1
|
Zhang C, Quan Y, Yang L, Bai Y, Yang Y. 6-Methoxyflavone induces S-phase arrest through the CCNA2/CDK2/p21CIP1 signaling pathway in HeLa cells. Bioengineered 2022; 13:7277-7292. [PMID: 35246013 PMCID: PMC8973872 DOI: 10.1080/21655979.2022.2047496] [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] [Indexed: 11/17/2022] Open
Abstract
This study aimed to elucidate the specific anticancer mechanism of 6-methoxyflavone in HeLa cells. A total of 178 putative targets of 6-methoxyflavone were obtained from the PharmMapper database. Microarray analyses, transcriptome sequencing analyses, functional enrichment analyses, and gene set enrichment analyses were performed to preliminarily explore the roles and mechanisms of the 178 targets in cervical cancer. Cell counting kit-8, cell cycle assays, polymerase chain reactions, and western blotting were used to clarify the mechanism of action of 6-methoxyflavone. Molecular docking and noncovalent interaction analyses were performed to further confirm the mechanism of action in three-dimensional structures. Functional enrichment analyses and gene set enrichment analyses indicated that high mRNA expression of cyclin A2 (CCNA2) and cyclin-dependent kinase 2 (CDK2) stimulated cell cycle progression in cervical cancer. Cell proliferation and cycle assays, transcriptome sequencing, polymerase chain reactions, and western blotting revealed that 6-methoxyflavone inhibited HeLa cell proliferation and induced S-phase arrest via the CCNA2/CDK2/ cyclin-dependent kinase inhibitor 1A (p21CIP1) pathway. Molecular docking and noncovalent interaction analyses showed that 6-methoxyflavone had the strongest affinity toward, inhibitory effect on, and noncovalent interactions with CDK2, and that the combination of CDK2 and CCNA2 enhanced these effects. An analysis of clinical characteristics showed that 6-methoxyflavone might be related to six clinicopathological parameters of cervical cancer patients. 6-Methoxyflavone induces S-phase arrest in HeLa cells via the CCNA2/CDK2/p21CIP1 pathway.
Collapse
Affiliation(s)
- Chaihong Zhang
- The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu, China.,Key Laboratory of Gynecological Oncology of Gansu Province, Lanzhou, Gansu, China
| | - Yuchong Quan
- College of Basic Medicine, Dalian Medical University, Dalian, Liaoning, China
| | - Lijuan Yang
- The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu, China.,Key Laboratory of Gynecological Oncology of Gansu Province, Lanzhou, Gansu, China
| | - Yingying Bai
- The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu, China.,Key Laboratory of Gynecological Oncology of Gansu Province, Lanzhou, Gansu, China
| | - Yongxiu Yang
- The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu, China.,Key Laboratory of Gynecological Oncology of Gansu Province, Lanzhou, Gansu, China.,Department of Obstetrics and Gynecology, First Hospital of Lanzhou University, Lanzhou, Gansu, China
| |
Collapse
|
2
|
Gu X, Zhang S, Zhang T. Abnormal Crosstalk between Endothelial Cells and Podocytes Mediates Tyrosine Kinase Inhibitor (TKI)-Induced Nephrotoxicity. Cells 2021; 10:cells10040869. [PMID: 33921219 PMCID: PMC8070074 DOI: 10.3390/cells10040869] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 04/04/2021] [Accepted: 04/09/2021] [Indexed: 12/12/2022] Open
Abstract
Vascular endothelial growth factor A (VEGFA) and its receptor VEGFR2 are the main targets of antiangiogenic therapies, and proteinuria is one of the common adverse events associated with the inhibition of the VEGFA/VEGFR2 pathway. The proteinuric kidney damage induced by VEGFR2 tyrosine kinase inhibitors (TKIs) is characterized by podocyte foot process effacement. TKI therapy promotes the formation of abnormal endothelial‒podocyte crosstalk, which plays a key role in TKI-induced podocyte injury and proteinuric nephropathy. This review article summarizes the underlying mechanism by which the abnormal endothelial‒podocyte crosstalk mediates podocyte injury and discusses the possible molecules and signal pathways involved in abnormal endothelial‒podocyte crosstalk. What is more, we highlight the molecules involved in podocyte injury and determine the essential roles of Rac1 and Cdc42; this provides evidence for exploring the abnormal endothelial‒podocyte crosstalk in TKI-induced nephrotoxicity.
Collapse
Affiliation(s)
| | | | - Ti Zhang
- Correspondence: ; Tel.: +86-21-6417-5590
| |
Collapse
|
3
|
Jonnalagadda B, Arockiasamy S, Krishnamoorthy S. Cellular growth factors as prospective therapeutic targets for combination therapy in androgen independent prostate cancer (AIPC). Life Sci 2020; 259:118208. [PMID: 32763294 DOI: 10.1016/j.lfs.2020.118208] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Revised: 07/27/2020] [Accepted: 08/02/2020] [Indexed: 12/21/2022]
Abstract
Cancer is the second leading cause of death worldwide, with prostate cancer, the second most commonly diagnosed cancer among men. Prostate cancer develops in the peripheral zone of the prostate gland, and the initial progression largely depends on androgens, the male reproductive hormone that regulates the growth and development of the prostate gland and testis. The currently available treatments for androgen dependent prostate cancer are, however, effective for a limited period, where the patients show disease relapse, and develop androgen-independent prostate cancer (AIPC). Studies have shown various intricate cellular processes such as, deregulation in multiple biochemical and signaling pathways, intra-tumoral androgen synthesis; AR over-expression and mutations and AR activation via alternative growth pathways are involved in progression of AIPC. The currently approved treatment strategies target a single cellular protein or pathway, where the cells slowly develop resistance and adapt to proliferate via other cellular pathways over a period of time. Therefore, an increased research aims to understand the efficacy of combination therapy, which targets multiple interlinked pathways responsible for acquisition of resistance and survival. The combination therapy is also shown to enhance efficacy as well as reduce toxicity of the drugs. Thus, the present review focuses on the signaling pathways involved in the progression of AIPC, comprising a heterogeneous population of cells and the advantages of combination therapy. Several clinical and pre-clinical studies on a variety of combination treatments have shown beneficial outcomes, yet further research is needed to understand the potential of combination therapy and its diverse strategies.
Collapse
Affiliation(s)
- Bhavana Jonnalagadda
- Department of Biomedical Sciences, Sri Ramachandra Institute of Higher Education and Research, Chennai, India
| | - Sumathy Arockiasamy
- Department of Biomedical Sciences, Sri Ramachandra Institute of Higher Education and Research, Chennai, India.
| | - Sriram Krishnamoorthy
- Department of Urology, Sri Ramachandra Medical Centre, Sri Ramachandra Institute of Higher Education and Research, Chennai, India
| |
Collapse
|
4
|
Bhatia K, Bhumika, Das A. Combinatorial drug therapy in cancer - New insights. Life Sci 2020; 258:118134. [PMID: 32717272 DOI: 10.1016/j.lfs.2020.118134] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 07/18/2020] [Accepted: 07/20/2020] [Indexed: 12/19/2022]
Abstract
Cancer can arise due to mutations in numerous pathways present in our body and thus has many alternatives for getting aggravated. Due to this attribute, it gets difficult to treat cancer patients with monotherapy alone and has a risk of not being eliminated to the full extent. This necessitates the introduction of combinatorial therapy as it employs cancer treatment using more than one method and shows a greater success rate. Combinatorial therapy involves a complementary combination of two different therapies like a combination of radio and immunotherapy or a combination of drugs that can target more than one pathway of cancer formation like combining CDK targeting drugs with Growth factors targeting drugs. In this review, we discuss the various aspects of cancer which include, its causes; four regulatory mechanisms namely: apoptosis, cyclin-dependent kinases, tumor suppressor genes, and growth factors; some of the pathways involved; treatment: monotherapy and combinatorial therapy and combinatorial drug formulation in chemotherapy. The present review gives a holistic account of the different mechanisms of therapies and also drug combinations that may serve to not only complement the monotherapy but can also surpass the resistance against monotherapy agents.
Collapse
Affiliation(s)
- Karanpreet Bhatia
- Department of Biotechnology, Delhi Technological University, Main Bawana Road, Delhi 110042, India
| | - Bhumika
- Department of Biotechnology, Delhi Technological University, Main Bawana Road, Delhi 110042, India
| | - Asmita Das
- Department of Biotechnology, Delhi Technological University, Main Bawana Road, Delhi 110042, India.
| |
Collapse
|
5
|
Sun Z, Wang L, Zhou Y, Dong L, Ma W, Lv L, Zhang J, Wang X. Glioblastoma Stem Cell-Derived Exosomes Enhance Stemness and Tumorigenicity of Glioma Cells by Transferring Notch1 Protein. Cell Mol Neurobiol 2019; 40:767-784. [PMID: 31853695 DOI: 10.1007/s10571-019-00771-8] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 12/03/2019] [Indexed: 02/05/2023]
Abstract
Exosomes contain plenty of bioactive information, playing an important role in intercellular communication by transfer their bioactive molecular contents to recipient cells. Glioblastoma stem cells (GSCs) and non-GSC glioma cells coexist in GBM microenvironment; GSC-released exosomes contain intracellular signaling molecules, which may affect the biological phenotypes of recipient cells. However, whether GSC exosomes could affect the biological phenotype of non-GSC glioma cells has not yet been defined. To explore whether GSC exosomes could reprogramme non-GSC glioma cells into GSCs and its possible mechanism involved, non-GSC glioma cells were treated with GSCs released exosomes; the potential mechanisms of action were studied with RNA interference, Notch inhibitors and Western blot analysis. The proliferation, neurosphere formation, invasive capacities, and tumorigenicity of non-GSC glioma cells were increased significantly after GSC exosome treatment; Notch1 signaling pathway was activated in GSCs; Notch1 protein was highly enriched in GSC exosomes; Notch1 signaling pathway and stemness-related protein expressions were increased in GSC exosome treated non-GSC glioma cells and these cell generated tumor tissues; Notch1 protein expression in GSCs and their exosomes, and the neurosphere formation of GSCs were decreased by Notch1 RNA interference; Notch1 signaling pathway protein and stemness protein expressions were decreased in GSC exosome treated non-GSC glioma cells by Notch1 RNA interference and Notch inhibitors. The findings in this study indicated that GSC exosomes act as information carriers, mediated non-GSC glioma cell dedifferentiation into GSCs by delivering Notch1 protein through Notch1 signaling activation, and enhanced stemness and tumorigenicity of non-GSC glioma cells.
Collapse
Affiliation(s)
- Zhen Sun
- Laboratory of Experimental Oncology, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, West China Clinical Medical School, Sichuan University, No. 1 Keyuan Road 4, Gaopeng Avenu, Hi-tech Zone, Chengdu, 610041, China
| | - Li Wang
- Laboratory of Experimental Oncology, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, West China Clinical Medical School, Sichuan University, No. 1 Keyuan Road 4, Gaopeng Avenu, Hi-tech Zone, Chengdu, 610041, China
| | - Yueling Zhou
- Laboratory of Experimental Oncology, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, West China Clinical Medical School, Sichuan University, No. 1 Keyuan Road 4, Gaopeng Avenu, Hi-tech Zone, Chengdu, 610041, China
| | - Lihua Dong
- Human Anatomy Department, School of Preclinical and Forensic Medcine, Sichuan University, Chengdu, 610041, China
| | - Weichao Ma
- Neurosurgery Department, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Liang Lv
- Neurosurgery Department, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jie Zhang
- Laboratory of Experimental Oncology, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, West China Clinical Medical School, Sichuan University, No. 1 Keyuan Road 4, Gaopeng Avenu, Hi-tech Zone, Chengdu, 610041, China
| | - Xiujie Wang
- Laboratory of Experimental Oncology, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, West China Clinical Medical School, Sichuan University, No. 1 Keyuan Road 4, Gaopeng Avenu, Hi-tech Zone, Chengdu, 610041, China.
| |
Collapse
|
6
|
Transcriptomic and Epigenomic Profiling of Histone Deacetylase Inhibitor Treatment Reveals Distinct Gene Regulation Profiles Leading to Impaired Neutrophil Development. Hemasphere 2019; 3:e270. [PMID: 31723844 PMCID: PMC6745919 DOI: 10.1097/hs9.0000000000000270] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Revised: 05/16/2019] [Accepted: 05/16/2019] [Indexed: 01/08/2023] Open
Abstract
Supplemental Digital Content is available in the text The clinical use of histone deacetylase inhibitors (HDACi) for the treatment of bone marrow failure and hematopoietic malignancies has increased dramatically over the last decades. Nonetheless, their effects on normal myelopoiesis remain poorly evaluated. Here, we treated cord blood derived CD34+ progenitor cells with two chemically distinct HDACi inhibitors MS-275 or SAHA and analyzed their effects on the transcriptome (RNA-seq), epigenome (H3K27ac ChIP-seq) and functional and morphological characteristics during neutrophil development. MS-275 (entinostat) selectively inhibits class I HDACs, with a preference for HDAC1, while SAHA (vorinostat) is a non-selective class I/II HDACi. Treatment with individual HDACi resulted in both overlapping and distinct effects on both transcriptome and epigenome, whereas functional effects were relatively similar. Both HDACi resulted in reduced expansion and increased apoptosis in neutrophil progenitor cells. Morphologically, HDACi disrupted normal neutrophil differentiation what was illustrated by decreased percentages of mature neutrophils. In addition, while SAHA treatment clearly showed a block at the promyelocytic stage, MS-275 treatment was characterized by dysplastic features and skewing towards the monocytic lineage. These effects could be mimicked using shRNA-mediated knockdown of HDAC1. Taken together, our data provide novel insights into the effects of HDAC inhibition on normal hematopoietic cells during neutrophil differentiation. These findings should be taken into account when considering the clinical use of MS-275 and SAHA, and can be potentially utilized to tailor more specific, hematopoietic-directed HDACi in the future.
Collapse
|
7
|
Combination of Anti-Cancer Drugs with Molecular Chaperone Inhibitors. Int J Mol Sci 2019; 20:ijms20215284. [PMID: 31652993 PMCID: PMC6862641 DOI: 10.3390/ijms20215284] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Revised: 10/21/2019] [Accepted: 10/22/2019] [Indexed: 12/15/2022] Open
Abstract
Most molecular chaperones belonging to heat shock protein (HSP) families are known to protect cancer cells from pathologic, environmental and pharmacological stress factors and thereby can hamper anti-cancer therapies. In this review, we present data on inhibitors of the heat shock response (particularly mediated by the chaperones HSP90, HSP70, and HSP27) either as a single treatment or in combination with currently available anti-cancer therapeutic approaches. An overview of the current literature reveals that the co-administration of chaperone inhibitors and targeting drugs results in proteotoxic stress and violates the tumor cell physiology. An optimal drug combination should simultaneously target cytoprotective mechanisms and trigger the imbalance of the tumor cell physiology.
Collapse
|
8
|
Tran LNK, Kichenadasse G, Sykes PJ. Combination Therapies Using Metformin and/or Valproic Acid in Prostate Cancer: Possible Mechanistic Interactions. Curr Cancer Drug Targets 2018; 19:368-381. [PMID: 30039761 DOI: 10.2174/1568009618666180724111604] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 05/25/2018] [Accepted: 07/10/2018] [Indexed: 12/22/2022]
Abstract
Prostate cancer (PCa) is the most frequent cancer in men. The evolution from local PCa to castration-resistant PCa, an end-stage of disease, is often associated with changes in genes such as p53, androgen receptor, PTEN, and ETS gene fusion products. Evidence is accumulating that repurposing of metformin (MET) and valproic acid (VPA) either when used alone, or in combination, with another therapy, could potentially play a role in slowing down PCa progression. This review provides an overview of the application of MET and VPA, both alone and in combination with other drugs for PCa treatment, correlates the responses to these drugs with common molecular changes in PCa, and then describes the potential for combined MET and VPA as a systemic therapy for prostate cancer, based on potential interacting mechanisms.
Collapse
Affiliation(s)
- Linh N K Tran
- Flinders Centre for Innovation in Cancer, Flinders University and Medical Centre, Bedford Park, Adelaide, Australia.,University of Medicine and Pharmacy at Ho Chi Minh City, Vietnam
| | - Ganessan Kichenadasse
- Flinders Centre for Innovation in Cancer, Flinders University and Medical Centre, Bedford Park, Adelaide, Australia
| | - Pamela J Sykes
- Flinders Centre for Innovation in Cancer, Flinders University and Medical Centre, Bedford Park, Adelaide, Australia
| |
Collapse
|
9
|
Zhang J, Zhang Y, Cheng L, Li C, Dai L, Zhang H, Yan F, Shi H, Dong G, Ning Z, Xu W, Si C, Deng H, Xiong H. Enrichment and characterization of cancer stem-like cells in ultra-low concentration of serum and non-adhesive culture system. Am J Transl Res 2018; 10:1552-1561. [PMID: 29887968 PMCID: PMC5992539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2018] [Accepted: 04/21/2018] [Indexed: 06/08/2023]
Abstract
Cancer stem cells (CSCs) play important roles in tumor initiation, metastasis, and progression. They are also mainly responsible for high treatment failure rates. Identification and characterization of CSCs are crucial for facilitating the detection, prevention, or therapy of cancer. Great efforts have been paid to develop an effective method and the ideal method for CSCs research is still in the going. In our study, we created an ultra-low concentration of serum and non-adhesive (ULCSN) culture system to enrich CSCs from murine lewis lung cancer cell line LL/2 with cell spheres structure and characterize the LL/2 CSCs properties. Their characteristics were investigated through colony formation, spheres formation, chemoresistance, flow cytometry for putative stem cell markers, such as CD133, CD34 and CD45, immunofluorescence staining and tumor initiation capacity in vivo. Tumor spheres were formed within 7-10 days under the condition of ULCSN culture system. Compared with adherent parental LL/2 cells, the colony capacity, chemo-resistance, and expression of stem cell markers increased significantly in addition to tumor-initiating capability in the tumor sphere cells. Using the ULCSN culture system, an available isolation method of lewis lung CSCs was established, which is simple, effective, and inexpensive compared with the cytokines attachment serum free culture method. The stem cell properties of the tumor sphere LL/2 cells reflected the CSCs phenotypes. We developed a useful CSCs model for basic and pre-clinical studies for lung cancer and other types of cancer.
Collapse
Affiliation(s)
- Junfeng Zhang
- Institute of Immunology and Molecular Medicine, Jining Medical UniversityJining 272067, Shandong, China
- State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan UniversityChengdu, Sichuan, China
| | - Yunsheng Zhang
- Institute of Immunology and Molecular Medicine, Jining Medical UniversityJining 272067, Shandong, China
| | - Lin Cheng
- State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan UniversityChengdu, Sichuan, China
| | - Chunlei Li
- State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan UniversityChengdu, Sichuan, China
- School of Pharmacy, Linyi UniversityLinyi 276000, Shandong, China
| | - Lei Dai
- State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan UniversityChengdu, Sichuan, China
| | - Hui Zhang
- Institute of Immunology and Molecular Medicine, Jining Medical UniversityJining 272067, Shandong, China
| | - Fenglian Yan
- Institute of Immunology and Molecular Medicine, Jining Medical UniversityJining 272067, Shandong, China
| | - Hui Shi
- Institute of Immunology and Molecular Medicine, Jining Medical UniversityJining 272067, Shandong, China
| | - Guanjun Dong
- Institute of Immunology and Molecular Medicine, Jining Medical UniversityJining 272067, Shandong, China
| | - Zhaochen Ning
- Institute of Immunology and Molecular Medicine, Jining Medical UniversityJining 272067, Shandong, China
| | - Wei Xu
- Shenzhen SiBiono GeneTech Co. Ltd.Shenzhen, China
| | - Chuanping Si
- Institute of Immunology and Molecular Medicine, Jining Medical UniversityJining 272067, Shandong, China
| | - Hongxin Deng
- State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan UniversityChengdu, Sichuan, China
| | - Huabao Xiong
- Institute of Immunology and Molecular Medicine, Jining Medical UniversityJining 272067, Shandong, China
- Department of Medicine, Immunology Institute, Icahn School of Medicine at Mount SinaiNew York, NY 10029, USA
| |
Collapse
|
10
|
El-Maqsoud NMRA, Osman NAA, El-Hamid AMA, El-Bab TKF, Galal EM. GOLPH3 and YB-1 Are Novel Markers Correlating With Poor Prognosis in Prostate Cancer. World J Oncol 2015; 6:473-484. [PMID: 28983350 PMCID: PMC5624675 DOI: 10.14740/wjon952w] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/14/2015] [Indexed: 02/06/2023] Open
Abstract
Background Prostate cancer is a common and aggressive cancer among men. Despite advances in the treatment, the mechanisms involved in progression are still unclear. New prognostic markers should be explored for better design of patient-specific therapeutic regimens. Methods This study was performed on 120 patients stratified as 76 with prostatic carcinoma, 12 with low-grade prostate intraepithelial lesion, 12 with high-grade prostate intraepithelial lesion and 20 with benign prostate hyperplasia. Immunohistochemical study was done for Golgi phosphoprotein 3 (GOLPH3) and Y-box binding protein-1 (YB-1) analysis. Correlation with clinicopathological data and overall survival was analyzed. Results Both GOLPH3 and YB-1 showed increased expression from benign to malignant tumors. In prostatic carcinoma, cytoplasmic GOLPH3 was associated with Gleason score, stage and androgen receptor (P = 0.034, P < 0.001, and P = 0.008 respectively). Nuclear YB-1 expression was associated with Gleason score and androgen receptor (P = 0.018 and P = 0.024 respectively). Cytoplasmic YB-1 expression was associated with Gleason score, stage and androgen receptor (P = 0.008, P = 0.027, and P < 0.001 respectively). High Gleason score (P = 0.004), high stage (P < 0.001) and androgen receptor (P = 0.006) were the only detected adverse prognostic clinicopathological factors. Moderate/intense GOLPH3 and high nuclear and cytoplasmic YB-1 expression were correlated with shorter overall survival (P < 0.001, P = 0.020, and P < 0.001 respectively). In the multivariate analysis, moderate/intense GOLPH3 expression was the only predictor of overall survival (P = 0.025). Conclusions High GOLPH3 and nuclear/cytoplasmic YB-1 expression correlated with poor prognosis in prostate cancer. Both markers can be promising targets for new treatment strategies.
Collapse
|
11
|
Abd El-Maqsoud NMR, Osman NAA, Abd El-Hamid AMA, Fath El-Bab TK, Galal EM. Golgi Phosphoprotein-3 and Y-Box-Binding Protein-1 Are Novel Markers Correlating With Poor Prognosis in Prostate Cancer. Clin Genitourin Cancer 2015; 14:e143-52. [PMID: 26794392 DOI: 10.1016/j.clgc.2015.12.015] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Revised: 12/13/2015] [Accepted: 12/16/2015] [Indexed: 12/18/2022]
Abstract
BACKGROUND Prostate cancer is a common and aggressive cancer among men. Despite advances in treatment, the mechanisms involved in progression are still unclear. New prognostic markers are needed to better design patient-specific therapeutic regimens. MATERIALS AND METHODS The present study included 120 patients: 76 with prostate carcinoma, 12 with low-grade prostate intraepithelial lesions, 12 with high-grade prostate intraepithelial lesions, and 20 with benign prostatic hyperplasia. Immunohistochemical study was performed for Golgi phosphoprotein-3 (GOLPH3) and Y-box-binding protein-1 (YB-1) analysis. The correlation with clinicopathologic data and overall survival was analyzed. RESULTS Both GOLPH3 and YB-1 showed increased expression from benign to malignant tumors. In prostate carcinoma, cytoplasmic GOLPH3 was associated with Gleason score, tumor stage, and androgen receptor status (P = .034, P < .001, and P = .008, respectively). Nuclear YB-1 expression was associated with Gleason score and androgen receptor status (P = .018 and P = .024, respectively). Cytoplasmic YB-1 expression was associated with Gleason score, tumor stage, and androgen receptor status (P = .008, P = .027, and P < .001, respectively). A high Gleason score (P = .004), high tumor stage (P < .001), and androgen receptor-independent cancer (P = .006) were the only detected adverse prognostic clinicopathologic factors. Moderate to intense GOLPH3 and high nuclear and cytoplasmic YB-1 expression correlated with shorter overall survival (P < .001, P = .020, and P < .001, respectively). On multivariate analysis, moderate to intense GOLPH3 expression was the only predictor of overall survival (P = .025). CONCLUSION High GOLPH3 and nuclear/cytoplasmic YB-1 expression correlated with a poor prognosis in patients with prostate cancer. Both markers could be promising targets for new treatment strategies.
Collapse
Affiliation(s)
| | - Nisreen A A Osman
- Department of Pathology, Faculty of Medicine, Minia University, Minia, Egypt
| | | | | | - Ehab M Galal
- Department of Urology, Faculty of Medicine, Minia University, Minia, Egypt
| |
Collapse
|
12
|
Berghauser Pont LM, Kleijn A, Kloezeman JJ, van den Bossche W, Kaufmann JK, de Vrij J, Leenstra S, Dirven CM, Lamfers ML. The HDAC Inhibitors Scriptaid and LBH589 Combined with the Oncolytic Virus Delta24-RGD Exert Enhanced Anti-Tumor Efficacy in Patient-Derived Glioblastoma Cells. PLoS One 2015; 10:e0127058. [PMID: 25993039 PMCID: PMC4436250 DOI: 10.1371/journal.pone.0127058] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2015] [Accepted: 04/10/2015] [Indexed: 01/12/2023] Open
Abstract
Background A phase I/II trial for glioblastoma with the oncolytic adenovirus Delta24-RGD was recently completed. Delta24-RGD conditionally replicates in cells with a disrupted retinoblastoma-pathway and enters cells via αvβ3/5 integrins. Glioblastomas are differentially sensitive to Delta24-RGD. HDAC inhibitors (HDACi) affect integrins and share common cell death pathways with Delta24-RGD. We studied the combination treatment effects of HDACi and Delta24-RGD in patient-derived glioblastoma stem-like cells (GSC), and we determined the most effective HDACi. Methods SAHA, Valproic Acid, Scriptaid, MS275 and LBH589 were combined with Delta24-RGD in fourteen distinct GSCs. Synergy was determined by Chou Talalay method. Viral infection and replication were assessed using luciferase and GFP encoding vectors and hexon-titration assays. Coxsackie adenovirus receptor and αvβ3 integrin levels were determined by flow cytometry. Oncolysis and mechanisms of cell death were studied by viability, caspase-3/7, LDH and LC3B/p62, phospho-p70S6K. Toxicity was studied on normal human astrocytes. MGMT promotor methylation status, TCGA classification, Rb-pathway and integrin gene expression levels were assessed as markers of responsiveness. Results Scriptaid and LBH589 acted synergistically with Delta24-RGD in approximately 50% of the GSCs. Both drugs moderately increased αvβ3 integrin levels and viral infection in responding but not in non-responding GSCs. LBH589 moderately increased late viral gene expression, however, virus titration revealed diminished viral progeny production by both HDACi, Scriptaid augmented caspase-3/7 activity, LC3B conversion, p62 and phospho-p70S6K consumption, as well as LDH levels. LBH589 increased LDH and phospho-p70S6K consumption. Responsiveness correlated with expression of various Rb-pathway genes and integrins. Combination treatments induced limited toxicity to human astrocytes. Conclusion LBH589 and Scriptaid combined with Delta24-RGD revealed synergistic anti-tumor activity in a subset of GSCs. Both HDACi moderately augmented viral infection and late gene expression, but slightly reduced progeny production. The drugs differentially activated multiple cell death pathways. The limited toxicity on astrocytes supports further evaluation of the proposed combination therapies.
Collapse
Affiliation(s)
| | - Anne Kleijn
- Department of Neurosurgery, Brain Tumor Center, Erasmus MC, Rotterdam, The Netherlands
| | - Jenneke J. Kloezeman
- Department of Neurosurgery, Brain Tumor Center, Erasmus MC, Rotterdam, The Netherlands
| | | | - Johanna K. Kaufmann
- Department of Neurosurgery, Harvey Cushing Neuro-Oncology Laboratories, Brigham & Women’s Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Jeroen de Vrij
- Department of Neurosurgery, Utrecht University Medical Center, Utrecht, The Netherlands
| | - Sieger Leenstra
- Department of Neurosurgery, Brain Tumor Center, Erasmus MC, Rotterdam, The Netherlands
- Department of Neurosurgery, Elisabeth Hospital, Tilburg, The Netherlands
| | - Clemens M.F. Dirven
- Department of Neurosurgery, Brain Tumor Center, Erasmus MC, Rotterdam, The Netherlands
| | - Martine L.M. Lamfers
- Department of Neurosurgery, Brain Tumor Center, Erasmus MC, Rotterdam, The Netherlands
- * E-mail:
| |
Collapse
|
13
|
Tsaur I, Hudak L, Makarević J, Juengel E, Mani J, Borgmann H, Gust KM, Schilling D, Bartsch G, Nelson K, Haferkamp A, Blaheta RA. Intensified antineoplastic effect by combining an HDAC-inhibitor, an mTOR-inhibitor and low dosed interferon alpha in prostate cancer cells. J Cell Mol Med 2015; 19:1795-804. [PMID: 25808196 PMCID: PMC4549030 DOI: 10.1111/jcmm.12583] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Accepted: 02/25/2015] [Indexed: 12/14/2022] Open
Abstract
A significant proportion of men diagnosed with prostate cancer (PCa) eventually develop metastatic disease, which progresses to castration resistance, despite initial response to androgen deprivation. As anticancer therapy has become increasingly effective, acquired drug resistance has emerged, limiting efficacy. Combination treatment, utilizing different drug classes, exemplifies a possible strategy to foil resistance development. The effects of the triple application of the histone deacetylase (HDAC) inhibitor valproic acid (VPA), the mammalian target of rapamycin inhibitor everolimus and low dosed interferon alpha (IFNα) on PCa cell growth and dissemination capacity were investigated. For that purpose, the human PCa cell lines, PC-3, DU-145 and LNCaP were treated with the combined regimen or separate single agents. Cell growth was investigated by the MTT dye reduction assay. Flow cytometry served to analyse cell cycle progression. Adhesion to vascular endothelium or immobilized collagen, fibronectin and laminin was quantified. Migration and invasion characteristics were determined by the modified Boyden chamber assay. Integrin α and β subtypes were investigated by flow cytometry, western blotting and RT-PCR. Integrin related signalling, Epidermal Growth Factor Receptor (EGFr), Akt, p70S6kinase and extracellular signal-regulated kinases (ERK)1/2 activation were also assessed. The triple application of VPA, everolimus and low dosed IFNα blocked tumour cell growth and dissemination significantly better than any agent alone. Antitumour effects were associated with pronounced alteration in the cell cycle machinery, intracellular signalling and integrin expression profile. Combining VPA, everolimus and low dosed IFNα might be a promising option to counteract resistance development and improve outcome in PCa patients.
Collapse
Affiliation(s)
- Igor Tsaur
- Department of Urology, Johann Wolfgang Goethe-University, Frankfurt am Main, Germany
| | - Lukasz Hudak
- Department of Urology, Johann Wolfgang Goethe-University, Frankfurt am Main, Germany
| | - Jasmina Makarević
- Department of Urology, Johann Wolfgang Goethe-University, Frankfurt am Main, Germany
| | - Eva Juengel
- Department of Urology, Johann Wolfgang Goethe-University, Frankfurt am Main, Germany
| | - Jens Mani
- Department of Urology, Johann Wolfgang Goethe-University, Frankfurt am Main, Germany
| | - Hendrik Borgmann
- Department of Urology, Johann Wolfgang Goethe-University, Frankfurt am Main, Germany
| | - Kilian M Gust
- Department of Urology, Johann Wolfgang Goethe-University, Frankfurt am Main, Germany
| | - David Schilling
- Department of Urology, Johann Wolfgang Goethe-University, Frankfurt am Main, Germany
| | - Georg Bartsch
- Department of Urology, Johann Wolfgang Goethe-University, Frankfurt am Main, Germany
| | - Karen Nelson
- Department of Vascular and Endovascular Surgery, Johann Wolfgang Goethe-University, Frankfurt am Main, Germany
| | - Axel Haferkamp
- Department of Urology, Johann Wolfgang Goethe-University, Frankfurt am Main, Germany
| | - Roman A Blaheta
- Department of Urology, Johann Wolfgang Goethe-University, Frankfurt am Main, Germany
| |
Collapse
|
14
|
Juengel E, Nowaz S, Makarevi J, Natsheh I, Werner I, Nelson K, Reiter M, Tsaur I, Mani J, Harder S, Bartsch G, Haferkamp A, Blaheta RA. HDAC-inhibition counteracts everolimus resistance in renal cell carcinoma in vitro by diminishing cdk2 and cyclin A. Mol Cancer 2014; 13:152. [PMID: 24935000 PMCID: PMC4073177 DOI: 10.1186/1476-4598-13-152] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Accepted: 06/03/2014] [Indexed: 02/06/2023] Open
Abstract
Background Targeted therapies have improved therapeutic options of treating renal cell carcinoma (RCC). However, drug response is temporary due to resistance development. Methods Functional and molecular changes in RCC Caki-1 cells, after acquired resistance to the mammalian target of rapamycin (mTOR)-inhibitor everolimus (Cakires), were investigated with and without additional application of the histone deacetylase (HDAC)-inhibitor valproic acid (VPA). Cell growth was evaluated by MTT assay, cell cycle progression and apoptosis by flow cytometry. Target molecules of everolimus and VPA, apoptotic and cell cycle regulating proteins were investigated by western blotting. siRNA blockade was performed to evaluate the functional relevance of the proteins. Results Everolimus resistance was accompanied by significant increases in the percentage of G2/M-phase cells and in the IC50. Akt and p70S6K, targets of everolimus, were activated in Cakires compared to drug sensitive cells. The most prominent change in Cakires cells was an increase in the cell cycle activating proteins cdk2 and cyclin A. Knock-down of cdk2 and cyclin A caused significant growth inhibition in the Cakires cells. The HDAC-inhibitor, VPA, counteracted everolimus resistance in Cakires, evidenced by a significant decrease in tumor growth and cdk2/cyclin A. Conclusion It is concluded that non-response to everolimus is characterized by increased cdk2/cyclin A, driving RCC cells into the G2/M-phase. VPA hinders everolimus non-response by diminishing cdk2/cyclin A. Therefore, treatment with HDAC-inhibitors might be an option for patients with advanced renal cell carcinoma and acquired everolimus resistance.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | - Roman A Blaheta
- Department of Urology, Goethe-University, Interdisciplinary Science Building, Building 25A, Room 404, Theodor-Stern-Kai 7, Frankfurt / Main D-60590, Germany.
| |
Collapse
|
15
|
Wang L, Guo H, Lin C, Yang L, Wang X. Enrichment and characterization of cancer stem‑like cells from a cervical cancer cell line. Mol Med Rep 2014; 9:2117-23. [PMID: 24676900 PMCID: PMC4055449 DOI: 10.3892/mmr.2014.2063] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2013] [Accepted: 02/19/2014] [Indexed: 02/05/2023] Open
Abstract
Cancer stem cells (CSCs) are proposed to be responsible for tumor recurrence, metastasis and the high mortality rate of cancer patients. Isolation and identification of CSCs is crucial for basic and preclinical studies. However, as there are currently no universal markers for the isolation and identification of CSCs in any type of cancer, the method for isolating CSCs from primary cancer tissues or cell lines is costly and ineffective. In order to establish a reliable model of cervical cancer stem cells for basic and preclinical studies, the present study was designed to enrich cervical cancer CSCs using a nonadhesive culture system and to characterize their partial stemness phenotypes. Human cervical cancer cells (HeLa) were cultured using a nonadhesive culture system to generate tumor spheres. Their stemness characteristics were investigated through colony formation, tumor sphere formation, self-renewal, toluidine blue staining, chemoresistance, invasion assays, reverse transcription-polymerase chain reaction, immunofluorescence staining of putative stem cell markers, including octamer-binding transcription factor 4, SRY-box 2 and aldehyde dehydrogenase 1 family, member A1, and adipogenic differentiation induction. Typical tumor spheres were formed within 5–7 days under this nonadhesive culture system. Compared with the adherent parental HeLa cells, the colony formation capacity, self-renewal potential, light cell population, cell invasion, chemoresistance and expression of putative stem cell markers of the tumor sphere cells increased significantly, and a subpopulation of tumor sphere cells were induced into adipogenic differentiation. Using the nonadhesive culture system, a reliable model of cervical cancer stem cells was established, which is inexpensive, effective and simple compared with the ultra-low attachment serum free culture method. The stemness characteristics of the tumor sphere HeLa cells mirrored the CSC phenotypes. This CSC model may be useful for basic and preclinical studies of cervical cancer and other types of cancer.
Collapse
Affiliation(s)
- Li Wang
- Laboratory of Experimental Oncology, State Key Laboratory of Biotherapy, West China Hospital, West China Clinical Medical School, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Huijie Guo
- Laboratory of Experimental Oncology, State Key Laboratory of Biotherapy, West China Hospital, West China Clinical Medical School, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Caiyu Lin
- Laboratory of Experimental Oncology, State Key Laboratory of Biotherapy, West China Hospital, West China Clinical Medical School, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Liuqi Yang
- Laboratory of Experimental Oncology, State Key Laboratory of Biotherapy, West China Hospital, West China Clinical Medical School, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Xiujie Wang
- Laboratory of Experimental Oncology, State Key Laboratory of Biotherapy, West China Hospital, West China Clinical Medical School, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| |
Collapse
|
16
|
Li J, Li C, Han J, Zhang C, Shang D, Yao Q, Zhang Y, Xu Y, Liu W, Zhou M, Yang H, Su F, Li X. The detection of risk pathways, regulated by miRNAs, via the integration of sample-matched miRNA-mRNA profiles and pathway structure. J Biomed Inform 2014; 49:187-97. [PMID: 24561483 DOI: 10.1016/j.jbi.2014.02.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2013] [Revised: 12/17/2013] [Accepted: 02/03/2014] [Indexed: 11/26/2022]
Abstract
The use of genome-wide, sample-matched miRNA (miRNAs)-mRNA expression data provides a powerful tool for the investigation of miRNAs and genes involved in diseases. The identification of miRNA-regulated pathways has been crucial for analysis of the role of miRNAs. However, the classical identification method fails to consider the structural information of pathways and the regulation of miRNAs simultaneously. We proposed a method that simultaneously integrated the change in gene expression and structural information in order to identify pathways. Our method used fold changes in miRNAs and gene products, along with the quantification of the regulatory effect on target genes, to measure the change in gene expression. Topological characteristics were investigated to measure the influence of gene products on entire pathways. Through the analysis of multiple myeloma and prostate cancer expression data, our method was proven to be effective and reliable in identifying disease risk pathways that are regulated by miRNAs. Further analysis showed that the structure of a pathway plays a crucial role in the recognition of the pathway as a factor in disease risk.
Collapse
Affiliation(s)
- Jing Li
- College of Bioinformatics Science and Technology and Bio-pharmaceutical Key Laboratory of Heilongjiang Province, Harbin Medical University, Harbin 150081, PR China; Department of Bioinformatics, School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350004, PR China
| | - Chunquan Li
- College of Bioinformatics Science and Technology and Bio-pharmaceutical Key Laboratory of Heilongjiang Province, Harbin Medical University, Harbin 150081, PR China
| | - Junwei Han
- College of Bioinformatics Science and Technology and Bio-pharmaceutical Key Laboratory of Heilongjiang Province, Harbin Medical University, Harbin 150081, PR China
| | - Chunlong Zhang
- College of Bioinformatics Science and Technology and Bio-pharmaceutical Key Laboratory of Heilongjiang Province, Harbin Medical University, Harbin 150081, PR China
| | - Desi Shang
- College of Bioinformatics Science and Technology and Bio-pharmaceutical Key Laboratory of Heilongjiang Province, Harbin Medical University, Harbin 150081, PR China
| | - Qianlan Yao
- College of Bioinformatics Science and Technology and Bio-pharmaceutical Key Laboratory of Heilongjiang Province, Harbin Medical University, Harbin 150081, PR China
| | - Yunpeng Zhang
- College of Bioinformatics Science and Technology and Bio-pharmaceutical Key Laboratory of Heilongjiang Province, Harbin Medical University, Harbin 150081, PR China
| | - Yanjun Xu
- College of Bioinformatics Science and Technology and Bio-pharmaceutical Key Laboratory of Heilongjiang Province, Harbin Medical University, Harbin 150081, PR China
| | - Wei Liu
- College of Bioinformatics Science and Technology and Bio-pharmaceutical Key Laboratory of Heilongjiang Province, Harbin Medical University, Harbin 150081, PR China
| | - Meng Zhou
- College of Bioinformatics Science and Technology and Bio-pharmaceutical Key Laboratory of Heilongjiang Province, Harbin Medical University, Harbin 150081, PR China
| | - Haixiu Yang
- College of Bioinformatics Science and Technology and Bio-pharmaceutical Key Laboratory of Heilongjiang Province, Harbin Medical University, Harbin 150081, PR China
| | - Fei Su
- College of Bioinformatics Science and Technology and Bio-pharmaceutical Key Laboratory of Heilongjiang Province, Harbin Medical University, Harbin 150081, PR China
| | - Xia Li
- College of Bioinformatics Science and Technology and Bio-pharmaceutical Key Laboratory of Heilongjiang Province, Harbin Medical University, Harbin 150081, PR China.
| |
Collapse
|
17
|
Jiang W, Zheng Y, Huang Z, Wang M, Zhang Y, Wang Z, Jin X, Xia Q. Role of SMAD4 in the mechanism of valproic acid’s inhibitory effect on prostate cancer cell invasiveness. Int Urol Nephrol 2013; 46:941-6. [DOI: 10.1007/s11255-013-0609-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2013] [Accepted: 11/09/2013] [Indexed: 01/20/2023]
|
18
|
Chen L, Li BQ, Zheng MY, Zhang J, Feng KY, Cai YD. Prediction of effective drug combinations by chemical interaction, protein interaction and target enrichment of KEGG pathways. BIOMED RESEARCH INTERNATIONAL 2013; 2013:723780. [PMID: 24083237 PMCID: PMC3780555 DOI: 10.1155/2013/723780] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/08/2013] [Accepted: 07/24/2013] [Indexed: 12/11/2022]
Abstract
Drug combinatorial therapy could be more effective in treating some complex diseases than single agents due to better efficacy and reduced side effects. Although some drug combinations are being used, their underlying molecular mechanisms are still poorly understood. Therefore, it is of great interest to deduce a novel drug combination by their molecular mechanisms in a robust and rigorous way. This paper attempts to predict effective drug combinations by a combined consideration of: (1) chemical interaction between drugs, (2) protein interactions between drugs' targets, and (3) target enrichment of KEGG pathways. A benchmark dataset was constructed, consisting of 121 confirmed effective combinations and 605 random combinations. Each drug combination was represented by 465 features derived from the aforementioned three properties. Some feature selection techniques, including Minimum Redundancy Maximum Relevance and Incremental Feature Selection, were adopted to extract the key features. Random forest model was built with its performance evaluated by 5-fold cross-validation. As a result, 55 key features providing the best prediction result were selected. These important features may help to gain insights into the mechanisms of drug combinations, and the proposed prediction model could become a useful tool for screening possible drug combinations.
Collapse
Affiliation(s)
- Lei Chen
- Institute of Systems Biology, Shanghai University, Shanghai 200444, China
- College of Information Engineering, Shanghai Maritime University, Shanghai 201306, China
| | - Bi-Qing Li
- Key Laboratory of Systems Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Ming-Yue Zheng
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Shanghai 201203, China
| | - Jian Zhang
- Department of Ophthalmology, Shanghai First People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200080, China
| | - Kai-Yan Feng
- Beijing Genomics Institute, Shenzhen Beishan Industrial Zone, Shenzhen 518083, China
| | - Yu-Dong Cai
- Institute of Systems Biology, Shanghai University, Shanghai 200444, China
| |
Collapse
|
19
|
Abstract
The phosphatidylinositol 3-kinase (PI3K)/Akt/mammalian target of rapamycin (mTOR) pathway is a key signaling pathway that has been linked to both tumorigenesis and resistance to therapy in prostate cancer and other solid tumors. Given the significance of the PI3K/Akt/mTOR pathway in integrating cell survival signals and the high prevalence of activating PI3K/Akt/mTOR pathway alterations in prostate cancer, inhibitors of this pathway have great potential for clinical benefit. Here, we review the role of the PI3K/Akt/mTOR pathway in prostate cancer and discuss the potential use of pathway inhibitors as single agents or in combination in the evolving treatment landscape of castration-resistant prostate cancer.
Collapse
Affiliation(s)
- Rhonda L Bitting
- Division of Medical Oncology, Duke Cancer Institute, Duke University, DUMC Box 102002, Durham, North Carolina 27710, USA
| | | |
Collapse
|
20
|
Sharma NL, Groselj B, Hamdy FC, Kiltie AE. The emerging role of histone deacetylase (HDAC) inhibitors in urological cancers. BJU Int 2013; 111:537-42. [PMID: 23551441 DOI: 10.1111/j.1464-410x.2012.11647.x] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
WHAT'S KNOWN ON THE SUBJECT? AND WHAT DOES THE STUDY ADD?: A growing body of evidence supports the anti-cancer effect of histone deacetylase inhibitors (HDACi) in vitro, via multiple pathways, and many Phase I clinical trials have shown them to be well-tolerated in a range of malignancies. Combined therapies, including with radiation, present an exciting area of current and planned study. This review summarises the evidence to date, including pre-clinical data and clinical trials, of the anti-cancer effect of HDACi in urological cancers. It provides an overview of epigenetics and the mechanisms of action of HDACi. It suggests areas of future development, including the current challenges for the successful introduction of HDACi into clinical therapy. Epigenetic modifications are known to play a critical role in the development and progression of many cancers. The opposing actions of histone deacetylases (HDACs) and histone acetyltransferases (HATs) modify chromatin and lead to epigenetic gene regulation, in addition to wider effects on non-histone proteins. There is growing interest in the clinical application of HDAC inhibitors (HDACi) in cancer. HDACi have been shown to inhibit cancer cell growth both in vitro and in vivo and recent clinical trials have shown encouraging results in various urological cancers. In this review, we discuss the existing evidence and potential role for HDACi in urological malignancies, including in combined therapies.
Collapse
Affiliation(s)
- Naomi L Sharma
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK.
| | | | | | | |
Collapse
|
21
|
Evidence from clinical trials for the use of valproic acid in solid tumors: focus on prostate cancer. ACTA ACUST UNITED AC 2013. [DOI: 10.4155/cli.13.23] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
22
|
Liu X, Chen L, Sun F, Zhang G. Enhanced suppression of proliferation and migration in highly-metastatic lung cancer cells by combination of valproic acid and coumarin-3-carboxylic acid and its molecular mechanisms of action. Cytotechnology 2012; 65:597-608. [PMID: 23161221 DOI: 10.1007/s10616-012-9513-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2012] [Accepted: 10/18/2012] [Indexed: 01/23/2023] Open
Abstract
Valproic acid (VPA) as a broad-spectrum inhibitor of histone deacetylase, has been used in cancer therapy. Recently, the combination of VPA with other anticancer agents has been considered as a useful and necessary strategy to inhibit tumor growth and progression. The coumarin derivates from natural plants have been shown to be the promising natural anticancer agents. However, no literature is available on the anticancer effects of the combination of VPA and coumarin-3-carboxylic acid (HCCA). Here we show that this combination significantly increases inhibitory effects against the proliferation and migration in highly-metastatic lung cancer cells by inducing apoptosis and cell cycle arrest as well as regulating related protein expressions. Our results indicate that this combination of VPA with HCCA not only enhances the protein levels of Bax, cytosolic cytochrome c, caspase-3 and PARP-1 but also reduces the protein expressions of Bcl-2, cyclin D1 and NF-κB as well as inhibits the phosphorylation and expressions of Akt, EGFR, VEGFR2 and c-Met in the cancer cells. Our results suggest that the combination of VPA with HCCA suppresses the proliferation and migration of lung cancer cells via EGFR/VEGFR2/c-Met-Akt-NF-κB signaling pathways; this combination may have a wide therapeutic and/or adjuvant therapeutic application in the treatment of lung cancer.
Collapse
Affiliation(s)
- Xin Liu
- Laboratory of Molecular Pharmacology, School of Pharmacy, Yantai University, No. 30, Qing Quan Lu, Lai Shan Qu, Yantai, Shandong Province, 264005, China
| | | | | | | |
Collapse
|
23
|
RGD-Binding Integrins in Prostate Cancer: Expression Patterns and Therapeutic Prospects against Bone Metastasis. Cancers (Basel) 2012; 4:1106-45. [PMID: 24213501 PMCID: PMC3712721 DOI: 10.3390/cancers4041106] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2012] [Revised: 10/09/2012] [Accepted: 10/22/2012] [Indexed: 12/26/2022] Open
Abstract
Prostate cancer is the third leading cause of male cancer deaths in the developed world. The current lack of highly specific detection methods and efficient therapeutic agents for advanced disease have been identified as problems requiring further research. The integrins play a vital role in the cross-talk between the cell and extracellular matrix, enhancing the growth, migration, invasion and metastasis of cancer cells. Progression and metastasis of prostate adenocarcinoma is strongly associated with changes in integrin expression, notably abnormal expression and activation of the β3 integrins in tumour cells, which promotes haematogenous spread and tumour growth in bone. As such, influencing integrin cell expression and function using targeted therapeutics represents a potential treatment for bone metastasis, the most common and debilitating complication of advanced prostate cancer. In this review, we highlight the multiple ways in which RGD-binding integrins contribute to prostate cancer progression and metastasis, and identify the rationale for development of multi-integrin antagonists targeting the RGD-binding subfamily as molecularly targeted agents for its treatment.
Collapse
|
24
|
Increased expression of Golgi phosphoprotein-3 is associated with tumor aggressiveness and poor prognosis of prostate cancer. Diagn Pathol 2012; 7:127. [PMID: 23006319 PMCID: PMC3488508 DOI: 10.1186/1746-1596-7-127] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2012] [Accepted: 08/10/2012] [Indexed: 12/22/2022] Open
Abstract
Background To investigate the expression of Golgi phosphoprotein-3 (GOLPH3) in prostate cancer and determine its prognostic value. Methods Immunohistochemical staining for GOLPH3 was performed on tissue microarrays of 342 prostate patients. The correlation between GOLPH3 expression with its clinicopathologic factors was also analyzed in order to determine its prognostic significance. Results GOLPH3 expression of normal prostate tissues, benign prostate hyperplasia, high-grade prostatic intraepithelial neoplasia, and hormone-dependent prostate cancer (HDPC) did not show any statistically significant difference. In contrast, statistically significant difference was reported in moderate/intense GOLPH3 expression in cases diagnosed with HDPC and castration resistant prostate cancer (CRPC) (P < 0.0005). Moderate /intense expression of GOLPH3 was associated with androgen independence (P = 0.012), higher Gleason score (P = 0.017), bone metastasis (P = 0.024), higher baseline prostate-specific antigen (PSA) (P = 0.038), and higher PSA nadir (P = 0.032). A significantly negative correlation was found between moderate/intense GOLPH3 expression and disease-free survival (DFS) (HR = 0.28, P = 0.012) and overall survival (OS) (HR = 0.42, P = 0.027). Univariated analysis indicated that moderate/intense GOLPH3 expression created a significantly prognostic impact in patients with CRPC. On the other hand, multivariate analysis indicated that GOLPH3 was a significantly independent prognostic factor of DFS (P = 0.027) in all prostate cancer patients. Conclusions In this study, it was discovered that the overexpression of GOLPH3 is associated with the transition of prostate cancer from hormone sensitive phase to hormone refractory phase. GOLPH3 might be an important prognostic factor of DFS and OS in patients with prostate cancer. In totality, GOLPH3 could be used as a novel candidate in devising a more effective therapeutic strategy to tackle CRPC. Virtual slides The virtual slide(s) for this article can be found here: http://www.diagnosticpathology.diagnomx.eu/vs/1452541171722856.
Collapse
|