Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Programs in Cancer Biology and Cancer Metastasis, The University of Texas Graduate School of Biomedical Sciences at Houston, TX, USA
| | - Andreas S Varkaris
- Department of Genitourinary Medical Oncology, The David Koch Center for Applied Research in Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Nila U Parikh
- Department of Genitourinary Medical Oncology, The David Koch Center for Applied Research in Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jian H Song
- Department of Genitourinary Medical Oncology, The David Koch Center for Applied Research in Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Chien-Jui Cheng
- Department of Pathology, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Department of Pathology, Taipei Medical University Hospital, Taipei Medical University, Taipei, Taiwan
| | - Rebecca E Schweppe
- Division of Endocrinology, Metabolism, and Diabetes, and Department of Pathology, University of Colorado Anschutz Medical Campus, University of Colorado Cancer Center, Aurora, CO, USA
| | - Stephanie Alexander
- Department of Genitourinary Medical Oncology, The David Koch Center for Applied Research in Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Department of Cell Biology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - John W Davis
- Department of Urology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Patricia Troncoso
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Peter Friedl
- Department of Genitourinary Medical Oncology, The David Koch Center for Applied Research in Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Division of Endocrinology, Metabolism, and Diabetes, and Department of Pathology, University of Colorado Anschutz Medical Campus, University of Colorado Cancer Center, Aurora, CO, USA
| | - Jian Kuang
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sue-Hwa Lin
- Department of Genitourinary Medical Oncology, The David Koch Center for Applied Research in Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Programs in Cancer Biology and Cancer Metastasis, The University of Texas Graduate School of Biomedical Sciences at Houston, TX, USA.,Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Gary E Gallick
- Department of Genitourinary Medical Oncology, The David Koch Center for Applied Research in Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Programs in Cancer Biology and Cancer Metastasis, The University of Texas Graduate School of Biomedical Sciences at Houston, TX, USA
| |
Collapse
|
22
|
Liu D. LYN, a Key Gene From Bioinformatics Analysis, Contributes to Development and Progression of Esophageal Adenocarcinoma. Med Sci Monit Basic Res 2015; 21:253-61. [PMID: 26708841 PMCID: PMC4701013 DOI: 10.12659/msmbr.895463] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Background Esophageal adenocarcinoma is a lethal malignancy whose incidence is rapidly growing in recent years. Previous reports suggested that Barrett’s esophagus (BE), which is represented by metaplasia-dysplasia-carcinoma transition, is regarded as the premalignant lesion of esophageal neoplasm. However, our knowledge about the development of esophageal adenocarcinoma is still very limited. Material/Methods In order to acquire better understanding about the pathological mechanisms in this field, we obtained gene profiling data on BE, esophageal adenocarcinoma patients, and normal controls from the Gene Expression Omnibus (GEO) database. Bioinformatics analyses, including Gene Ontology (GO) analysis and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis, were conducted. Results Our results revealed that several pathways, such as the wound healing, complement, and coagulation pathways, were closely correlated with cancer development and progression. The mitogen-activated protein kinase (MAPK) pathway was discovered to be responsible for the predisposition stage of cancer; while response to stress, cytokine-cytokine receptor interaction, nod-like receptor signaling pathway, and ECM-receptor interaction were chief contributors of cancer progression. More importantly, we discovered in this study that LYN was a critical gene. It was found to be the key nodule of several significant biological networks, which suggests its close correlation with cancer initiation and progression. Conclusions These results provided more information on the mechanisms of esophageal adenocarcinoma, which enlightened our way to the clinical discovery of novel therapeutic makers for conquering esophageal cancer. Keywords: esophageal adenocarcinoma; LYN; Go analysis; KEGG pathway.
Collapse
Affiliation(s)
- Dabiao Liu
- Department of Clinical Laboratory, Zhenjiang No. 4 Hospital, Zhenjiang, Jiangsu, China (mainland)
| |
Collapse
|
23
|
Shah K, Bradbury NA. Kinase modulation of androgen receptor signaling: implications for prostate cancer. ACTA ACUST UNITED AC 2015; 2. [PMID: 28580371 DOI: 10.14800/ccm.1023] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Androgens and androgen receptors play essential roles in the development and progression of prostate cancer, a disease that claims roughly 28,000 lives annually. In addition to androgen biding, androgen receptor activity can be regulated via several post-translational modifications such as ubiquitination, acetylation, phosphorylation, methylation & SUMO-ylation. Off these modifications, phosphorylation has been the most extensively studied. Modification by phosphorylation can alter androgen receptor localization, protein stability and transcriptional activity, ultimately leading to changes in the biology of cancer cells and cancer progression. Understanding, role of phosphorylated androgen receptor species holds the key to identifying a potential therapeutic drug target for patients with prostate cancer and castrate resistant prostate cancer. Here, we present a brief review of recently discovered protein kinases phosphorylating AR, focusing on the functional role of phosphorylated androgen receptor species in prostate cancer and castrate resistant prostate cancer.
Collapse
Affiliation(s)
- Kalpit Shah
- Department of Physiology and Biophysics, The Chicago Medical School, Rosalind Franklin University of Medicine & Sciences, North Chicago, IL, 60064, USA
| | - Neil A Bradbury
- Department of Physiology and Biophysics, The Chicago Medical School, Rosalind Franklin University of Medicine & Sciences, North Chicago, IL, 60064, USA
| |
Collapse
|
24
|
Mello AA, Leal MF, Rey JA, Pinto GR, Lamarão LM, Montenegro RC, Alves APNN, Assumpção PP, Borges BDN, Smith MC, Burbano RR. Deregulated Expression of SRC, LYN and CKB Kinases by DNA Methylation and Its Potential Role in Gastric Cancer Invasiveness and Metastasis. PLoS One 2015; 10:e0140492. [PMID: 26460485 PMCID: PMC4604160 DOI: 10.1371/journal.pone.0140492] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Accepted: 09/25/2015] [Indexed: 12/29/2022] Open
Abstract
Kinases are downstream modulators and effectors of several cellular signaling cascades and play key roles in the development of neoplastic disease. In this study, we aimed to evaluate SRC, LYN and CKB protein and mRNA expression, as well as their promoter methylation, in gastric cancer. We found elevated expression of SRC and LYN kinase mRNA and protein but decreased levels of CKB kinase, alterations that may have a role in the invasiveness and metastasis of gastric tumors. Expression of the three studied kinases was also associated with MYC oncogene expression, a possible biomarker for gastric cancer. To understand the mechanisms that regulate the expression of these genes, we evaluated the DNA promoter methylation of the three kinases. We found that reduced SRC and LYN methylation and increased CKB methylation was associated with gastric cancer. The reduced SRC and LYN methylation was associated with increased levels of mRNA and protein expression, suggesting that DNA methylation is involved in regulating the expression of these kinases. Conversely, reduced CKB methylation was observed in samples with reduced mRNA and protein expression, suggesting CKB expression was found to be only partly regulated by DNA methylation. Additionally, we found that alterations in the DNA methylation pattern of the three studied kinases were also associated with the gastric cancer onset, advanced gastric cancer, deeper tumor invasion and the presence of metastasis. Therefore, SRC, LYN and CKB expression or DNA methylation could be useful markers for predicting tumor progression and targeting in anti-cancer strategies.
Collapse
Affiliation(s)
- Adriano Azevedo Mello
- Centro de Ciências Biológicas e da Saúde, Universidade Federal de Campina Grande, Campina Grande, PB, Brazil
| | - Mariana Ferreira Leal
- Disciplina de Genética, Departamento de Morfologia e Genética, Universidade Federal de São Paulo, São Paulo, SP, Brazil
- Departamento de Ortopedia e Traumatologia, Universidade Federal de São Paulo, São Paulo, SP, Brazil
- * E-mail:
| | - Juan Antonio Rey
- Laboratorio de Oncogenética Molecular, Hospital Universitario La Paz, Madrid, Madrid, Spain
| | | | - Leticia Martins Lamarão
- Laboratório de Testes de Ácidos Nucleicos, Fundação Centro de Hemoterapia e Hematologia do Pará, Belém, PA, Brazil
- Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, PA, Brazil
| | | | | | - Paulo Pimentel Assumpção
- Núcleo de Pesquisa em Oncologia, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém, PA, Brazil
| | - Barbara do Nascimento Borges
- Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, PA, Brazil
- Centro de Tecnologia Agropecuária, Instituto Socioambiental e dos Recursos Hídricos, Universidade Federal Rural da Amazônia, Belém, PA, Brazil
| | - Marília Cardoso Smith
- Disciplina de Genética, Departamento de Morfologia e Genética, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Rommel Rodriguez Burbano
- Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, PA, Brazil
- Núcleo de Pesquisa em Oncologia, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém, PA, Brazil
| |
Collapse
|