Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters











Publication year range
1.
Int J Biol Macromol ; 209(Pt A): 1205-1210, 2022 Jun 01.
Article in English | MEDLINE | ID: mdl-35461862

ABSTRACT

BJcuL is a snake venom C-type lectin (SVCTL) purified from the snake's venom Bothrops jararacussu. It has been previously demonstrated that BJcuL induces the accumulation of pro-apoptotic proteins of the extrinsic pathway, such as FADD and caspase-8, in the colorectal cancer cell line HT29, suggesting that the lectin may be able to enhance TRAIL-induced apoptosis. To test this hypothesis, we exposed two colorectal cancer cell lines, HT29 and HCT116, to increasing concentrations of BJcuL (1-20 µg/mL) in the presence or absence of TRAIL. Contrary to our expectations, however, BJcuL was unable to induce apoptosis in these cells, as shown by annexin-V/7AAD, clonogenic assays, and immunoblotting. Nevertheless, BJcuL was able to induce the accumulation of FADD and caspase-8, as well as anti-apoptotic proteins such as c-FLIP and survivin and poly-ubiquitinated proteins. Incubation with the deubiquitinase inhibitor WP1130 (10 µM) resulted in decreased BJcuL-induced survivin levels. Altogether, our results evince the effects of SVCTL on the ubiquitin-proteasome system in vitro for the first time. Compounds that can influence such system are important tools in the search for new therapeutic or diagnostic targets in cancer since they can elucidate the molecular mechanisms involved in determining cell fate as well as contributing to drug-development strategies in partnership with the pharmaceutical industry.


Subject(s)
Bothrops , Colorectal Neoplasms , Crotalid Venoms , Animals , Apoptosis Regulatory Proteins/metabolism , Bothrops/metabolism , Caspase 8 , Cell Line , Crotalid Venoms/pharmacology , Deubiquitinating Enzymes , Lectins, C-Type/metabolism , Snake Venoms , Survivin/metabolism
2.
BMC Cancer ; 21(1): 1248, 2021 Nov 19.
Article in English | MEDLINE | ID: mdl-34798868

ABSTRACT

BACKGROUND: Glioblastoma is a fatal brain tumour with a poor patient survival outcome. Hypoxia has been shown to reprogram cells towards a stem cell phenotype associated with self-renewal and drug resistance properties. Activation of hypoxia-inducible factors (HIFs) helps in cellular adaptation mechanisms under hypoxia. Similarly, miRNAs are known to be dysregulated in GBM have been shown to act as critical mediators of the hypoxic response and to regulate key processes involved in tumorigenesis. METHODS: Glioblastoma (GBM) cells were exposed to oxygen deprivation to mimic a tumour microenvironment and different cell aspects were analysed such as morphological changes and gene expression of miRNAs and survival genes known to be associated with tumorigenesis. RESULTS: It was observed that miR-128a-3p, miR-34-5p, miR-181a/b/c, were down-regulated in 6 GBM cell lines while miR-17-5p and miR-221-3p were upregulated when compared to a non-GBM control. When the same GBM cell lines were cultured under hypoxic microenvironment, a further 4-10-fold downregulation was observed for miR-34-5p, miR-128a-3p and 181a/b/c while a 3-6-fold upregulation was observed for miR-221-3p and 17-5p for most of the cells. Furthermore, there was an increased expression of SOX2 and Oct4, GLUT-1, VEGF, Bcl-2 and survivin, which are associated with a stem-like state, increased metabolism, altered angiogenesis and apoptotic escape, respectively. CONCLUSION: This study shows that by mimicking a tumour microenvironment, miRNAs are dysregulated, stemness factors are induced and alteration of the survival genes necessary for the cells to adapt to the micro-environmental factors occurs. Collectively, these results might contribute to GBM aggressiveness.


Subject(s)
Brain Neoplasms/genetics , Glioblastoma/genetics , MicroRNAs/metabolism , Tumor Hypoxia/genetics , Tumor Microenvironment/genetics , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Line, Tumor , Down-Regulation , Genotype , Glioblastoma/metabolism , Glioblastoma/pathology , Glucose Transporter Type 1/metabolism , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Octamer Transcription Factor-3/metabolism , Phenotype , Proto-Oncogene Proteins c-bcl-2/metabolism , SOXB1 Transcription Factors/metabolism , Survivin/metabolism , Up-Regulation , Vascular Endothelial Growth Factor A/metabolism
3.
Head Neck Pathol ; 15(4): 1127-1136, 2021 Dec.
Article in English | MEDLINE | ID: mdl-33840043

ABSTRACT

To evaluate molecular epithelial changes, we investigated whether a profile of survivin, cyclin dependent kinase inhibitor 2A (CDKN2A), epidermal growth factor receptor (EGFR), polo like kinase 1 (PLK1), p63, p40 (Δnp63 isoform), cyclin D1 (CCND1) and BCL2 apoptosis regulator (BCL2) proteins could predict malignant transformation. Different tissue segments (tumor adjacent epithelium; dysplasia and tumor) from a total of 109 patients were analyzed by immunohistochemistry. An increased expression of survivin (p < 0.001), PLK1 (p = 0.001), and p63 (p < 0.001) in parallel to reduced immunostaining of p40 (p < 0.001) and BCL2 (p = 0.029) was observed among the tissue segments analyzed. Our study revealed that survivin, PLK1, p63, p40 and BCL2 play a role in oral tumorigenesis and represent promising biomarkers able to recognize mesenchymal phenotype induction in the transition from nonmalignant cells to tumor cells. These results reveals critical interaction between survivin, PLK1, p63, p40 promising proteins during invasive carcinoma development.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Mouth Mucosa/metabolism , Mouth Neoplasms/metabolism , Biomarkers, Tumor/metabolism , Carcinoma, Squamous Cell/pathology , Cell Cycle Proteins/metabolism , Cell Transformation, Neoplastic/pathology , Cyclin D1/metabolism , Cyclin-Dependent Kinase Inhibitor p16/metabolism , ErbB Receptors/metabolism , Female , Humans , Immunohistochemistry , Male , Middle Aged , Mouth Neoplasms/pathology , Protein Isoforms , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Survivin/metabolism , Transcription Factors/metabolism , Polo-Like Kinase 1
4.
Int J Mol Sci ; 21(9)2020 May 08.
Article in English | MEDLINE | ID: mdl-32397263

ABSTRACT

Photodynamic therapy (PDT) has been used to treat certain types of non-melanoma skin cancer with promising results. However, some skin lesions have not fully responded to this treatment, suggesting a potential PDT-resistant phenotype. Therefore, novel therapeutic alternatives must be identified that improve PDT in resistant skin cancer. In this study, we analyzed the cell viability, intracellular protoporphyrin IX (PpIX) content and subcellular localization, proliferation profile, cell death, reactive oxygen species (ROS) detection and relative gene expression in PDT-resistant HSC-1 cells. PDT-resistant HSC-1 cells show a low quantity of protoporphyrin IX and low levels of ROS, and thus a low rate of death cell. Furthermore, the resistant phenotype showed a downregulation of HSPB1, SLC15A2, FECH, SOD2 and an upregulation of HMBS and BIRC5 genes. On the other hand, epigallocatechin gallate catechin enhanced the MAL-PDT effect, increasing levels of protoporphyrin IX and ROS, and killing 100% of resistant cells. The resistant MAL-PDT model of skin cancer squamous cells (HSC-1) is a reliable and useful tool to understand PDT cytotoxicity and cellular response. These resistant cells were successfully sensitized with epigallocatechin gallate catechin. The in vitro epigallocatechin gallate catechin effect as an enhancer of MAL-PDT in resistant cells is promising in the treatment of difficult skin cancer lesions.


Subject(s)
Anticarcinogenic Agents/pharmacology , Carcinoma, Squamous Cell/drug therapy , Catechin/analogs & derivatives , Cell Death/drug effects , Cell Proliferation/drug effects , Combined Modality Therapy/methods , Photochemotherapy/methods , Skin Neoplasms/drug therapy , Aminolevulinic Acid/analogs & derivatives , Aminolevulinic Acid/pharmacology , Carcinoma, Squamous Cell/radiotherapy , Catechin/pharmacology , Cell Death/radiation effects , Cell Hypoxia/drug effects , Cell Hypoxia/genetics , Cell Hypoxia/radiation effects , Cell Line, Tumor , Cell Proliferation/radiation effects , Cell Survival/drug effects , Cell Survival/radiation effects , Ferrochelatase/genetics , Ferrochelatase/metabolism , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Humans , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Photosensitizing Agents/metabolism , Protoporphyrins/metabolism , Reactive Oxygen Species/metabolism , Skin Neoplasms/radiotherapy , Stress, Physiological/drug effects , Stress, Physiological/genetics , Stress, Physiological/radiation effects , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Survivin/genetics , Survivin/metabolism , Symporters/genetics , Symporters/metabolism
5.
Cells ; 9(3)2020 03 13.
Article in English | MEDLINE | ID: mdl-32183150

ABSTRACT

(1) Aim: In the present paper we analyzed the transcriptome of CSCs (Cancer Stem Cells), in order to find defining molecular processes of breast cancer. (2) Methods: We performed RNA-Seq from CSCs isolated from the basal cell line MDA-MB-468. Enriched processes and networks were studied using the IPA (Ingenuity Pathway Analysis) tool. Validation was performed with qRT-PCR and the analysis of relevant genes was evaluated by overexpression, flow cytometry and in vivo zebrafish studies. Finally, the clinical relevance of these results was assessed using reported cohorts. (3) Results: We found that CSCs presented marked differences from the non-CSCs, including enrichment in transduction cascades related to stemness, cellular growth, proliferation and apoptosis. Interestingly, CSCs overexpressed a module of co-regulated Chromosomal Passenger Proteins including BIRC5 (survivin), INCENP and AURKB. Overexpression of BIRC5 increased the number of CSCs, as assessed by in vitro and in vivo zebrafish xenotransplant analyses. Analysis of previously published cohorts showed that this co-regulated module was not only overexpressed in basal breast tumors but also associated with relapse-free and overall survival in these patients. (4) Conclusions: These results underline the importance of Cancer Stem Cells in breast cancer progression and point toward the possible use of chromosomal passenger proteins as prognostic factors.


Subject(s)
Breast Neoplasms/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Neoplastic Stem Cells/metabolism , Animals , Apoptosis , Aurora Kinase B/genetics , Aurora Kinase B/metabolism , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Differentiation , Cell Line, Tumor , Cell Proliferation , Chromosomal Proteins, Non-Histone/genetics , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , Humans , Neoplastic Stem Cells/pathology , Prognosis , Survivin/genetics , Survivin/metabolism , Zebrafish
6.
Clin Transl Oncol ; 22(9): 1491-1498, 2020 Sep.
Article in English | MEDLINE | ID: mdl-31965535

ABSTRACT

BACKGROUND: This work aimed to investigate the inhibitory effect of regorafenib in combination with ginsenoside on the growth of HepG2 liver cancer cells. METHODS: HepG2 liver cancer cells were divided into blank control group, regorafenib single-drug group, ginsenoside single-drug group, and regorafenib/ginsenoside combination group. Cells in the regorafenib single-drug group were treated with regorafenib at 0.25 mg/L, 0.5 mg/L, and 1 mg/L, respectively, while cells in the ginsenoside single-drug group were treated with ginsenoside at 5.0 mg/L, 10.0 mg/L, and 20.0 mg/L, respectively. HepG2 cell proliferation, expression of survivin mRNA, and the apoptotic effector caspase-3 in HepG2 liver cancer cells were assessed. RESULTS: An inhibitory effect on the growth of HepG2 liver cancer cells was observed for both the single-drug therapies and the combination therapy. The synergistic inhibitory effect presented by the combination therapy was dependent on the gradient concentration and treatment time. RT-qPCR results showed that both regorafenib and ginsenoside significantly reduced the expression of survivin mRNA in HepG2 liver cancer cells and the expression level of survivin mRNA in the regorafenib/ginsenoside combination group was much lower than those in the regorafenib single-drug group and ginsenoside single-drug group. The two drugs demonstrated synergistic inhibitory effect when used in combination. CONCLUSIONS: The findings in this study offered a theoretical insight into clinical use of regorafenib and ginsenoside for treatment of liver cancer.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Caspase 3/biosynthesis , Ginsenosides/pharmacology , Liver Neoplasms/drug therapy , Phenylurea Compounds/pharmacology , Pyridines/pharmacology , Survivin/biosynthesis , Apoptosis/drug effects , Caspase 3/genetics , Caspase 3/metabolism , Cell Proliferation/drug effects , Drug Synergism , Ginsenosides/administration & dosage , Hep G2 Cells , Humans , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Phenylurea Compounds/administration & dosage , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/pharmacology , Pyridines/administration & dosage , Survivin/genetics , Survivin/metabolism
7.
Hum Exp Toxicol ; 39(3): 338-354, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31726875

ABSTRACT

Fridericia platyphylla (Cham.) L.G. Lohmann (FP) has cytotoxic, anti-inflammatory, and analgesic properties. We aimed to characterize the cytotoxic and antiproliferative effects of FP extract on normal (GAS) and tumor-derived (ACP02 and HepG2) cell lines. The effective concentrations (EC50s) by tetrazolium bromide assay (MTT) were 56.16, 43.68, and 42.57 µg mL-1 and 69.38, 41.73, and 52.39 µg mL-1 by neutral red assay for GAS, ACP02, and HepG2 cells, respectively. The extract decreased nuclear division indices, which was not reflected in cell proliferation curves. Flow cytometric analyses showed that even 30 µg mL-1 extract (shown to be noncytotoxic by MTT assay) increased the sub-G1 population, indicating cell death due to apoptosis and necrosis. A cytokinesis-block micronucleus cytome assay showed that 30 µg mL-1 of the extract increased the frequency of nuclear buds in tumor cells. Real-time quantitative polymerase chain reaction showed CCND1 upregulation in doxorubicin-treated GAS cells and BCL-XL, BIRC5, and MET downregulation in 5 or 30 µg mL-1 in FP extract-treated ACP02 cells. In conclusion, FP extract modulated apoptosis- and cell cycle-related genes and presented selective cytotoxicity toward tumor cells that deserves further investigation by testing other cell types. Our results demonstrated that even medicinal plants exert adverse effects depending on the extract concentrations used and tissues investigated.


Subject(s)
Bignoniaceae/chemistry , Plant Extracts/pharmacology , Plant Roots/chemistry , Stomach Neoplasms/drug therapy , Survivin/metabolism , bcl-X Protein/metabolism , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Down-Regulation/drug effects , Gene Expression Regulation/drug effects , Humans , Necrosis , Plant Extracts/chemistry , Proto-Oncogene Proteins c-met/genetics , Proto-Oncogene Proteins c-met/metabolism , Reactive Oxygen Species , Survivin/genetics , bcl-X Protein/genetics
8.
Viruses ; 11(11)2019 10 30.
Article in English | MEDLINE | ID: mdl-31671627

ABSTRACT

It is known that levels of the anti-apoptotic protein survivin are reduced during Murine norovirus MNV-1 and Feline calicivirus (FCV) infection as part of the apoptosis establishment required for virus release and propagation in the host. Recently, our group has reported that overexpression of survivin causes a reduction of FCV protein synthesis and viral progeny production, suggesting that survivin may affect early steps of the replicative cycle. Using immunofluorescence assays, we observed that overexpression of survivin, resulted in the reduction of FCV infection not only in transfected but also in the neighboring nontransfected CrFK cells, thus suggesting autocrine and paracrine protective effects. Cells treated with the supernatants collected from CrFK cells overexpressing survivin showed a reduction in FCV but not MNV-1 protein production and viral yield, suggesting that FCV binding and/or entry were specifically altered. The reduced ability of FCV to bind to the surface of the cells overexpressing survivin, or treated with the supernatants collected from these cells, correlate with the reduction in the cell surface of the FCV receptor, the feline junctional adhesion molecule (fJAM) 1, while no effect was observed in the cells transfected with the pAm-Cyan vector or in cells treated with the corresponding supernatants. Moreover, the overexpression of survivin affects neither Vaccinia virus (VACV) production in CrFK cells nor MNV-1 virus production in RAW 267.4 cells, indicating that the effect is specific for FCV. All of these results taken together indicate that cells that overexpress survivin, or cell treatment with the conditioned medium from these cells, results in the reduction of the fJAM-1 molecule and, therefore, a specific reduction in FCV entry and infection.


Subject(s)
Caliciviridae Infections/virology , Calicivirus, Feline/physiology , Survivin/metabolism , Animals , Caliciviridae Infections/genetics , Caliciviridae Infections/metabolism , Calicivirus, Feline/metabolism , Cats , Cell Line , Culture Media, Conditioned/metabolism , Culture Media, Conditioned/pharmacology , Gene Expression , Host-Pathogen Interactions , Junctional Adhesion Molecules/metabolism , Receptors, Virus/metabolism , Species Specificity , Survivin/genetics , Viral Proteins/biosynthesis , Virus Internalization/drug effects , Virus Replication/drug effects
9.
Int J Mol Sci ; 20(20)2019 Oct 12.
Article in English | MEDLINE | ID: mdl-31614718

ABSTRACT

Drug resistance represents a major issue in treating breast cancer, despite the identification of novel therapeutic strategies, biomarkers, and subgroups. We have previously identified the LQB-223, 11a-N-Tosyl-5-deoxi-pterocarpan, as a promising compound in sensitizing doxorubicin-resistant breast cancer cells, with little toxicity to non-neoplastic cells. Here, we investigated the mechanisms underlying LQB-223 antitumor effects in 2D and 3D models of breast cancer. MCF-7 and MDA-MB-231 cells had migration and motility profile assessed by wound-healing and phagokinetic track motility assays, respectively. Cytotoxicity in 3D conformation was evaluated by measuring spheroid size and performing acid phosphatase and gelatin migration assays. Protein expression was analyzed by immunoblotting. Our results show that LQB-223, but not doxorubicin treatment, suppressed the migratory and motility capacity of breast cancer cells. In 3D conformation, LQB-223 remarkably decreased cell viability, as well as reduced 3D culture size and migration. Mechanistically, LQB-223-mediated anticancer effects involved decreased proteins levels of XIAP, c-IAP1, and Mcl-1 chemoresistance-related proteins, but not survivin. Survivin knockdown partially potentiated LQB-223-induced cytotoxicity. Additionally, cell treatment with LQB-223 resulted in changes in the mRNA levels of epithelial-mesenchymal transition markers, suggesting that it might modulate cell plasticity. Our data demonstrate that LQB-223 impairs 3D culture growth and migration in 2D and 3D models of breast cancer exhibiting different phenotypes.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis , Breast Neoplasms/metabolism , Drug Resistance, Neoplasm , Pterocarpans/pharmacology , Antineoplastic Agents/toxicity , Cell Movement , Cell Proliferation , Female , Humans , Inhibitor of Apoptosis Proteins/metabolism , MCF-7 Cells , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Pterocarpans/toxicity , Spheroids, Cellular/drug effects , Survivin/genetics , Survivin/metabolism , Tumor Cells, Cultured , X-Linked Inhibitor of Apoptosis Protein/metabolism
10.
Virology ; 527: 146-158, 2019 01 15.
Article in English | MEDLINE | ID: mdl-30529563

ABSTRACT

Calicivirus infection causes intrinsic apoptosis, leading to viral propagation in the host. During murine norovirus infection, a reduction in the anti-apoptotic protein survivin has been documented. Here we report that in feline calicivirus infection, a downregulation of the anti-apoptotic proteins survivin and XIAP occur, which correlates with the translocation of the pro-apoptotic protein Smac/DIABLO from the mitochondria to the cytoplasm and the activation of caspase-3. Inhibition of survivin degradation by lactacystin treatment caused a delay in apoptosis progression, reducing virus release, without affecting virus production. However, the overexpression of survivin caused a negative effect in viral progeny production. Overexpression of the leader of the capsid protein (LC), but not of the protease-polymerase NS6/7, results in the downregulation of survivin and XIAP, caspase activation and mitochondrial damage. These results indicate that LC is responsible for the induction of apoptosis in transfected cells and most probably in FCV infection.


Subject(s)
Apoptosis , Caliciviridae Infections/metabolism , Calicivirus, Feline/physiology , Capsid Proteins/metabolism , Down-Regulation , Survivin/genetics , X-Linked Inhibitor of Apoptosis Protein/genetics , Animals , Caliciviridae Infections/virology , Capsid Proteins/chemistry , Cats , Cell Line , Gene Expression , Host-Pathogen Interactions , Mitochondrial Proteins/metabolism , Protein Transport , Survivin/metabolism , Viral Proteins/biosynthesis , Virus Replication
11.
Bioorg Med Chem ; 27(2): 375-382, 2019 01 15.
Article in English | MEDLINE | ID: mdl-30579801

ABSTRACT

Malignant neoplasms are one of the leading causes of death worldwide and hematologic malignancies, including acute leukemia (AL) is one of the most relevant cancer types. Current available chemotherapeutics are associated with high morbidity and mortality rates, therefore, the search for new molecules with antitumor activity, specific and selective for neoplastic cells, became a great challenge for researchers in the oncology field. As pyrazolines stand out in the literature for their great variety of biological activities, the aim of this study was to synthesize and evaluate the antileukemic activity of five new pyrazoline derivatives. All pyrazolines showed adequate physicochemical properties for a good oral bioavailability. The two unpublished and most effective pyrazoline derivatives have been selected for further experiments. These compounds are highly selective for leukemic cells when compared to non-neoplastic cells and did not cause lysis on human red blood cells. Additionally, selected pyrazolines induced cell cycle arrest at G0/G1 phase and decreased cell proliferation marker KI67. Apoptotic cell death induced by selected pyrazolines was confirmed by morphological analysis, assessment of phosphatidylserine residue exposure and DNA fragmentation. Several factors indicate that both intrinsic and extrinsic apoptosis occurred. These were: increased FasR expression; the predominance of Bax in relation to Bcl-2; the loss of mitochondrial membrane potential; AIF release; decreased expression of survivin (an antiapoptotic protein); and the activation of caspase-3. The selected pyrazolines were also found to be cytotoxic against neoplastic cells collected from the peripheral blood and bone marrow of patients with different subtypes of acute leukemia.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Pyrazoles/pharmacology , Acute Disease , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Apoptosis Inducing Factor/metabolism , Caspase 3/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , Humans , Leukemia/drug therapy , Membrane Potential, Mitochondrial/drug effects , Proto-Oncogene Proteins c-bcl-2/metabolism , Pyrazoles/chemical synthesis , Pyrazoles/chemistry , S Phase Cell Cycle Checkpoints/drug effects , Survivin/metabolism , bcl-2-Associated X Protein/metabolism
12.
Biomed Pharmacother ; 107: 464-474, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30107342

ABSTRACT

Cardiac glycosides (CGs) are natural compounds used to treat congestive heart failure. They have garnered attention as a potential cancer treatment option, especially because they bind to Na+/K+-ATPase as a target and activate intracellular signaling pathways leading to a variety of cellular responses. In this study we evaluated AMANTADIG, a semisynthetic cardenolide derivative, for its cytotoxic activity in two human androgen-insensitive prostate carcinoma cell lines, and the potential synergistic effects with docetaxel. AMANTADIG induced cytotoxic effects in both cell lines, and a combination with docetaxel showed a moderate and strong synergism in DU145 and PC-3 cells, respectively, at concentrations considerably lower than their IC50 values. Cell cycle analyses showed that AMANTADIG and its synergistic combination induced G2/M arrest of DU145 and PC-3 cells by modulating Cyclin B1, CDK1, p21 and, mainly, survivin expression, a promising target in cancer therapy. Furthermore, AMANTADIG presented reduced toxicity toward non-cancerous cell type (PBMC), and computational docking studies disclosed high-affinity binding to the Na+/K+-ATPase α subunit, a result that was experimentally confirmed by Na+/K+-ATPase inhibition assays. Hence, AMANTADIG inhibited Na+/K+-ATPase activity in PC-3 cells, as well as in purified pig kidney at nanomolar range. Altogether, these data highlight the potent effects of AMANTADIG in combination with docetaxel and offer important insights for the development of more effective and selective therapies against prostate cancer.


Subject(s)
Apoptosis/drug effects , Digitoxigenin/analogs & derivatives , Docetaxel/pharmacology , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/pathology , Sodium-Potassium-Exchanging ATPase/antagonists & inhibitors , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Digitoxigenin/chemistry , Digitoxigenin/pharmacology , Drug Synergism , Gene Expression Regulation, Neoplastic/drug effects , Humans , Male , Molecular Docking Simulation , Necrosis , Prostatic Neoplasms/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism , Survivin/genetics , Survivin/metabolism
13.
Sci Rep ; 7(1): 6707, 2017 07 27.
Article in English | MEDLINE | ID: mdl-28751665

ABSTRACT

Molecular data generation and their combination in penile carcinomas (PeCa), a significant public health problem in poor and underdeveloped countries, remain virtually unexplored. An integrativemethodology combin ing genome-wide copy number alteration, DNA methylation, miRNA and mRNA expression analysis was performed in a set of 20 usual PeCa. The well-ranked 16 driver candidates harboring genomic alterations and regulated by a set of miRNAs, including hsa-miR-31, hsa-miR-34a and hsa-miR-130b, were significantly associated with over-represented pathways in cancer, such as immune-inflammatory system, apoptosis and cell cycle. Modules of co-expressed genes generated from expression matrix were associated with driver candidates and classified according to the over-representation of passengers, thus suggesting an alteration of the pathway dynamics during the carcinogenesis. This association resulted in 10 top driver candidates (AR, BIRC5, DNMT3B, ERBB4, FGFR1, PML, PPARG, RB1, TNFSF10 and STAT1) selected and confirmed as altered in an independent set of 33 PeCa samples. In addition to the potential driver genes herein described, shorter overall survival was associated with BIRC5 and DNMT3B overexpression (log-rank test, P = 0.026 and P = 0.002, respectively) highlighting its potential as novel prognostic marker for penile cancer.


Subject(s)
Carcinoma/genetics , DNA (Cytosine-5-)-Methyltransferases/genetics , Gene Expression Regulation, Neoplastic , Neoplasm Proteins/genetics , Penile Neoplasms/genetics , Survivin/genetics , Aged , Apoptosis/genetics , Carcinogenesis/genetics , Carcinogenesis/metabolism , Carcinogenesis/pathology , Carcinoma/diagnosis , Carcinoma/metabolism , Carcinoma/mortality , Case-Control Studies , Cell Cycle/genetics , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Copy Number Variations , DNA Methylation , Epithelial Cells/metabolism , Epithelial Cells/pathology , Genome, Human , Humans , Male , MicroRNAs/genetics , MicroRNAs/metabolism , Middle Aged , Multigene Family , Neoplasm Proteins/metabolism , Penile Neoplasms/diagnosis , Penile Neoplasms/metabolism , Penile Neoplasms/mortality , Prognosis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction , Survival Analysis , Survivin/metabolism , DNA Methyltransferase 3B
SELECTION OF CITATIONS
SEARCH DETAIL