151
|
Sun J, Yan J, Yuan X, Yang R, Dan T, Wang X, Kong G, Gao S. A computationally constructed ceRNA interaction network based on a comparison of the SHEE and SHEEC cell lines. Cell Mol Biol Lett 2016; 21:21. [PMID: 28536623 PMCID: PMC5415789 DOI: 10.1186/s11658-016-0022-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Accepted: 09/10/2016] [Indexed: 12/16/2022] Open
Abstract
Long non-coding RNAs (lncRNAs) play critical and complicated roles in the regulation of various biological processes, including chromatin modification, transcription and post-transcriptional processing. Interestingly, some lncRNAs serve as miRNA "sponges" that inhibit interaction with miRNA targets in post-transcriptional regulation. We constructed a putative competing endogenous RNA (ceRNA) network by integrating lncRNA, miRNA and mRNA expression based on high-throughput RNA sequencing and microarray data to enable a comparison of the SHEE and SHEEC cell lines. Using Targetscan and miRanda bioinformatics algorithms and miRTarbase microRNA-target interactions database, we established that 51 miRNAs sharing 13,623 MREs with 2260 genes and 82 lncRNAs were involved in this ceRNA network. Through a biological function analysis, the ceRNA network appeared to be primarily involved in cell proliferation, apoptosis, the cell cycle, invasion and metastasis. Functional pathway analyses demonstrated that the ceRNA network potentially modulated multiple signaling pathways, such as the MAPK, Ras, HIF-1, Rap1, and PI3K/Akt signaling pathways. These results might provide new clues to better understand the regulation of the ceRNA network in cancer.
Collapse
Affiliation(s)
- Jiachun Sun
- Department of Oncology, Cancer Institute, First Affiliated Hospital of Henan University of Science and Technology, Luoyang, People’s Republic of China
| | - Junqiang Yan
- Department of Neurology, Cancer Institute, First Affiliated Hospital of Henan University of Science and Technology, Luoyang, People’s Republic of China
| | - Xiaozhi Yuan
- Department of Oncology, Cancer Institute, First Affiliated Hospital of Henan University of Science and Technology, Luoyang, People’s Republic of China
| | - Ruina Yang
- Department of Oncology, Cancer Institute, First Affiliated Hospital of Henan University of Science and Technology, Luoyang, People’s Republic of China
| | - Tanyou Dan
- Department of Oncology, Cancer Institute, First Affiliated Hospital of Henan University of Science and Technology, Luoyang, People’s Republic of China
| | - Xinshuai Wang
- Department of Oncology, Cancer Institute, First Affiliated Hospital of Henan University of Science and Technology, Luoyang, People’s Republic of China
| | - Guoqiang Kong
- Department of Oncology, Cancer Institute, First Affiliated Hospital of Henan University of Science and Technology, Luoyang, People’s Republic of China
| | - Shegan Gao
- Department of Oncology, Cancer Institute, First Affiliated Hospital of Henan University of Science and Technology, Luoyang, People’s Republic of China
| |
Collapse
|
152
|
Li Y, Chen H, Pan T, Jiang C, Zhao Z, Wang Z, Zhang J, Xu J, Li X. LncRNA ontology: inferring lncRNA functions based on chromatin states and expression patterns. Oncotarget 2016; 6:39793-805. [PMID: 26485761 PMCID: PMC4741861 DOI: 10.18632/oncotarget.5794] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Accepted: 09/05/2015] [Indexed: 02/01/2023] Open
Abstract
Accumulating evidences suggest that long non-coding RNAs (lncRNAs) perform important functions. Genome-wide chromatin-states area rich source of information about cellular state, yielding insights beyond what is typically obtained by transcriptome profiling. We propose an integrative method for genome-wide functional predictions of lncRNAs by combining chromatin states data with gene expression patterns. We first validated the method using protein-coding genes with known function annotations. Our validation results indicated that our integrative method performs better than co-expression analysis, and is accurate across different conditions. Next, by applying the integrative model genome-wide, we predicted the probable functions for more than 97% of human lncRNAs. The putative functions inferred by our method match with previously annotated by the targets of lncRNAs. Moreover, the linkage from the cellular processes influenced by cancer-associated lncRNAs to the cancer hallmarks provided a “lncRNA point-of-view” on tumor biology. Our approach provides a functional annotation of the lncRNAs, which we developed into a web-based application, LncRNA Ontology, to provide visualization, analysis, and downloading of lncRNA putative functions.
Collapse
Affiliation(s)
- Yongsheng Li
- College of Bioinformatics Science and Technology and Bio-Pharmaceutical Key Laboratory of Heilongjiang Province, Harbin Medical University, Nangang, Harbin, Heilongjiang, China
| | - Hong Chen
- College of Bioinformatics Science and Technology and Bio-Pharmaceutical Key Laboratory of Heilongjiang Province, Harbin Medical University, Nangang, Harbin, Heilongjiang, China
| | - Tao Pan
- College of Bioinformatics Science and Technology and Bio-Pharmaceutical Key Laboratory of Heilongjiang Province, Harbin Medical University, Nangang, Harbin, Heilongjiang, China
| | - Chunjie Jiang
- College of Bioinformatics Science and Technology and Bio-Pharmaceutical Key Laboratory of Heilongjiang Province, Harbin Medical University, Nangang, Harbin, Heilongjiang, China
| | - Zheng Zhao
- College of Bioinformatics Science and Technology and Bio-Pharmaceutical Key Laboratory of Heilongjiang Province, Harbin Medical University, Nangang, Harbin, Heilongjiang, China
| | - Zishan Wang
- College of Bioinformatics Science and Technology and Bio-Pharmaceutical Key Laboratory of Heilongjiang Province, Harbin Medical University, Nangang, Harbin, Heilongjiang, China
| | - Jinwen Zhang
- College of Bioinformatics Science and Technology and Bio-Pharmaceutical Key Laboratory of Heilongjiang Province, Harbin Medical University, Nangang, Harbin, Heilongjiang, China
| | - Juan Xu
- College of Bioinformatics Science and Technology and Bio-Pharmaceutical Key Laboratory of Heilongjiang Province, Harbin Medical University, Nangang, Harbin, Heilongjiang, China
| | - Xia Li
- College of Bioinformatics Science and Technology and Bio-Pharmaceutical Key Laboratory of Heilongjiang Province, Harbin Medical University, Nangang, Harbin, Heilongjiang, China
| |
Collapse
|
153
|
Al Faraj A. SWCNTs as novel theranostic nanocarriers for cancer diagnosis and therapy: towards safe translation to the clinics. Nanomedicine (Lond) 2016; 11:1431-45. [DOI: 10.2217/nnm-2016-0065] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
With their unique physicochemical properties, single walled carbon nanotubes (SWCNTs) hold great promise for applications as drug delivery systems (DDS) for early and better diagnosis and therapy of cancer. While several in vitro and in vivo studies have validated their potential benefit, no SWCNT-based formulation has yet reached clinical trials. Towards prospective safe clinical applications, the main properties that were adopted to enhance the biocompatibility of SWCNTs were highlighted. Then, the recent progresses in the in vivo applications of SWCNTs as diagnostic nanoprobes using multimodality imaging techniques and as therapeutic nanocarriers delivering wide range of anticancer efficient drugs to tumors were reviewed. Finally, the efforts required for safe clinical applications of SWCNTs as DDS for cancer diagnosis and therapy were discussed.
Collapse
Affiliation(s)
- Achraf Al Faraj
- Department of Radiological Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
154
|
The Philadelphia chromosome in leukemogenesis. CHINESE JOURNAL OF CANCER 2016; 35:48. [PMID: 27233483 PMCID: PMC4896164 DOI: 10.1186/s40880-016-0108-0] [Citation(s) in RCA: 109] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/25/2015] [Accepted: 05/03/2016] [Indexed: 02/07/2023]
Abstract
The truncated chromosome 22 that results from the reciprocal translocation t(9;22)(q34;q11) is known as the Philadelphia chromosome (Ph) and is a hallmark of chronic myeloid leukemia (CML). In leukemia cells, Ph not only impairs the physiological signaling pathways but also disrupts genomic stability. This aberrant fusion gene encodes the breakpoint cluster region-proto-oncogene tyrosine-protein kinase (BCR-ABL1) oncogenic protein with persistently enhanced tyrosine kinase activity. The kinase activity is responsible for maintaining proliferation, inhibiting differentiation, and conferring resistance to cell death. During the progression of CML from the chronic phase to the accelerated phase and then to the blast phase, the expression patterns of different BCR-ABL1 transcripts vary. Each BCR-ABL1 transcript is present in a distinct leukemia phenotype, which predicts both response to therapy and clinical outcome. Besides CML, the Ph is found in acute lymphoblastic leukemia, acute myeloid leukemia, and mixed-phenotype acute leukemia. Here, we provide an overview of the clinical presentation and cellular biology of different phenotypes of Ph-positive leukemia and highlight key findings regarding leukemogenesis.
Collapse
|
155
|
Wang F, Zhang N, Wang J, Wu H, Zheng X. Tumor purity and differential methylation in cancer epigenomics. Brief Funct Genomics 2016; 15:408-419. [PMID: 27199459 DOI: 10.1093/bfgp/elw016] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
DNA methylation is an epigenetic modification of DNA molecule that plays a vital role in gene expression regulation. It is not only involved in many basic biological processes, but also considered an important factor for tumorigenesis and other human diseases. Study of DNA methylation has been an active field in cancer epigenomics research. With the advances of high-throughput technologies and the accumulation of enormous amount of data, method development for analyzing these data has gained tremendous interests in the fields of computational biology and bioinformatics. In this review, we systematically summarize the recent developments of computational methods and software tools in high-throughput methylation data analysis with focus on two aspects: differential methylation analysis and tumor purity estimation in cancer studies.
Collapse
|
156
|
An Oncogenic Virus Promotes Cell Survival and Cellular Transformation by Suppressing Glycolysis. PLoS Pathog 2016; 12:e1005648. [PMID: 27187079 PMCID: PMC4871371 DOI: 10.1371/journal.ppat.1005648] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Accepted: 04/29/2016] [Indexed: 11/19/2022] Open
Abstract
Aerobic glycolysis is essential for supporting the fast growth of a variety of cancers. However, its role in the survival of cancer cells under stress conditions is unclear. We have previously reported an efficient model of gammaherpesvirus Kaposi’s sarcoma-associated herpesvirus (KSHV)-induced cellular transformation of rat primary mesenchymal stem cells. KSHV-transformed cells efficiently induce tumors in nude mice with pathological features reminiscent of Kaposi’s sarcoma tumors. Here, we report that KSHV promotes cell survival and cellular transformation by suppressing aerobic glycolysis and oxidative phosphorylation under nutrient stress. Specifically, KSHV microRNAs and vFLIP suppress glycolysis by activating the NF-κB pathway to downregulate glucose transporters GLUT1 and GLUT3. While overexpression of the transporters rescues the glycolytic activity, it induces apoptosis and reduces colony formation efficiency in softagar under glucose deprivation. Mechanistically, GLUT1 and GLUT3 inhibit constitutive activation of the AKT and NF-κB pro-survival pathways. Strikingly, GLUT1 and GLUT3 are significantly downregulated in KSHV-infected cells in human KS tumors. Furthermore, we have detected reduced levels of aerobic glycolysis in several KSHV-infected primary effusion lymphoma cell lines compared to a Burkitt’s lymphoma cell line BJAB, and KSHV infection of BJAB cells reduced aerobic glycolysis. These results reveal a novel mechanism by which an oncogenic virus regulates a key metabolic pathway to adapt to stress in tumor microenvironment, and illustrate the importance of fine-tuning the metabolic pathways for sustaining the proliferation and survival of cancer cells, particularly under stress conditions. KSHV is causally associated with the development of Kaposi’s sarcoma and primary effusion lymphoma; however, the mechanism underlying KSHV-induced malignant transformation remains unclear. The recent development of an efficient KSHV-induced cellular transformation model of primary rat mesenchymal stem cells should facilitate the delineation of KSHV-induced oncogenesis. In this report, we have used this model to investigate the metabolic pathways mediating the proliferation and survival of KSHV-transformed cells. In contrast to most other cancers that depend on aerobic glycolysis for their fast growth, we demonstrate that KSHV suppresses aerobic glycolysis and oxidative phosphorylation in the transformed cells. Significantly, suppression of aerobic glycolysis enhances the survival of the KSHV-transformed cells under nutrient deprivation. Mechanistically, KSHV-encoded microRNAs and vFLIP suppress aerobic glycolysis by activating the NF-κB pathway to downregulate glucose transporters GLUT1 and GLUT3. We have further shown that GLUT1 and GLUT3 inhibit constitutive activation of the AKT and NF-κB pro-survival pathways. Strikingly, GLUT1 and GLUT3 are significantly downregulated in KSHV-infected cells in human KS tumors. Furthermore, we have detected reduced levels of aerobic glycolysis in several KSHV-infected primary effusion lymphoma cell lines and a KSHV-infected Burkitt’s lymphoma cell line BJAB. Our results reveal a novel mechanism by which an oncogenic virus regulates a key metabolic pathway to adapt to stress in tumor microenvironment, and illustrate the importance of fine-tuning the metabolic pathways for sustaining the proliferation and survival of cancer cells, particularly under nutrient stress microenvironment.
Collapse
|
157
|
Wang L, Chen G, Chen K, Ren Y, Li H, Jiang X, Jia L, Fu S, Li Y, Liu X, Wang S, Yang J, Wu C. Dual targeting of retinoid X receptor and histone deacetylase with DW22 as a novel antitumor approach. Oncotarget 2016; 6:9740-55. [PMID: 25762635 PMCID: PMC4496394 DOI: 10.18632/oncotarget.3149] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Accepted: 01/17/2015] [Indexed: 11/26/2022] Open
Abstract
Retinoid X receptor (RXR) and Histone deacetylase (HDAC) are considered important targets for cancer therapy due to their crucial roles in genetic or epigenetic regulations of cancer development and progression. Here, we evaluated the potential of dual targeting of RXR and HDAC using DW22 as a novel therapeutic approach to cancer treatment. We found that the co-expression of RXR-α and HDAC1 was frequently appeared in lung cancer and breast cancer tissues and cell lines. RXR was activated by DW22 in RXRα and HDAC1 overexpressed A549 and MDA-MB-435 cell lines. Meanwhile, DW22 inhibited the activity of HDAC by decreasing its expression in A549 and MDA-MB-435 cell lines, but not in RXRα and HDAC1 deficient cell lines. Moreover, DW22 suppressed cell growth, induced cell differentiation, prompted cell apoptosis and arrested cell cycle in A549, MDA-MB-435 or HL60 cell lines. Treatment human umbilical vascular endothelial cells (HUVECs) with DW22 suppressed migration, invasion and tube formation through decreasing VEGF expression. The up-regulation of Ac-H3 and p21, and down-regulation of VEGF caused by DW22 was markedly attenuated by silencing of HDAC1. Furthermore, knockdown of RXRα by siRNA completely blocked DW22-induced cell differentiation, but partially attenuated DW22-caused inhibition of cell proliferation, induction of cell apoptosis, and suppression of cell migration, invasion and tube formation. Moreover, intravenous administration of DW22 significantly retarded tumor growth of A549 and MDA-MB-435 xenograft mice models, and induced no substantial weight loss and gross toxicity. In addition, DW22 also reduced cell proliferation, angiogenesis, and induced cell apoptosis in vivo. Collectively, our data demonstrates that dual targeting of RXR and HDAC using DW22 possesses pleiotropic antitumor activities both in vitro and in vivo, providing a novel therapeutic approach for cancer treatment.
Collapse
Affiliation(s)
- Lihui Wang
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, P.R. China.,Benxi Institute of Pharmaceutical Research, Shenyang Pharmaceutical University, Benxi, P.R. China
| | - Guoliang Chen
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang, P.R. China
| | - Kang Chen
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, P.R. China.,Benxi Institute of Pharmaceutical Research, Shenyang Pharmaceutical University, Benxi, P.R. China
| | - Yong Ren
- Department of Pathology, Wuhan General Hospital of Guangzhou Command, People's Liberation Army, Wuhan, P.R. China
| | - Huahuan Li
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, P.R. China.,Benxi Institute of Pharmaceutical Research, Shenyang Pharmaceutical University, Benxi, P.R. China
| | - Xiaorui Jiang
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, P.R. China.,Benxi Institute of Pharmaceutical Research, Shenyang Pharmaceutical University, Benxi, P.R. China
| | - Lina Jia
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, P.R. China
| | - Shiyuan Fu
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, P.R. China.,Benxi Institute of Pharmaceutical Research, Shenyang Pharmaceutical University, Benxi, P.R. China
| | - Yi Li
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, P.R. China.,Benxi Institute of Pharmaceutical Research, Shenyang Pharmaceutical University, Benxi, P.R. China
| | - Xinwei Liu
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, P.R. China.,Benxi Institute of Pharmaceutical Research, Shenyang Pharmaceutical University, Benxi, P.R. China
| | - Shuang Wang
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, P.R. China.,Benxi Institute of Pharmaceutical Research, Shenyang Pharmaceutical University, Benxi, P.R. China
| | - Jingyu Yang
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, P.R. China.,Benxi Institute of Pharmaceutical Research, Shenyang Pharmaceutical University, Benxi, P.R. China
| | - Chunfu Wu
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, P.R. China.,Benxi Institute of Pharmaceutical Research, Shenyang Pharmaceutical University, Benxi, P.R. China
| |
Collapse
|
158
|
Achyut BR, Arbab AS. Myeloid cell signatures in tumor microenvironment predicts therapeutic response in cancer. Onco Targets Ther 2016; 9:1047-55. [PMID: 27042097 PMCID: PMC4780185 DOI: 10.2147/ott.s102907] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Tumor microenvironment (TME) consists of several immune and nonimmune cell populations including tumor cells. For many decades, experimental studies have depicted profound contribution of TME toward cancer progression and metastasis development. Several therapeutic strategies have been tested against TME through preclinical studies and clinical trials. Unfortunately, most of them have shown transient effect, and have largely failed due to aggressive tumor growth and without improving survival. Solid tumors are known to have a strong myeloid component (eg, tumor-associated macrophages) in tumor development. Recent data suggest that therapeutic responses in tumor are characterized by alterations in immune cell signatures, including tumor-associated myeloid cells. Polarized tumor-associated myeloid cells (M1–M2) are critical in impairing therapeutic effect and promoting tumor growth. The present review is intended to compile all the literatures related to the emerging contribution of different populations of myeloid cells in the development of tumor and therapeutic failures. Finally, we have discussed targeting of myeloid cell populations as a combination therapy with chemo-, targeted-, or radiation therapies.
Collapse
Affiliation(s)
- Bhagelu R Achyut
- Tumor Angiogenesis Laboratory, Department of Biochemistry and Molecular Biology, Cancer Center, Georgia Regents University, Augusta, GA, USA
| | - Ali S Arbab
- Tumor Angiogenesis Laboratory, Department of Biochemistry and Molecular Biology, Cancer Center, Georgia Regents University, Augusta, GA, USA
| |
Collapse
|
159
|
Mao Y, Wu S, Zhao R, Deng Q. MiR-205 promotes proliferation, migration and invasion of nasopharyngeal carcinoma cells by activation of AKT signalling. J Int Med Res 2016; 44:231-40. [PMID: 26880795 PMCID: PMC5580060 DOI: 10.1177/0300060515576556] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Accepted: 02/16/2015] [Indexed: 01/11/2023] Open
Abstract
Objective To examine the role of microRNA (miR)-205 in proliferation, migration and invasion of nasopharyngeal carcinoma (NPC). Methods The human NPC cell line CNE2 was transfected with miR-205 mimic, anti-miR-205 inhibitor or scrambled oligonucleotide (control). Cell proliferation was assessed via MTT assay. Cell migration and invasion were evaluated by transwell migration and Matrigel® invasion assay, respectively. Radiation induced apoptosis was quantified via Caspase-Glo3/7 assay. Apoptotic proteins and epithelial–mesenchymal transition (EMT) proteins were semiquantified by Western blot analysis. Results Overexpression of miR-205 increased the proliferation, migration and invasion of CNE2 cells, and decreased radiation-induced apoptosis compared with control cells. MiR-205 overexpression downregulated E-cadherin and upregulated Snail expression via downregulation of PTEN and upregulation of AKT. Conclusion MiR-205 plays vital roles in tumourigenesis and tumour progression in NPC, and may be a potential treatment target.
Collapse
Affiliation(s)
- Yanjiao Mao
- Department of Radiation Oncology, Hangzhou Cancer Hospital, Hangzhou First People's Hospital, Hangzhou, China
| | - Shixiu Wu
- Department of Radiation Oncology, Hangzhou Cancer Hospital, Hangzhou First People's Hospital, Hangzhou, China
| | - Ruping Zhao
- Department of Radiation Oncology, Hangzhou Cancer Hospital, Hangzhou First People's Hospital, Hangzhou, China
| | - Qinghua Deng
- Department of Radiation Oncology, Hangzhou Cancer Hospital, Hangzhou First People's Hospital, Hangzhou, China
| |
Collapse
|
160
|
Kim P, Cheng F, Zhao J, Zhao Z. ccmGDB: a database for cancer cell metabolism genes. Nucleic Acids Res 2015; 44:D959-68. [PMID: 26519468 PMCID: PMC4702820 DOI: 10.1093/nar/gkv1128] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Accepted: 10/15/2015] [Indexed: 12/24/2022] Open
Abstract
Accumulating evidence has demonstrated that rewiring of metabolism in cells is an important hallmark of cancer. The percentage of patients killed by metabolic disorder has been estimated to be 30% of the advanced-stage cancer patients. Thus, a systematic annotation of cancer cell metabolism genes is imperative. Here, we present ccmGDB (Cancer Cell Metabolism Gene DataBase), a comprehensive annotation database for cell metabolism genes in cancer, available at http://bioinfo.mc.vanderbilt.edu/ccmGDB. We assembled, curated, and integrated genetic, genomic, transcriptomic, proteomic, biological network and functional information for over 2000 cell metabolism genes in more than 30 cancer types. In total, we integrated over 260 000 somatic alterations including non-synonymous mutations, copy number variants and structural variants. We also integrated RNA-Seq data in various primary tumors, gene expression microarray data in over 1000 cancer cell lines and protein expression data. Furthermore, we constructed cancer or tissue type-specific, gene co-expression based protein interaction networks and drug-target interaction networks. Using these systematic annotations, the ccmGDB portal site provides 6 categories: gene summary, phenotypic information, somatic mutations, gene and protein expression, gene co-expression network and drug pharmacological information with a user-friendly interface for browsing and searching. ccmGDB is developed and maintained as a useful resource for the cancer research community.
Collapse
Affiliation(s)
- Pora Kim
- Department of Biomedical Informatics, Vanderbilt University School of Medicine, Nashville, TN 37203, USA
| | - Feixiong Cheng
- Department of Biomedical Informatics, Vanderbilt University School of Medicine, Nashville, TN 37203, USA
| | - Junfei Zhao
- Department of Biomedical Informatics, Vanderbilt University School of Medicine, Nashville, TN 37203, USA
| | - Zhongming Zhao
- Department of Biomedical Informatics, Vanderbilt University School of Medicine, Nashville, TN 37203, USA Department of Cancer Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA Department of Psychiatry, Vanderbilt University School of Medicine, Nashville, TN 37212, USA
| |
Collapse
|
161
|
Gao C, Zhang W, He S, Li S, Liu F, Jiang Y. Synthesis and antiproliferative activity of 2,7-diamino l0-(3,5-dimethoxy)benzyl-9(10H)-acridone derivatives as potent telomeric G-quadruplex DNA ligands. Bioorg Chem 2015; 60:30-6. [DOI: 10.1016/j.bioorg.2015.04.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2014] [Revised: 04/01/2015] [Accepted: 04/02/2015] [Indexed: 12/30/2022]
|
162
|
Zhang L, Li Z, Gai F, Wang Y. MicroRNA-137 suppresses tumor growth in epithelial ovarian cancer in vitro and in vivo. Mol Med Rep 2015; 12:3107-14. [PMID: 25955305 DOI: 10.3892/mmr.2015.3756] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Accepted: 04/08/2015] [Indexed: 11/05/2022] Open
Abstract
Epithelial ovarian cancer (EOC) remains a major gynecological problem, with a poor 5-year-survival rate due to distant metastases. The identification of microRNAs (miRNAs) may provide a novel avenue for diagnostic and treatment regimens for EOC. Several miRNAs have been reported to be involved in the progression of EOC, among which miRNA (miR)-137 has been observed to be downregulated in the ovarian tissues of patients with EOC. However, the functions of miR-137 in EOC cell apoptosis, migration and invasion remain to be elucidated. In the present study, the expression of miR-137 was measured in clinical ovarian cancer specimens and cell lines using reverse transcription-quantitative polymerase chain reaction. The role of miR-137 in the growth and survival of the SKOV3 human ovarian cancer cell line was determined using several in vitro approaches and in nude mouse models. The results demonstrated that the expression of miR-137 was downregulated in the ovarian cancer specimens and cell lines. It was also observed that enforced expression of miR-137 in the EOC cell lines decreased cell proliferation, clonogenicity, migration and invasion, and induced G1 arrest and cell apoptosis in vitro. Notably, the enforced expression of miR-137 suppressed tumor growth in the nude mice models. These findings suggested that miR-137 may act as a tumor suppressor and be used as a potential therapeutic agent for the treatment of EOC.
Collapse
Affiliation(s)
- Liyu Zhang
- Department of Obstetrics and Gynecology, The Affiliated Hospital, Changchun University of Chinese Medicine, Changchun, Jilin 130117, P.R. China
| | - Zhihong Li
- Department of Obstetrics and Gynecology, The Affiliated Hospital, Changchun University of Chinese Medicine, Changchun, Jilin 130117, P.R. China
| | - Fengchun Gai
- Department of Infectious Diseases, The Affiliated Hospital, Changchun University of Chinese Medicine, Changchun, Jilin 130117, P.R. China
| | - Yanping Wang
- Department of Obstetrics and Gynecology, The Affiliated Hospital, Changchun University of Chinese Medicine, Changchun, Jilin 130117, P.R. China
| |
Collapse
|
163
|
Kale VP, Amin SG, Pandey MK. Targeting ion channels for cancer therapy by repurposing the approved drugs. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2015; 1848:2747-55. [PMID: 25843679 DOI: 10.1016/j.bbamem.2015.03.034] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Revised: 03/18/2015] [Accepted: 03/27/2015] [Indexed: 12/21/2022]
Abstract
Ion channels have been shown to be involved in oncogenesis and efforts are being poured in to target the ion channels. There are many clinically approved drugs with ion channels as "off" targets. The question is, can these drugs be repurposed to inhibit ion channels for cancer treatment? Repurposing of drugs will not only save investors' money but also result in safer drugs for cancer patients. Advanced bioinformatics techniques and availability of a plethora of open access data on FDA approved drugs for various indications and omics data of large number of cancer types give a ray of hope to look for possibility of repurposing those drugs for cancer treatment. This article is part of a Special Issue entitled: Membrane channels and transporters in cancers.
Collapse
Affiliation(s)
- Vijay Pralhad Kale
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Shantu G Amin
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Manoj K Pandey
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA.
| |
Collapse
|
164
|
Yang JJ, Hu ZG, Shi WX, Deng T, He SQ, Yuan SG. Prognostic significance of neutrophil to lymphocyte ratio in pancreatic cancer: A meta-analysis. World J Gastroenterol 2015; 21:2807-2815. [PMID: 25759553 PMCID: PMC4351235 DOI: 10.3748/wjg.v21.i9.2807] [Citation(s) in RCA: 159] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Revised: 08/08/2014] [Accepted: 10/15/2014] [Indexed: 02/06/2023] Open
Abstract
AIM: To conduct a meta-analysis evaluating the association between the peripheral blood neutrophil to lymphocyte ratio (NLR) and the outcome of patients with pancreatic cancer.
METHODS: Studies evaluating the relationship between the peripheral blood NLR and outcome of patients with pancreatic cancer published up to May 2014 were searched using electronic databases, including PubMed, Web of Science, Embase and Ovid. A meta-analysis was performed to pool the hazard ratios (HRs) or odds ratios (ORs) and their 95% confidence intervals (CIs) using either a fixed-effects model or a random-effects model to quantitatively assess the prognostic value of NLR and its association with clinicopathological parameters.
RESULTS: Eleven studies containing a total of 1804 patients were eligible according to our selection criteria, and combined hazard ratios indicated that high NLR was a poor prognostic marker for pancreatic cancer patients because it had an unfavorable impact on the overall survival (OS) (HR = 2.61, 95%CI: 1.68-4.06, P = 0.000) and cancer specific survival (HR = 1.66, 95%CI: 1.08-2.57, P = 0.021). Subgroup analysis revealed that high NLR was associated with poor OS in patients with mixed treatment (HR = 4.36, 95%CI: 2.50-7.61, P = 0.000), chemotherapy (HR = 2.08, 95%CI: 1.49-2.9, P = 0.000), or surgical resection (HR = 1.2, 95%CI: 1.00-1.44, P = 0.048). Additionally, high NLR was significantly correlated with tumor metastasis (OR = 1.69, 95%CI: 1.10-2.59, P = 0.016), poor tumor differentiation (OR = 2.75, 95%CI: 1.19-6.36, P = 0.016), poor performance status (OR = 2.56, 95%CI: 1.63-4.03, P = 0.000), high cancer antigen 199 (OR = 2.62, 95%CI: 1.49-4.60, P = 0.000), high C-reactive protein (OR = 4.32, 95%CI: 2.71-6.87, P = 0.000), and low albumin (OR = 3.56, 95%CI: 1.37-9.27, P = 0.009).
CONCLUSION: High peripheral blood NLR suggested a poor prognosis for patients with pancreatic cancer, and it could be a novel marker of survival evaluation and could help clinicians develop therapeutic strategies for pancreatic cancer patients.
Collapse
|
165
|
Comprehensive network analysis of genes expressed in human oropharyngeal cancer. Am J Otolaryngol 2015; 36:235-41. [PMID: 25484365 DOI: 10.1016/j.amjoto.2014.11.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Accepted: 11/13/2014] [Indexed: 01/02/2023]
Abstract
PURPOSE Oropharyngeal cancer (OPC) is the eighth most common cancer worldwide, however the genes involved in the development of OPC have been reported few. We constructed a co-expression network to extend knowledge of the molecular biomarkers in OPC development. MATERIALS AND METHODS Microarray data of HPV-active, -inactive, -negative OPC and normal benign tissue (uvula, tonsil) (Series GSE55550) were retrieved from NCBI GEO DataSets. We performed co-expression analysis of OPC transcriptome data by the Pearson correlation coefficient (PCC) method with the mutual rank (MR)-based cut-off using 13 guide genes. RESULTS The OPC subnetwork contained three clusters: cell cycle (62 node genes and 125 edge genes), immune system (44 node genes and 70 edge genes) and organ morphogenesis (128 node gene and 215 edge genes) process separately. CONCLUSION Our co-expression analysis includes separated transcriptomes of OPC, which is a useful resource for OPC researchers to elucidate important and complex biological events, to prevent and to predict cancer.
Collapse
|
166
|
Cho H, Lai TC, Tomoda K, Kwon GS. Polymeric micelles for multi-drug delivery in cancer. AAPS PharmSciTech 2015; 16:10-20. [PMID: 25501872 DOI: 10.1208/s12249-014-0251-3] [Citation(s) in RCA: 97] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Accepted: 11/25/2014] [Indexed: 01/16/2023] Open
Abstract
Drug combinations are common in cancer treatment and are rapidly evolving, moving beyond chemotherapy combinations to combinations of signal transduction inhibitors. For the delivery of drug combinations, i.e., multi-drug delivery, major considerations are synergy, dose regimen (concurrent versus sequential), pharmacokinetics, toxicity, and safety. In this contribution, we review recent research on polymeric micelles for multi-drug delivery in cancer. In concurrent drug delivery, polymeric micelles deliver multi-poorly water-soluble anticancer agents, satisfying strict requirements in solubility, stability, and safety. In sequential drug delivery, polymeric micelles participate in pretreatment strategies that "prime" solid tumors and enhance the penetration of secondarily administered anticancer agent or nanocarrier. The improved delivery of multiple poorly water-soluble anticancer agents by polymeric micelles via concurrent or sequential regimens offers novel and interesting strategies for drug combinations in cancer treatment.
Collapse
|
167
|
Li Z, Park HR, Shi Z, Li Z, Pham CD, Du Y, Khuri FR, Zhang Y, Han Q, Fu H. Pro-oncogenic function of HIP-55/Drebrin-like (DBNL) through Ser269/Thr291-phospho-sensor motifs. Oncotarget 2015; 5:3197-209. [PMID: 24912570 PMCID: PMC4102803 DOI: 10.18632/oncotarget.1900] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
HIP-55 (HPK1-interacting protein of 55 kDa, also named DBNL, SH3P7, and mAbp1) is a multidomain adaptor protein that is critical for organ development and the immune response. Here, we report the coupling of HIP-55 to cell growth control through its 14-3-3-binding phospho-Ser/Thr-sensor sites. Using affinity chromatography, we found HIP-55 formed a complex with 14-3-3 proteins, revealing a new node in phospho-Ser/Thr-mediated signaling networks. In addition, we demonstrated that HIP-55 is required for proper cell growth control. Enforced HIP-55 expression promoted proliferation, colony formation, migration, and invasion of lung cancer cells while silencing of HIP-55 reversed these effects. Importantly, HIP-55 was found to be upregulated in lung cancer cell lines and in tumor tissues of lung cancer patients. Upregulated HIP-55 was required to promote the growth of tumors in a xenograft animal model. However, tumors with S269A/T291A-mutated HIP-55, which ablates 14-3-3 binding, exhibited significantly reduced sizes, supporting a vital role of the HIP-55/14-3-3 protein interaction node in transmitting oncogenic signals. Mechanistically, HIP-55-mediated tumorigenesis activity appears to be in part mediated by antagonizing the tumor suppressor function of HPK1. Thus, the HIP-55–mediated oncogenic pathway, through S269/T291, may be exploited for the development of new therapeutic strategies.
Collapse
Affiliation(s)
- Zijian Li
- Department of Pharmacology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
168
|
Song D, Tian Y, Huang S, Li B, Yuan Y, Hu A. An acyclic enediyne with a furyl tethering group for efficient inhibition of tumor cell viability. J Mater Chem B 2015; 3:8584-8588. [DOI: 10.1039/c5tb01867f] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
An acyclic enediyne with a furyl tethering group and two pH-sensitive orthoester groups at the alkynyl termini was synthesized. The introduction of a furyl tethering group represents a new design strategy of “intelligent” antitumor antibiotics that can distinguish tumor and normal cells.
Collapse
Affiliation(s)
- Depeng Song
- Shanghai Key Laboratory of Advanced Polymeric Materials
- School of Materials Science and Engineering
- East China University of Science and Technology
- Shanghai
- China
| | - Yu Tian
- The State Key Laboratory of Bioreactor Engineering
- East China University of Science and Technology
- Shanghai
- China
| | - Shuai Huang
- Shanghai Key Laboratory of Advanced Polymeric Materials
- School of Materials Science and Engineering
- East China University of Science and Technology
- Shanghai
- China
| | - Baojun Li
- Shanghai Key Laboratory of Advanced Polymeric Materials
- School of Materials Science and Engineering
- East China University of Science and Technology
- Shanghai
- China
| | - Yuan Yuan
- The State Key Laboratory of Bioreactor Engineering
- East China University of Science and Technology
- Shanghai
- China
| | - Aiguo Hu
- Shanghai Key Laboratory of Advanced Polymeric Materials
- School of Materials Science and Engineering
- East China University of Science and Technology
- Shanghai
- China
| |
Collapse
|
169
|
Pramanik S, Kutzner A, Heese K. Lead discovery and in silico 3D structure modeling of tumorigenic FAM72A (p17). Tumour Biol 2014; 36:239-49. [PMID: 25234718 DOI: 10.1007/s13277-014-2620-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2014] [Accepted: 09/09/2014] [Indexed: 12/30/2022] Open
Abstract
FAM72A (p17) is a novel neuronal protein that has been linked to tumorigenic effects in non-neuronal tissue. Using state of the art in silico physicochemical analyses (e.g., I-TASSER, RaptorX, and Modeller), we determined the three-dimensional (3D) protein structure of FAM72A and further identified potential ligand-protein interactions. Our data indicate a Zn(2+)/Fe(3+)-containing 3D protein structure, based on a 3GA3_A model template, which potentially interacts with the organic molecule RSM ((2s)-2-(acetylamino)-N-methyl-4-[(R)-methylsulfinyl] butanamide). The discovery of RSM may serve as potential lead for further anti-FAM72A drug screening tests in the pharmaceutical industry because interference with FAM72A's activities via RSM-related molecules might be a novel option to influence the tumor suppressor protein p53 signaling pathways for the treatment of various types of cancers.
Collapse
Affiliation(s)
- Subrata Pramanik
- Graduate School of Biomedical Science and Engineering, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul, 133-791, Republic of Korea
| | | | | |
Collapse
|
170
|
Rana A, Alex JM, Chauhan M, Joshi G, Kumar R. A review on pharmacophoric designs of antiproliferative agents. Med Chem Res 2014. [DOI: 10.1007/s00044-014-1196-5] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
171
|
Guo S, Lin CM, Xu Z, Miao L, Wang Y, Huang L. Co-delivery of cisplatin and rapamycin for enhanced anticancer therapy through synergistic effects and microenvironment modulation. ACS NANO 2014; 8:4996-5009. [PMID: 24720540 PMCID: PMC4046782 DOI: 10.1021/nn5010815] [Citation(s) in RCA: 144] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/07/2023]
Abstract
The tumor microenvironment plays an important role in the tumor's progression and metastasis. Therefore, successful alteration of this delicate setting against the tumor's favor can open a window for therapeutic efficacy. We have developed a modality to bring about treatment-induced alterations in the tumor microenvironment by employing the synergistic effects between two drugs. Co-delivery of rapamycin (RAPA), an mTOR inhibitor that may offer notable therapy through antiangiogenic activity, alongside cisplatin can foster significant potency as RAPA sensitizes A375 melanoma cells to cisplatin therapy through microenvironment modulation. However, encapsulation of these drugs into poly(lactic-co-glycolic acid) (PLGA) NPs was inefficient due to the incompatibility between the two free drugs and the polymer matrix. Here, we show cisplatin can be made hydrophobic by coating a nanoprecipitate (cores) of the drug with dioleoylphosphatidic acid (DOPA). These DOPA coated cisplatin cores are compatible with PLGA and can be coencapsulated in PLGA NPs alongside RAPA at a molar ratio to promote synergistic antitumor activity. The presence of the cisplatin cores significantly improved the encapsulation of RAPA into PLGA NPs. Furthermore, PLGA NPs containing both cisplatin cores and RAPA induced significant apoptosis on A375-luc human melanoma cells in vitro. Additionally, they inhibited the growth of A375-luc melanoma in a xenograft tumor model through modulation of the tumor vasculature and permitted enhanced penetration of NPs into the tumor.
Collapse
|
172
|
Synergistic effects of glycated chitosan with high-intensity focused ultrasound on suppression of metastases in a syngeneic breast tumor model. Cell Death Dis 2014; 5:e1178. [PMID: 24743733 PMCID: PMC4001313 DOI: 10.1038/cddis.2014.159] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2014] [Revised: 03/13/2014] [Accepted: 03/13/2014] [Indexed: 01/12/2023]
Abstract
Stimulation of the host immune system is crucial in cancer treatment. In particular, nonspecific immunotherapies, when combined with other traditional therapies such as radiation and chemotherapy, may induce immunity against primary and metastatic tumors. In this study, we demonstrate that a novel, non-toxic immunoadjuvant, glycated chitosan (GC), decreases the motility and invasion of mammalian breast cancer cells in vitro and in vivo. Lung metastatic ratios were reduced in 4T1 tumor-bearing mice when intratumoral GC injection was combined with local high-intensity focused ultrasound (HIFU) treatment. We postulate that this treatment modality stimulates the host immune system to combat cancer cells, as macrophage accumulation in tumor lesions was detected after GC-HIFU treatment. In addition, plasma collected from GC-HIFU-treated tumor-bearing mice exhibited tumor-specific cytotoxicity. We also investigated the effect of GC on epithelial–mesenchymal transition-related markers. Our results showed that GC decreased the expression of Twist-1 and Slug, proto-oncogenes commonly implicated in metastasis. Epithelial-cadherin, which is regulated by these genes, was also upregulated. Taken together, our current data suggest that GC alone can reduce cancer cell motility and invasion, whereas GC-HIFU treatment can induce immune responses to suppress tumor metastasis in vivo.
Collapse
|
173
|
Xu C, Lin L, Cao G, Chen Q, Shou P, Huang Y, Han Y, Wang Y, Shi Y. Interferon-α-secreting mesenchymal stem cells exert potent antitumor effect in vivo. Oncogene 2013; 33:5047-52. [PMID: 24186200 DOI: 10.1038/onc.2013.458] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2013] [Revised: 07/24/2013] [Accepted: 08/13/2013] [Indexed: 12/13/2022]
Abstract
Mesenchymal stem cells (MSCs) are a kind of adult stem cells that can be isolated easily from bone marrow, adipose tissue, umbilical cord and many other tissues. MSCs have been shown to specifically migrate to inflammatory sites, including tumors, and hold great promise as tumor-specific vectors to deliver antitumor agents. Interferon-α (IFNα) has been used in clinic to treat various types of tumors; however, because of its short half-life, significant therapeutic effects require high doses that often results in serious side effects. Here, we tested whether MSCs continuingly secreting IFNα can exert a persistent antitumor effect and eliminate the side effects associated with high clinical doses of recombinant IFNα. We found that even a small number of IFNα-secreting MSCs could potently halt B16 tumor growth in vivo. The antitumor activity of IFNα-secreting MSCs was largely abolished in immunodeficient mice, an effect largely attributed to natural killer cells and CD8(+) T cells. Therefore, IFNα-secreting MSCs provide an innovative strategy for tumor therapy.
Collapse
Affiliation(s)
- C Xu
- Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences/Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - L Lin
- Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences/Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - G Cao
- Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences/Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Q Chen
- Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences/Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - P Shou
- Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences/Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Y Huang
- Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences/Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Y Han
- Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences/Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Y Wang
- Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences/Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Y Shi
- Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences/Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
174
|
Li GH, Huang JF. Inferring therapeutic targets from heterogeneous data: HKDC1 is a novel potential therapeutic target for cancer. Bioinformatics 2013; 30:748-52. [DOI: 10.1093/bioinformatics/btt606] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
|
175
|
miRNA and mRNA cancer signatures determined by analysis of expression levels in large cohorts of patients. Proc Natl Acad Sci U S A 2013; 110:19160-5. [PMID: 24101511 DOI: 10.1073/pnas.1316991110] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Toward identifying a cancer-specific gene signature we applied surprisal analysis to the RNAs expression behavior for a large cohort of breast, lung, ovarian, and prostate carcinoma patients. We characterize the cancer phenotypic state as a shared response of a set of mRNA or microRNAs (miRNAs) in cancer patients versus noncancer controls. The resulting signature is robust with respect to individual patient variability and distinguishes with high fidelity between cancer and noncancer patients. The mRNAs and miRNAs that are implicated in the signature are correlated and are known to contribute to the regulation of cancer-signaling pathways. The miRNA and mRNA networks are common to the noncancer and cancer patients, but the disease modulates the strength of the connectivities. Furthermore, we experimentally assessed the cancer-specific signatures as possible therapeutic targets. Specifically we restructured a single dominant connectivity in the cancer-specific gene network in vitro. We find a deflection from the cancer phenotype, significantly reducing cancer cell proliferation and altering cancer cellular physiology. Our approach is grounded in thermodynamics augmented by information theory. The thermodynamic reasoning is demonstrated to ensure that the derived signature is bias-free and shows that the most significant redistribution of free energy occurs in programming a system between the noncancer and cancer states. This paper introduces a platform that can elucidate miRNA and mRNA behavior on a systems level and provides a comprehensive systematic view of both the energetics of the expression levels of RNAs and of their changes during tumorigenicity.
Collapse
|
176
|
Zheng Y, Liu Y, Jin H, Pan S, Qian Y, Huang C, Zeng Y, Luo Q, Zeng M, Zhang Z. Scavenger receptor B1 is a potential biomarker of human nasopharyngeal carcinoma and its growth is inhibited by HDL-mimetic nanoparticles. Am J Cancer Res 2013; 3:477-86. [PMID: 23843895 PMCID: PMC3706691 DOI: 10.7150/thno.6617] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2013] [Accepted: 06/04/2013] [Indexed: 12/11/2022] Open
Abstract
Nasopharyngeal carcinoma (NPC) is a very regional malignant head and neck cancer that has attracted widespread attention for its unique etiology, epidemiology and therapeutic options. To achieve high cure rates in NPC patients, theranostic approaches are actively being pursued and improved efforts remain desirable in identifying novel biomarkers and establishing effective therapeutic approaches with low long-term toxicities. Here, we discovered that the scavenger receptor class B type I (SR-B1) was overexpressed in all investigated NPC cell lines and 75% of NPC biopsies, demonstrating that SR-B1 is a potential biomarker of NPC. Additional functional analysis showed that SR-B1 has great effect on cell motility while showing no significant impact on cell proliferation. As high-density lipoproteins (HDL) exhibit strong binding affinities to SR-B1 and HDL mimetic peptides are reportedly capable of inhibiting tumor growth, we further examined the SR-B1 targeting ability of a highly biocompatible HDL-mimicking peptide-phospholipid scaffold (HPPS) nanocarrier and investigated its therapeutic effect on NPC. Results show that NPC cells with higher SR-B1 expression have superior ability in taking up the core constituents of HPPS. Moreover, HPPS inhibited the motility and colony formation of 5-8F cells, and significantly suppressed the NPC cell growth in nude mice without inducing tumor cell necrosis or apoptosis. These results indicate that HPPS is not only a NPC-targeting nanocarrier but also an effective anti-NPC drug. Together, the identification of SR-B1 as a potential biomarker and the use of HPPS as an effective anti-NPC agent may shed new light on the diagnosis and therapeutics of NPC.
Collapse
|
177
|
Roumeliotis TI, Halabalaki M, Alexi X, Ankrett D, Giannopoulou EG, Skaltsounis AL, Sayan BS, Alexis MN, Townsend PA, Garbis SD. Pharmacoproteomic study of the natural product Ebenfuran III in DU-145 prostate cancer cells: the quantitative and temporal interrogation of chemically induced cell death at the protein level. J Proteome Res 2013; 12:1591-603. [PMID: 23418717 DOI: 10.1021/pr300968q] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
A naturally occurring benzofuran derivative, Ebenfuran III (Eb III), was investigated for its antiproliferative effects using the DU-145 prostate cell line. Eb III was isolated from Onobrychis ebenoides of the Leguminosae family, a plant endemic in Central and Southern Greece. We have previously reported that Eb III exerts significant cytotoxic effects on certain cancer cell lines. This effect is thought to occur via the isoprenyl moiety at the C-5 position of the molecule. The study aim was to gain a deeper understanding of the pharmacological effect of Eb III on DU-145 cell death at the translational level using a relative quantitative and temporal proteomics approach. Proteins extracted from the cell pellets were subjected to solution phase trypsin proteolysis followed by iTRAQ-labeling. The labeled tryptic peptide extracts were then fractionated using strong cation exchange chromatography and the fractions were analyzed by nanoflow reverse phase ultraperformance liquid chromatography-nanoelectrospray ionization-tandem mass spectrometry analysis using a hybrid QqTOF platform. Using this approach, we compared the expression levels of 1360 proteins analyzed at ≤ 1% global protein false discovery rate (FDR), commonly present in untreated (control, vehicle only) and Eb III-treated cells at the different exposure time points. Through the iterative use of Ingenuity Pathway Analysis with hierarchical clustering of protein expression patterns, followed by bibliographic research, the temporal regulation of the Calpain-1, ERK2, PAR-4, RAB-7, and Bap31 proteins were identified as potential nodes of multipathway convergence to Eb III induced DU-145 cell death. These proteins were further verified with Western blot analysis. This gel-free, quantitative 2DLC-MS/MS proteomics method effectively captured novel modulated proteins in the DU-145 cell line as a response to Eb III treatment. This approach also provided greater insight to the multifocal and combinatorial signaling pathways implicated in Eb III-induced cell death.
Collapse
Affiliation(s)
- Theodoros I Roumeliotis
- Institute for Life Sciences, ‡Cancer Sciences Unit, Cancer Research U.K., Faculty of Medicine, University of Southampton , Southampton, United Kingdom
| | | | | | | | | | | | | | | | | | | |
Collapse
|