Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
Add more filters










Publication year range
1.
Signal Transduct Target Ther ; 7(1): 296, 2022 08 23.
Article in English | MEDLINE | ID: mdl-35999218

ABSTRACT

Metastasis is a pivotal event that accelerates the prognosis of cancer patients towards mortality. Therapies that aim to induce cell death in metastatic cells require a more detailed understanding of the metastasis for better mitigation. Towards this goal, we discuss the details of two distinct but overlapping pathways of metastasis: a classical reversible epithelial-to-mesenchymal transition (hybrid-EMT)-driven transport pathway and an alternative cell death process-driven blebbishield metastatic-witch (BMW) transport pathway involving reversible cell death process. The knowledge about the EMT and BMW pathways is important for the therapy of metastatic cancers as these pathways confer drug resistance coupled to immune evasion/suppression. We initially discuss the EMT pathway and compare it with the BMW pathway in the contexts of coordinated oncogenic, metabolic, immunologic, and cell biological events that drive metastasis. In particular, we discuss how the cell death environment involving apoptosis, ferroptosis, necroptosis, and NETosis in BMW or EMT pathways recruits immune cells, fuses with it, migrates, permeabilizes vasculature, and settles at distant sites to establish metastasis. Finally, we discuss the therapeutic targets that are common to both EMT and BMW pathways.


Subject(s)
Epithelial-Mesenchymal Transition , Neoplasms , Carcinogenesis , Cell Death , Epithelial-Mesenchymal Transition/genetics , Humans , Neoplasms/metabolism , Oncogenes
2.
Sci Rep ; 11(1): 12673, 2021 06 16.
Article in English | MEDLINE | ID: mdl-34135394

ABSTRACT

A subset of hepatocellular carcinoma (HCC) overexpresses the chromosome 19 miRNA cluster (C19MC) and is associated with an undifferentiated phenotype marked by overexpression of cancer testis antigens (CTAs) including anti-apoptotic melanoma-A antigens (MAGEAs). However, the regulation of C19MC miRNA and MAGEA expression in HCCs are not understood. Here we show that, C19MC overexpression is tightly linked to a sub-set of HCCs with transcription-incompetent p53. Using next-generation and Sanger sequencing we found that, p53 in Hep3B cells is impaired by TP53-FXR2 fusion, and that overexpression of the C19MC miRNA-520G in Hep3B cells promotes the expression of MAGEA-3, 6 and 12 mRNAs. Furthermore, overexpression of p53-R175H and p53-R273H mutants promote miR-520G and MAGEA RNA expression and cellular transformation. Moreover, IFN-γ co-operates with miR-520G to promote MAGEA expression. On the other hand, metals such as nickel and zinc promote miR-526B but not miR-520G, to result in the suppression of MAGEA mRNA expression, and evoke cell death through mitochondrial membrane depolarization. Therefore our study demonstrates that a MAGEA-promoting network involving miR-520G, p53-defects and IFN-γ that govern cellular transformation and cell survival pathways, but MAGEA expression and survival are counteracted by nickel and zinc combination.


Subject(s)
Antigens, Neoplasm , Carcinoma, Hepatocellular , Cell Transformation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic , Antigens, Neoplasm/genetics , Antigens, Neoplasm/metabolism , Carcinoma, Hepatocellular/etiology , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Chromosomes, Human, Pair 19 , Humans , Interferon-gamma/metabolism , Liver Neoplasms/etiology , Liver Neoplasms/pathology , Male , MicroRNAs/metabolism , Mutation , Oncogene Fusion , Peptide Fragments/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Testis/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
3.
Cell Death Discov ; 7(1): 80, 2021 Apr 14.
Article in English | MEDLINE | ID: mdl-33854056

ABSTRACT

Cellular transformation is a major event that helps cells to evade apoptosis, genomic instability checkpoints, and immune surveillance to initiate tumorigenesis and to promote progression by cancer stem cell expansion. However, the key molecular players that govern cellular transformation and ways to target cellular transformation for therapy are poorly understood to date. Here we draw key evidences from the literature on K-Ras-driven cellular transformation in the context of apoptosis to shed light on the key players that are required for cellular transformation and explain how aiming p53 could be useful to target cellular transformation. The defects in key apoptosis regulators such as p53, Bax, and Bak lead to apoptosis evasion, cellular transformation, and genomic instability to further lead to stemness, tumorigenesis, and metastasis via c-Myc-dependent transcription. Therefore enabling key apoptotic checkpoints in combination with K-Ras inhibitors will be a promising therapeutic target in cancer therapy.

4.
Sci Rep ; 10(1): 12371, 2020 07 23.
Article in English | MEDLINE | ID: mdl-32704163

ABSTRACT

MYO18B has been proposed to contribute to the progression of hepatocellular carcinoma (HCC). However, the signals that govern MYO18B transcription are not known. Here we show that, a network of C19MC miRNA-520G, IFN-γ, CEBPB and p53 transcriptional-defects promote MYO18B mRNA expression in HCCs. IFN-γ by itself suppresses MYO18B transcription, but promotes it when miRNA-520G is stably overexpressed. Similarly, CEBPB-liver-enriched activator protein (LAP) isoform overexpression suppresses MYO18B transcription but promotes transcription when the cells are treated with IFN-γ. Furthermore, miR-520G together with mutant-p53 promotes MYO18B transcription. Conversely, bFGF suppresses MYO18B mRNA irrespective of CEBPB, miR-520G overexpression or IFN-γ treatment. Finally high MYO18B expression reflects poor prognosis while high MYL5 or MYO1B expression reflects better survival of HCC patients. Thus, we identified a network of positive and negative regulators of MYO18B mRNA expression which reflects the survival of HCC patients.


Subject(s)
CCAAT-Enhancer-Binding Protein-beta/biosynthesis , Carcinoma, Hepatocellular/metabolism , Fibroblast Growth Factor 2/biosynthesis , Gene Expression Regulation, Neoplastic , Interferon-gamma/biosynthesis , Liver Neoplasms/metabolism , MicroRNAs/biosynthesis , Myosins/biosynthesis , RNA, Messenger/biosynthesis , RNA, Neoplasm/biosynthesis , Tumor Suppressor Protein p53/biosynthesis , Tumor Suppressor Proteins/biosynthesis , CCAAT-Enhancer-Binding Protein-beta/genetics , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Line , Female , Fibroblast Growth Factor 2/genetics , Humans , Interferon-gamma/genetics , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Male , MicroRNAs/genetics , Myosins/genetics , RNA, Messenger/genetics , RNA, Neoplasm/genetics , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Proteins/genetics
5.
PLoS Genet ; 16(4): e1008642, 2020 04.
Article in English | MEDLINE | ID: mdl-32310940

ABSTRACT

Undifferentiated embryonal sarcoma of the liver (UESL) is a rare and aggressive malignancy. Though the molecular underpinnings of this cancer have been largely unexplored, recurrent chromosomal breakpoints affecting a noncoding region on chr19q13, which includes the chromosome 19 microRNA cluster (C19MC), have been reported in several cases. We performed comprehensive molecular profiling on samples from 14 patients diagnosed with UESL. Congruent with prior reports, we identified structural variants in chr19q13 in 10 of 13 evaluable tumors. From whole transcriptome sequencing, we observed striking expressional activity of the entire C19MC region. Concordantly, in 7 of 7 samples undergoing miRNAseq, we observed hyperexpression of the miRNAs within this cluster to levels >100 fold compared to matched normal tissue or a non-C19MC amplified cancer cell line. Concurrent TP53 mutation or copy number loss was identified in all evaluable tumors with evidence of C19MC overexpression. We find that C19MC miRNAs exhibit significant negative correlation to TP53 regulatory miRNAs and K-Ras regulatory miRNAs. Using RNA-seq we identified that pathways relevant to cellular differentiation as well as mRNA translation machinery are transcriptionally enriched in UESL. In summary, utilizing a combination of next-generation sequencing and high-density arrays we identify the combination of C19MC hyperexpression via chromosomal structural event with TP53 mutation or loss as highly recurrent genomic features of UESL.


Subject(s)
Chromosome Breakpoints , Chromosomes, Human, Pair 19/genetics , Gene Expression Regulation, Neoplastic , Liver Neoplasms/genetics , MicroRNAs/genetics , Mutation , Neoplasms, Germ Cell and Embryonal/genetics , Sarcoma/genetics , Tumor Suppressor Protein p53/genetics , Aneuploidy , Child , Child, Preschool , Female , Genes, ras/genetics , Genomic Instability/genetics , Humans , Infant , Male , Transcription Initiation Site , Tumor Suppressor Protein p53/deficiency , Up-Regulation
6.
Biol Rev Camb Philos Soc ; 95(1): 244-266, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31663259

ABSTRACT

Metastasis is a pivotal event that changes the course of cancers from benign and treatable to malignant and difficult to treat, resulting in the demise of patients. Understanding the genetic control of metastasis is thus crucial to develop efficient and sustainable targeted therapies. Here we discuss the alterations in epigenetic mechanisms, transcription, chromosomal instability, chromosome imprinting, non-coding RNAs, coding RNAs, mutant RNAs, enhancers, G-quadruplexes, and copy number variation to dissect the genetic control of metastasis. We conclude that the genetic control of metastasis is predominantly executed through epithelial to mesenchymal transition and evasion of cell death. We discuss how genetic regulatory mechanisms can be harnessed for therapeutic purposes to achieve sustainable control over cancer metastasis.

7.
PLoS One ; 13(10): e0206008, 2018.
Article in English | MEDLINE | ID: mdl-30335837

ABSTRACT

Triple negative breast cancers (TNBCs) are known to express low PGR, ESR1, and ERBB2, and high KRT5, KRT14, and KRT17. However, the reasons behind the increased expressions of KRT5, KRT14, KRT17 and decreased expressions of PGR, ESR1, and ERBB2 in TNBCs are not fully understood. Here we show that, expression of chromosome 19 miRNA cluster (C19MC) specifically marks human TNBCs. Low REST and high CEBPB correlate with expression of C19MC, KRT5, KRT14, and KRT17 and enhancers of these genes/cluster are regulated by CEBPB and REST binding sites. The C19MC miRNAs in turn can potentially target REST to offer a positive feedback loop, and might target PGR, ESR1, ERBB2, GATA3, SCUBE2, TFF3 mRNAs to contribute towards TNBC phenotype. Thus our study demonstrates that C19MC miRNA expression marks TNBCs and that C19MC miRNAs and CEBPB might together determine the TNBC marker expression pattern.


Subject(s)
CCAAT-Enhancer-Binding Protein-beta/genetics , Chromosomes, Human, Pair 19/genetics , Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , Multigene Family , Triple Negative Breast Neoplasms/genetics , CCAAT-Enhancer-Binding Protein-beta/metabolism , Cell Line, Tumor , CpG Islands/genetics , DNA Methylation/genetics , Down-Regulation/genetics , Enhancer Elements, Genetic/genetics , Female , Humans , Keratins/genetics , MicroRNAs/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Triple Negative Breast Neoplasms/classification
8.
Oncoscience ; 4(9-10): 124-125, 2017 Sep.
Article in English | MEDLINE | ID: mdl-29142901
9.
Cancer Res ; 77(22): 6144-6156, 2017 11 15.
Article in English | MEDLINE | ID: mdl-28855211

ABSTRACT

Genomic instability and immune evasion are hallmarks of cancer. Apoptotic cancer stem cells can evade cell death by undergoing cellular transformation by constructing "blebbishields" from apoptotic bodies. In this study, we report a novel linkage between genomic instability and phagocytosis evasion that is coordinated by the blebbishield emergency program. Blebbishield emergency program evaded genomic instability checkpoint, expressed genomic instability-associated genes at distinct phases of cellular transformation, exhibited chromosomal instability, and promoted increase in nuclear size. Blebbishields fused with immune cells to evade phagocytosis, and the resultant hybrid cells exhibited increased migration, tumorigenesis, metastasis, red blood cell recruitment to tumors, and induced hepatosplenomegaly with signatures of genomic instability, blebbishield emergency program, and phagocytosis evasion to offer poor prognosis. Overall, our data demonstrate that the blebbishield emergency program drives evasion of chromosomal instability and phagocytosis checkpoints by apoptotic cancer stem cells. Cancer Res; 77(22); 6144-56. ©2017 AACR.


Subject(s)
Apoptosis/genetics , Genomic Instability , Neoplasms/genetics , Neoplastic Stem Cells/metabolism , Phagocytosis/genetics , Animals , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic , Humans , Male , Mice, Nude , Microscopy, Electron, Transmission , Neoplasms/metabolism , Neoplasms/pathology , Neoplastic Stem Cells/ultrastructure , Spheroids, Cellular/metabolism , Spheroids, Cellular/pathology , Spheroids, Cellular/ultrastructure , Transplantation, Heterologous
10.
Sci Rep ; 7(1): 9602, 2017 08 29.
Article in English | MEDLINE | ID: mdl-28851898

ABSTRACT

Cancer cells require both migratory and tumorigenic property to establish metastatic tumors outside the primary microenvironment. Identifying the characteristic features of migratory cancer stem cells with tumorigenic property is important to predict patient prognosis and combat metastasis. Here we established one epithelial and two mesenchymal cell lines from ascites of a bladder cancer patient (i.e. cells already migrated outside primary tumor). Analyses of these cell lines demonstrated that the epithelial cells with surface expression of PD-L1, E-cadherin, CD24, and VEGFR2 rapidly formed tumors outside the primary tumor microenvironment in nude mice, exhibited signatures of immune evasion, increased stemness, increased calcium signaling, transformation, and novel E-cadherin-RalBP1 interaction. The mesenchymal cells on the other hand, exhibited constitutive TGF-ß signaling and were less tumorigenic. Hence, targeting epithelial cancer stem cells with rapid tumorigenesis signatures in future might help to combat metastasis.


Subject(s)
B7-H1 Antigen/metabolism , CD24 Antigen/metabolism , Carcinoma/metabolism , Cell Membrane/metabolism , Cell Transformation, Neoplastic/metabolism , Neoplastic Stem Cells/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Biomarkers, Tumor , Carcinoma/genetics , Carcinoma/pathology , Cell Cycle , Cell Transformation, Neoplastic/genetics , DNA Fingerprinting , Epithelial-Mesenchymal Transition/genetics , Gene Expression Profiling , Humans , Microsatellite Repeats , Oligonucleotide Array Sequence Analysis , Reactive Oxygen Species/metabolism , Reproducibility of Results , Signal Transduction
11.
Cell Death Discov ; 3: 17023, 2017.
Article in English | MEDLINE | ID: mdl-28580172

ABSTRACT

Cancer stem cells evade apoptotic death by blebbishield emergency program, which constructs blebbishields from apoptotic bodies and drives cellular transformation. Von Hippel-Lindau (VHL) plays both tumor suppressor and oncogenic roles, and the reason behind is poorly understood. Here we demonstrate that dimers and trimers of p19-VHL interact with RalBP1 to construct blebbishields. Expression of RalBP1, p19-VHL, and high-molecular weight VHL is required to evade apoptosis by blebbishield-mediated transformation. In contrast, p30-VHL plays a tumor suppressor role by inhibiting blebbishield-mediated transformation. Furthermore, target genes of VHL that suppress oxidative stress were elevated during blebbishield-mediated cellular transformation. Thus, RalBP1 and p19-VHL play an oncogenic role, whereas p30-VHL plays a tumor suppressor role during the blebbishield emergency program by regulating oxidative stress management genes.

12.
Apoptosis ; 22(5): 719-729, 2017 05.
Article in English | MEDLINE | ID: mdl-28283889

ABSTRACT

Cancer stem cells are capable of undergoing cellular transformation after commencement of apoptosis through the blebbishield emergency program in a VEGF-VEGFR2-dependent manner. Development of therapeutics targeting the blebbishield emergency program would thus be important in cancer therapy. Specificity protein 1 (Sp1) orchestrates the transcription of both VEGF and VEGFR2; hence, Sp1 could act as a therapeutic target. Here, we demonstrate that CF3DODA-Me induced apoptosis, degraded Sp1, inhibited the expression of multiple drivers of the blebbishield emergency program such as VEGFR2, p70S6K, and N-Myc through activation of caspase-3, inhibited reactive oxygen species; and inhibited K-Ras activation to abolish transformation from blebbishields as well as transformation in soft agar. These findings confirm CF3DODA-Me as a potential therapeutic candidate that can induce apoptosis and block transformation from blebbishields.


Subject(s)
Apoptosis/genetics , Hydrocarbons, Fluorinated/administration & dosage , Sp1 Transcription Factor/genetics , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor Receptor-2/genetics , Apoptosis/drug effects , Caspase 3/genetics , Cell Line, Tumor , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic/drug effects , Glycyrrhetinic Acid/administration & dosage , Glycyrrhetinic Acid/analogs & derivatives , Humans , Hydrocarbons, Fluorinated/chemistry , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Reactive Oxygen Species/metabolism , Ribosomal Protein S6 Kinases, 70-kDa/genetics , Triterpenes/administration & dosage
13.
Biofactors ; 43(2): 181-186, 2017 Mar.
Article in English | MEDLINE | ID: mdl-27671897

ABSTRACT

Cancer stem cells can survive and undergo transformation after apoptosis by initiating robust endocytosis. Endocytosis in-turn drives formation of serpentine filopodia, which promote construction of blebbishields from apoptotic bodies. However, the status and role of macropinocytosis in blebbishields is not known. Here, we show by scanning electron microscopy and by macropinocytosis assays that blebbishields exhibit robust macropinocytosis. Inhibiting dynamin-mediated endocytosis does not affect macropinocytosis in blebbishields or in mitotic cells. In addition, inhibiting macropinocytosis did not inhibit construction of blebbishields from apoptotic bodies. Thus, although apoptotic cancer stem cells exhibit robust macropinocytosis, macropinocytosis is not essential to generate blebbishields, although it may play other roles in blebbishield biology. © 2016 BioFactors, 43(2):181-186, 2017.


Subject(s)
Mitosis/genetics , Neoplastic Stem Cells/metabolism , Pinocytosis , Apoptosis/genetics , Endocytosis/genetics , Humans , Microscopy, Electron, Scanning , Neoplastic Stem Cells/ultrastructure , Pseudopodia/metabolism , Pseudopodia/ultrastructure
14.
Apoptosis ; 21(11): 1203-1213, 2016 11.
Article in English | MEDLINE | ID: mdl-27651368

ABSTRACT

Pim kinases are being implicated in oncogenic process in various human cancers. Pim kinases primarily deal with three broad categories of functions such as tumorigenesis, protecting cells from apoptotic signals and evading immune attacks. Here in this review, we discuss the regulation of Pim kinases and their expression, and how these kinases defend cancer cells from therapeutic and immune attacks with special emphasis on how Pim kinases maintain their own expression during apoptosis and cellular transformation, defend mitochondria during apoptosis, defend cancer cells from immune attack, defend cancer cells from therapeutic attack, choose localization, self-regulation, activation of oncogenic transcription, metabolic regulation and so on. In addition, we also discuss how Pim kinases contribute to tumorigenesis by regulating cellular transformation and glycolysis to reinforce the importance of Pim kinases in cancer and cancer stem cells.


Subject(s)
Apoptosis , Neoplasms/enzymology , Proto-Oncogene Proteins c-pim-1/metabolism , Animals , Antineoplastic Agents/therapeutic use , Humans , Isoenzymes/genetics , Isoenzymes/metabolism , Neoplasms/drug therapy , Neoplasms/immunology , Neoplasms/physiopathology , Proto-Oncogene Proteins c-pim-1/genetics
15.
Article in English | MEDLINE | ID: mdl-27226900

ABSTRACT

The blebbishield emergency program helps to resurrect apoptotic cancer stem cells (CSCs) themselves. Understanding the mechanisms behind this program is essential to block resurrection of CSCs during cancer therapy. Here we demonstrate that endocytosis drives serpentine filopodia to construct blebbishields from apoptotic bodies and that a VEGF-VEGFR2-endocytosis-p70S6K axis governs subsequent transformation. Disengagement of RalGDS from E-cadherin initiates endocytosis of RalGDS and its novel interaction partners cdc42, VEGFR2, cleaved ß-catenin, and PKC-ζ as well as its known interaction partner K-Ras. We also report novel interactions of p45S6K (cleaved p70S6K) and PKM-ζ with PAK-1 filopodia-forming machinery specifically in blebbishields. Thus, a RalGDS-endocytosis-filopodia-VEGFR2-K-Ras-p70S6K axis drives the blebbishield emergency program, and therapeutic targeting of this axis might prevent resurrection of CSCs during cancer therapy.

16.
Sci Rep ; 6: 23965, 2016 Apr 04.
Article in English | MEDLINE | ID: mdl-27040869

ABSTRACT

Cancer stem cells are capable of transformation after apoptosis through the blebbishield emergency program. Reactive oxygen species (ROS) play an essential role in transformation. Understanding how ROS are linked to blebbishield-mediated transformation is necessary to develop efficient therapeutics that target the resurrection of cancer stem cells. Here we demonstrate that a novel PKC-ζ to p47(phox) interaction is required for ROS production in cancer cells. The combined use of the S6K inhibitor BI-D1870 with TNF-α inhibited the PKC-ζ to p47(phox) interaction, inhibited ROS production, degraded PKC-ζ, and activated caspases-3 and -8 to block transformation from blebbishields. BI-D1870 also inhibited transformation from cycloheximide-generated blebbishields. Thus ROS and the PKC-ζ to p47(phox) interaction are valid therapeutic targets to block transformation from blebbishields.


Subject(s)
Cell Transformation, Neoplastic/metabolism , NADPH Oxidases/metabolism , Protein Kinase C/metabolism , Reactive Oxygen Species/metabolism , Caspase 3/metabolism , Caspase 8/metabolism , Cell Line, Tumor , Cell Transformation, Neoplastic/drug effects , Cycloheximide/pharmacology , Humans , Neoplastic Stem Cells/metabolism , Protein Binding/drug effects , Pteridines/pharmacology , Tumor Necrosis Factor-alpha/pharmacology
17.
Urol Oncol ; 34(8): 337.e11-8, 2016 08.
Article in English | MEDLINE | ID: mdl-27038699

ABSTRACT

OBJECTIVE: The objective is to determine whether methyl 2-cyano-3,11-dioxo-18b-olean-1,12-dien-30-oate (CDODA-Me) has therapeutic potential in bladder cancer. We investigated the effects of CDODA-Me on the growth and survival of bladder cancer cells, and expression of specificity protein (Sp) transcription factors that regulate genes associated with cancer cell proliferation and survival. METHODS: J82, RT4P, and 253JB-V bladder cancer cell lines were treated with vehicle alone or with CDODA-Me with or without the antioxidant l-glutathione. Cell viability and DNA fragmentation were measured by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay and propidium iodide-fluorescence-activated cell sorting (FACS) analysis, respectively. Intracellular reactive oxygen species (ROS) were measured by 2',7'-dichlorofluorescin diacetate-FACS analysis. We assessed CDODA's effects on the levels of Sp and Sp-regulated proteins and induction of apoptosis in bladder cancer cells by Western blotting. We also assessed the anticancer effects of CDODA-Me in nude mice bearing RT4v6 bladder cancer. RESULTS: 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay and FACS analysis revealed that CDODA-Me inhibited the proliferation and survival of the 3 bladder cancer cell lines in a dose-dependent manner. FACS analysis also indicated that CDODA-Me-induced intracellular ROS, and Western blot analysis indicated that CDODA-Me decreased levels of Sp and Sp-regulated proteins and induced apoptosis in a dose-dependent and time-dependent manner. l-Glutathione attenuated CDODA-Me's down-regulation of Sp and Sp-regulated proteins. Compared with the control treatment, CDODA-Me substantially inhibited tumor growth in vivo. CONCLUSIONS: CDODA-Me has antineoplastic activity in bladder cancer cells by inducing ROS, which down-regulate Sp and Sp-regulated proteins. Thus, CDODA-Me has therapeutic potential in bladder cancer, and additional studies of the agent's efficacy and mode of action are warranted.


Subject(s)
Apoptosis , Glycyrrhetinic Acid/analogs & derivatives , Reactive Oxygen Species/metabolism , Sp Transcription Factors/metabolism , Urinary Bladder Neoplasms/drug therapy , Animals , Cell Line, Tumor , Cell Proliferation , Glycyrrhetinic Acid/pharmacology , Humans , Male , Mice , Mice, Nude , Xenograft Model Antitumor Assays
18.
Biochem J ; 473(1): 99-107, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26508734

ABSTRACT

Cancer cells are capable of sphere formation (transformation) through reactive oxygen species (ROS) and glycolysis shift. Transformation is linked to tumorigenesis and therapy resistance, hence targeting regulators of ROS and glycolysis is important for cancer therapeutic candidates. Here, we demonstrate that Smac mimetic AZ58 in combination with tumour necrosis factor-α (TNF-α) was able to inhibit the production of ROS, inhibit glycolysis through Pim-1 kinase-mediated Ser-112 phosphorylation of BAD, and increase depolarization of mitochondria. We also identified mitochondrial isoforms of Pim-1 kinase that were targeted for degradation by AZ58 in combination with TNF-α or AZ58 in combination with Fas ligand (FasL) plus cycloheximide (CHX) through caspase-3 to block transformation. Our study demonstrates that Smac mimetic in combination with TNF-α is an ideal candidate to target Pim-1 expression, inhibit ROS production and to block transformation from blebbishields.


Subject(s)
Biomimetic Materials/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Mitochondrial Proteins/metabolism , Proto-Oncogene Proteins c-pim-1/metabolism , Reactive Oxygen Species/metabolism , Transformation, Genetic/physiology , Tumor Necrosis Factor-alpha/metabolism , Apoptosis Regulatory Proteins , Biomimetic Materials/administration & dosage , Cell Line, Tumor , Humans , Intracellular Signaling Peptides and Proteins/administration & dosage , Mitochondrial Proteins/administration & dosage , Protein Isoforms/metabolism , Transformation, Genetic/drug effects , Tumor Necrosis Factor-alpha/administration & dosage
19.
Oncol Rep ; 34(5): 2738-44, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26323344

ABSTRACT

Bladder cancer is a common malignancy for which regional or metastatic disease is identified at diagnosis. The aim of this study was to determine whether tamoxifen (Tam), an estrogen receptor (ER) antagonist, can sensitize bladder cancer cell lines to gemcitabine (Gem) chemotherapy. ERα and ERß protein levels were determined in each cell line using western blot analysis. The TCC-Sup, 5637, and RT4 bladder cancer cells were exposed to various concentrations and regimens of Tam or Gem alone or in combination. Cell viability and apoptosis were assessed by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay and propidium iodide followed by flow cytometry. Apoptosis was then evaluated by western blot analysis. Treated TCC-Sup cells were subjected to soft agar colony formation assay to determine the cellular transformation. Western blot analysis results revealed ER expression in the three cell lines. TCC-Sup and 5637 cells treated with a combination of Tam and Gem had lower cell viabilities than those treated with Tam or Gem alone for 72 h in TCC-Sup and 5637. Compared with the other treatments, sequential Gem followed by Tam (Gem→Tam) treatment caused the largest increase in DNA fragmentation at 72 h in TCC-Sup cells. Western blot analysis results revealed that this sequential Gem→Tam treatment increased poly(ADP-ribose) polymerase cleavage in TCC-Sup cells. Sequential Gem→Tam inhibited the cell transformation in TCC-Sup cells. In conclusion, sequential Gem→Tam enhanced the cytotoxicity of Gem in vitro. This regimen be useful to enhance the efficacy of Gem in bladder cancer. However, future in vivo studies are required to verify the results.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Transformation, Neoplastic/drug effects , Deoxycytidine/analogs & derivatives , Tamoxifen/pharmacology , Urinary Bladder Neoplasms/drug therapy , Apoptosis , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Deoxycytidine/pharmacology , Drug Synergism , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Humans , Urinary Bladder Neoplasms/metabolism , Gemcitabine
20.
Urol Oncol ; 31(8): 1676-1682, 2013 Nov.
Article in English | MEDLINE | ID: mdl-22717623

ABSTRACT

OBJECTIVE: Intravesical bacillus Calmette-Guerin (BCG) is the gold standard for high-grade non-muscle-invasive bladder cancer (NMIBC); however, some patients do not respond to initial therapy while others relapse and/or progress. Therefore, combination strategies that can enhance the efficacy and sustainability of BCG are needed. Herein, we explore the efficacy of lenalidomide, a thalidomide derivative with immunomodulatory effects, in combination with BCG, both in vitro and in vivo. MATERIALS AND METHODS: We explored the outcomes of lenalidomide in combination with BCG in vivo using the MBT-2 cell line implanted in C3H immunocompetent mice. Apoptosis, cell proliferation, and microvessel density were measured by immunohistochemistry. In vitro, we performed Western blotting for cell cycle and apoptosis regulatory proteins and a chromatin condensation assay to evaluate TNF-α and FasL in combination with lenalidomide. RESULTS: In the mouse model, combination therapy with BCG and lenalidomide resulted in a statistically significant decrease in tumor size compared with the control group. IHC demonstrated a nonsignificant increase in apoptosis in the combination condition and no effect on cellular proliferation. Microvessel density was decreased in all treated conditions. In vitro, caspase-3 activation and chromatin condensation studies demonstrated increased cell death in the combinations of lenalidomide and TNF-α. CONCLUSIONS: The immunomodulatory molecule lenalidomide augments the response to BCG in an in vivo mouse model. This provides the rationale for studying the combination in patients with high grade NMIBC.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cell Proliferation/drug effects , Microvessels/drug effects , Urinary Bladder Neoplasms/drug therapy , Animals , Apoptosis/drug effects , BCG Vaccine/administration & dosage , BCG Vaccine/pharmacology , Blotting, Western , Caspase 3/metabolism , Cell Line, Tumor , Drug Synergism , Fas Ligand Protein/administration & dosage , Fas Ligand Protein/pharmacology , Female , Immunohistochemistry , Lenalidomide , Mice , Mice, Inbred C3H , Microvessels/metabolism , Microvessels/pathology , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Thalidomide/administration & dosage , Thalidomide/analogs & derivatives , Thalidomide/pharmacology , Tumor Burden/drug effects , Tumor Necrosis Factor-alpha/administration & dosage , Tumor Necrosis Factor-alpha/pharmacology , Urinary Bladder Neoplasms/metabolism , Urinary Bladder Neoplasms/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...