Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 375
Filter
1.
Int J Mol Sci ; 22(4)2021 Feb 12.
Article in English | MEDLINE | ID: mdl-33673050

ABSTRACT

The BIRC (baculoviral IAP repeat-containing; BIRC) family genes encode for Inhibitor of Apoptosis (IAP) proteins. The dysregulation of the expression levels of the genes in question in cancer tissue as compared to normal tissue suggests that the apoptosis process in cancer cells was disturbed, which may be associated with the development and chemoresistance of triple negative breast cancer (TNBC). In our study, we determined the expression level of eight genes from the BIRC family using the Real-Time PCR method in patients with TNBC and compared the obtained results with clinical data. Additionally, using bioinformatics tools (Ualcan and The Breast Cancer Gene-Expression Miner v4.5 (bc-GenExMiner v4.5)), we compared our data with the data in the Cancer Genome Atlas (TCGA) database. We observed diverse expression pattern among the studied genes in breast cancer tissue. Comparing the expression level of the studied genes with the clinical data, we found that in patients diagnosed with breast cancer under the age of 50, the expression levels of all studied genes were higher compared to patients diagnosed after the age of 50. We observed that in patients with invasion of neoplastic cells into lymphatic vessels and fat tissue, the expression levels of BIRC family genes were lower compared to patients in whom these features were not noted. Statistically significant differences in gene expression were also noted in patients classified into three groups depending on the basis of the Scarff-Bloom and Richardson (SBR) Grading System.


Subject(s)
Databases, Nucleic Acid , Gene Expression Regulation, Neoplastic , Inhibitor of Apoptosis Proteins , Multigene Family , Neoplasm Proteins , Triple Negative Breast Neoplasms , Adult , Aged , Female , Humans , Inhibitor of Apoptosis Proteins/biosynthesis , Inhibitor of Apoptosis Proteins/genetics , Middle Aged , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/metabolism
2.
Oncol Rep ; 45(4)2021 04.
Article in English | MEDLINE | ID: mdl-33649820

ABSTRACT

Anaplastic thyroid cancer (ATC) is characterized by a rapid and aggressive course of progression. Despite significant advances in surgery, radiotherapy and chemotherapy, the disease­specific mortality due to ATC is approximately 100%. New strategies, such as molecular targeted therapies, are imperative for improving survival. Livin, a member of the human inhibitor of apoptosis protein family, has been found to be associated with tumor progression and poor prognosis in various human cancers. The aim of the present study was to evaluate the role of Livin in cancer progression and chemoradioresistance of ATC and to investigate its potential as a therapeutic target. Endogenous Livin expression in the human BHT101 ATC cell line was silenced by Livin­specific small interfering RNA. To assess the impact of Livin on cancer cell behavior in human ATC cells, various methods such as cell invasion, cell viability and cell apoptosis assays were applied. To assess the expression of Livin and the change of apoptosis­related proteins associated with Livin expression, reverse transcription­quantitative PCR and western blotting were performed. Immunohistochemistry was performed to detect Livin protein expression in human ATC tissues. The association between Livin expression and apoptotic/proliferation index was analyzed in human ATC cells. Livin­knockdown suppressed tumor cell invasion; and conversely, it enhanced cell apoptosis, with elevated expression levels of cleaved caspase­3 and ­7 and cleaved PARP. Livin­knockdown enhanced radiation­induced apoptosis, while reducing cell viability following radiotherapy, as well as lenvatinib treatment. In addition, human ATC tissues with high Livin­expression exhibited a high Ki­67 labeling index and low apoptotic index. In summary, these findings indicate the contribution of Livin to tumor progression and chemoradioresistance in ATC.


Subject(s)
Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Inhibitor of Apoptosis Proteins/antagonists & inhibitors , Neoplasm Proteins/antagonists & inhibitors , Thyroid Carcinoma, Anaplastic/metabolism , Thyroid Carcinoma, Anaplastic/therapy , Thyroid Neoplasms/therapy , Adaptor Proteins, Signal Transducing/biosynthesis , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Aged , Apoptosis/drug effects , Apoptosis/radiation effects , Drug Resistance, Neoplasm , Female , Gene Knockdown Techniques , Humans , Inhibitor of Apoptosis Proteins/biosynthesis , Inhibitor of Apoptosis Proteins/genetics , Inhibitor of Apoptosis Proteins/metabolism , Male , Neoplasm Invasiveness , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Phenylurea Compounds/pharmacology , Quinolines/pharmacology , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , Radiation Tolerance , Thyroid Carcinoma, Anaplastic/genetics , Thyroid Neoplasms/genetics , Thyroid Neoplasms/metabolism
3.
Metab Brain Dis ; 36(4): 669-683, 2021 04.
Article in English | MEDLINE | ID: mdl-33547995

ABSTRACT

Tauopathies belong to a heterogeneous class of neuronal diseases resulting in the metabolic disturbance. A disulfide natural compound of Alpha-Lipoic acid (ALA) has shown numerous pharmacologic, antioxidant, and neuroprotective activities under neuropathological conditions. The aim of this study was to investigate the neuroprotective effects of ALA on the tauopathy-induced oxidative disturbance and behavioral deficits. The transgenic Drosophila model of tauopathy induced by human tauR406W using GAL4/UAS system and effects of ALA (0.001, 0.005, and 0.025 % w/w of diet) on the neuropathology of tau in younger (20 days) and older (30 days) adults were investigated via biochemical, molecular, behavioral and in-situ tissue analyses. Expression of apoptosis-related proteins involving Drosophila Cyt-c-d (trigger of intrinsic apoptosis) and DrICE (effector caspase) were upregulated in both ages (20 and 30 days) and DIAP1 (caspase inhibitor) has reduced only in older model flies compared to the controls. Remarkably, all doses of ALA increased DIAP1 and glutathione (GSH) as well as reducing Cyt-c-d and lipid peroxidation (LPO) in the younger flies compared to the model flies. Moreover, the higher doses of ALA were able to decrease thiol concentrations, to increase total antioxidant capacity, and to improve the behavioral deficits (locomotor function, olfactory memory, and ethanol sensitivity) in the younger flies. On the other hand, only a higher dose of ALA was able to decrease DrICE, Cyt-c-d, LPO, and thiol as well as increasing antioxidant capacity and decreasing ethanol sensitivity (ST50, RT50) in the older flies. TUNEL assay showed that all doses of ALA could potentially increase the DIAP1/DrICE ratio and exert anti-apoptotic effects on younger, but not on the older adults. Furthermore, data obtained from the in-situ ROS assay confirmed that only a higher dose of ALA significantly decreased the ROS level at both ages. Our data showed that an effective neuroprotective dose of ALA and its mechanism of action on this model of tauopathy could potentially be influenced by longevity. Moreover, it was shown that ALA prevents apoptosis and decreases the redox homeostasis, and this partially explains the mechanism by which ALA diminishes behavioral deficits.


Subject(s)
Caspases/biosynthesis , Drosophila Proteins/biosynthesis , Inhibitor of Apoptosis Proteins/biosynthesis , Locomotion/physiology , Oxidative Stress/physiology , Tauopathies/metabolism , Thioctic Acid/therapeutic use , Age Factors , Animals , Animals, Genetically Modified , Antioxidants/pharmacology , Antioxidants/therapeutic use , Apoptosis/drug effects , Apoptosis/physiology , Caspases/genetics , Drosophila , Drosophila Proteins/genetics , Female , Homeostasis/drug effects , Homeostasis/physiology , Inhibitor of Apoptosis Proteins/genetics , Locomotion/drug effects , Male , Oxidation-Reduction/drug effects , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Tauopathies/drug therapy , Tauopathies/genetics , Thioctic Acid/pharmacology
4.
Oxid Med Cell Longev ; 2020: 9084730, 2020.
Article in English | MEDLINE | ID: mdl-32322338

ABSTRACT

The knowledge of factors affecting the viability as well as proliferation and therapeutic potential of perinatal stem cells is of great importance for the decisions concerning their collection, multiplication, and storing. The aim of this work is to evaluate the expression of the BIRC2, BIRC3, and BIRC5 genes at the level of transcription in mesenchymal stem cells derived from the umbilical cord Wharton's jelly. The study examined the relationship between the expression level of the studied genes and selected biophysical parameters of umbilical blood: pH, pCO2, pO2, and cHCO3. Moreover, the relationship between the pregnant age, the type of delivery (natural delivery or cesarean section), and the level of expression of the BIRC2, BIRC3, and BIRC5 genes was assessed. The research was carried out on mesenchymal stem cells derived from the umbilical cord Wharton's jelly (WJSC) taken from 55 women immediately after delivery. Expression of the examined genes was assessed with the qPCR method using commercially available reagent kits. On the basis of the conducted research, it was demonstrated that WJSCs collected from younger women giving birth naturally, and in the acidic environment of the umbilical cord blood, are characterized by a higher expression of the BIRC2, BIRC3, and BIRC5 genes. It was shown that the expression of the BIRC2 and BIRC3 genes in Wharton's jelly mesenchymal stem cells declines with the mother's age. Our research suggests that stem cells collected from younger women giving birth naturally can be more resistant to apoptosis and show a more stem cell-like character, which can increase their therapeutic potential and clinical utility, but this conclusion needs to be approved in the next studies.


Subject(s)
Baculoviral IAP Repeat-Containing 3 Protein/biosynthesis , Fetal Blood/metabolism , Inhibitor of Apoptosis Proteins/biosynthesis , Mesenchymal Stem Cells/metabolism , Survivin/biosynthesis , Ubiquitin-Protein Ligases/biosynthesis , Wharton Jelly/metabolism , Adult , Baculoviral IAP Repeat-Containing 3 Protein/genetics , Baculoviral IAP Repeat-Containing 3 Protein/metabolism , Female , Humans , Inhibitor of Apoptosis Proteins/genetics , Inhibitor of Apoptosis Proteins/metabolism , Pregnancy , Survivin/genetics , Survivin/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Wharton Jelly/cytology , Young Adult
5.
Int J Mol Med ; 45(4): 1103-1111, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32124942

ABSTRACT

A pterygium is an inflammatory, invasive and proliferative lesion on the ocular surface, which can decrease visual acuity, damage the ocular surface and affect the appearance of the eye. However, the underlying molecular mechanisms of the pathogenesis remain unclear. In the present study, the role of apoptosis­associated protein Livin in the occurrence and development of pterygium was investigated. Primary samples from quiescent or advanced clinical stages of pterygium and normal human conjunctival tissues were used to assess mRNA and protein expression levels of Livin using reverse transcription­quantitative PCR and immunohistochemistry, respectively. Livin was knocked down in pterygium epithelial cells (PECs) using small interfering RNA (siRNA), to investigate the role of Livin in PEC viability, migration, invasion ability and apoptosis. The cell viability, invasion ability and apoptosis of PECs following ultraviolet B (UVB) radiation alone or in combination with Livin silencing were also analyzed. Expression levels of Livin increased in the pterygium tissues compared with those in the normal conjunctiva at both the mRNA and protein levels. Livin expression levels in advanced pterygium were significantly higher compared with those in quiescent pterygium samples. Knockdown of Livin expression levels significantly reduced cell migration, invasion ability and cell viability, and induced apoptosis of PECs. Inhibition of Livin expression in PECs increased the expression levels of caspase­7, caspase­3 and E­cadherin, whereas expression levels of Snail were downregulated. Cell viability and invasion ability in PECs was enhanced following UVB radiation and Livin expression upregulated. UVB irradiation induced cell invasion ability of PECs and this was attenuated by Livin­silencing. Transfection with Livin siRNA also partially recovered the apoptosis rate of PECs, which was reduced by UVB irradiation. In conclusion, Livin was upregulated in pterygium, and UVB radiation functions in the development of pterygium by inducing Livin expression.


Subject(s)
Adaptor Proteins, Signal Transducing/biosynthesis , Conjunctiva/metabolism , Epithelial Cells/metabolism , Inhibitor of Apoptosis Proteins/biosynthesis , Neoplasm Proteins/biosynthesis , Pterygium/metabolism , Ultraviolet Rays/adverse effects , Up-Regulation/radiation effects , Adaptor Proteins, Signal Transducing/genetics , Adult , Aged , Cell Line , Conjunctiva/pathology , Epithelial Cells/pathology , Female , Humans , Inhibitor of Apoptosis Proteins/genetics , Male , Middle Aged , Neoplasm Proteins/genetics , Pterygium/genetics , Pterygium/pathology , Up-Regulation/genetics
6.
Oncol Res ; 27(9): 1035-1042, 2019 Sep 23.
Article in English | MEDLINE | ID: mdl-30982490

ABSTRACT

Although arsenic trioxide (ATO) is a well-known antileukemic drug used for acute promyelocytic leukemia treatment, the development of ATO resistance is still a big challenge. We previously reported that microRNA-204 (miR-204) was involved in the regulation of acute myeloid leukemia (AML) cell apoptosis, but its role in chemoresistance is poorly understood. In the present study, we showed that miR-204 was significantly increased in AML cells after ATO treatment. Interestingly, the increased miR-204 level that was negatively correlated with ATO induced the decrease in cell viability and baculoviral inhibition of apoptosis protein repeat-containing 6 (BIRC6) expression. Overexpression of miR-204 potentiated ATO-induced AML cell growth inhibition and apoptosis. Furthermore, miR-204 directly targets to the 3'-UTR of BIRC6. Upregulation of miR-204 decreased BIRC6 luciferase activity and expression, which subsequently enhanced the expression of p53. Restoration of BIRC6 markedly reversed the effect of miR-204 on the regulation of AML cell sensitivity to ATO. Taken together, our study demonstrates that miR-204 decreases ATO chemoresistance in AML cells at least partially via promoting BIRC6/p53-mediated apoptosis. miR-204 represents a novel target of ATO, and upregulation of miR-204 may be a useful strategy to improve the efficacy of ATO in AML treatment.


Subject(s)
Arsenic Trioxide/pharmacology , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , MicroRNAs/metabolism , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , HL-60 Cells , Humans , Inhibitor of Apoptosis Proteins/biosynthesis , Inhibitor of Apoptosis Proteins/genetics , Leukemia, Myeloid, Acute/metabolism , MicroRNAs/genetics , Tumor Suppressor Protein p53/biosynthesis , Tumor Suppressor Protein p53/genetics , Up-Regulation/drug effects
7.
Cell Death Dis ; 10(2): 57, 2019 01 22.
Article in English | MEDLINE | ID: mdl-30670698

ABSTRACT

Physiological stress conditions attenuate global mRNA translation via modifications of key eukaryotic initiation factors. However, non-canonical translation initiation mechanisms allow cap-independent translation of certain mRNAs. We have previously demonstrated that eIF5B promotes cap-independent translation of the mRNA encoding the antiapoptotic factor, XIAP, during cellular stress. Here, we show that depletion of eIF5B sensitizes glioblastoma multiforme cells to TRAIL-induced apoptosis by a pathway involving caspases-8, -9, and -7, with no significant effect on cell cycle progression. eIF5B promotes evasion of apoptosis by promoting the translation of several IRES-containing mRNAs, encoding the antiapoptotic proteins XIAP, Bcl-xL, cIAP1, and c-FLIPS. We also show that eIF5B promotes translation of nuclear factor erythroid 2-related factor 2 and suggest that reactive oxygen species contribute to increased apoptosis under conditions of eIF5B depletion. Finally, eIF5B depletion leads to decreased activation of the canonical NF-κB pathway. Taken together, our data suggest that eIF5B represents a regulatory node, allowing cancer cells to evade apoptosis by promoting the translation of pro-survival proteins from IRES-containing mRNAs.


Subject(s)
Eukaryotic Initiation Factors/genetics , Glioblastoma/genetics , Glioblastoma/pathology , Apoptosis/drug effects , Apoptosis/physiology , Cell Line, Tumor , Cell Survival/physiology , Cyclin-Dependent Kinase Inhibitor p21/biosynthesis , Cyclin-Dependent Kinase Inhibitor p21/genetics , Drug Resistance, Neoplasm , Eukaryotic Initiation Factors/deficiency , Eukaryotic Initiation Factors/metabolism , Glioblastoma/drug therapy , Glioblastoma/metabolism , HEK293 Cells , Humans , Inhibitor of Apoptosis Proteins/biosynthesis , Inhibitor of Apoptosis Proteins/genetics , NF-E2-Related Factor 2/biosynthesis , NF-E2-Related Factor 2/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , Recombinant Proteins/pharmacology , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Transfection , Up-Regulation , X-Linked Inhibitor of Apoptosis Protein/biosynthesis , X-Linked Inhibitor of Apoptosis Protein/genetics , bcl-X Protein/biosynthesis , bcl-X Protein/genetics
8.
Medicine (Baltimore) ; 97(31): e11697, 2018 Aug.
Article in English | MEDLINE | ID: mdl-30075571

ABSTRACT

Excision repair cross-complementing group 1 (ERCC1) functions as a nucleotide excision repair (NER) enzyme. Altered ERCC1 expression or function is closely associated with cancer development and progression. This study determined the association of ERCC1 expression with survivin expression, clinicopathological characteristics, and survival of esophageal squamous cell carcinoma (ESCC) patients after postoperative concurrent chemoradiotherapy.Tissue specimens from 102 resected ESCC patients were acquired for immunohistochemical analysis of ERCC1 and survivin protein expression.ERCC1 expression was detected in 62.7% of ESCC tissues and in 9.8% of normal squamous epithelium tissues (P < .01), while survivin expression was detected in 60.8% of ESCC tissues and in 19.6% of normal squamous epithelia (P < .01). ERCC1 overexpression associated with advanced tumor clinical stage and lymph node metastasis (P < .05), but not with tumor size, depth of invasion, or differentiation (P > .05). ERCC1 overexpression was also associated with survivin levels (r = 0.42, P < .01) and worse progression-free survival of ESCC patients after concurrent chemoradiotherapy. Multivariate analysis data revealed that ERCC1 and survivin protein expression were independent predictors of overall survival of ESCC patients after chemotherapy and/or radiotherapy (P < .05).ERCC1 overexpression is an important phenotype that is associated with ESCC lymph node metastasis and advanced tumor clinical stages. ERCC1 expression may also inhibit ESCC cell apoptosis via regulating survivin expression, and ERCC1 and survivin overexpression are independent predictors of prognosis for ESCC patients who receive chemotherapy and/or radiotherapy.


Subject(s)
Carcinoma, Squamous Cell/pathology , DNA-Binding Proteins/biosynthesis , Endonucleases/biosynthesis , Esophageal Neoplasms/pathology , Inhibitor of Apoptosis Proteins/biosynthesis , Adult , Aged , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/therapy , Chemoradiotherapy , Disease-Free Survival , Esophageal Neoplasms/genetics , Esophageal Neoplasms/therapy , Esophageal Squamous Cell Carcinoma , Female , Gene Expression , Humans , Lymphatic Metastasis , Male , Middle Aged , Neoplasm Invasiveness , Neoplasm Staging , Prognosis , Survivin
9.
Toxicol Lett ; 296: 163-172, 2018 Oct 15.
Article in English | MEDLINE | ID: mdl-29908847

ABSTRACT

Roundup® is extensively used for weed control worldwide. Residues of this compound may lead to side effects of the male reproductive system. However, the toxic effects and mechanisms of Roundup® of male germ cells remain unclear. We aimed to investigate the apoptosis-inducing effects of Roundup® on mouse male germ cells and explore the role of a novel tumor suppressor XAF1 (X-linked inhibitor of apoptosis-associated factor 1) involved in this process. We demonstrated that Roundup® can impair spermatogenesis, decrease sperm motility and concentration, and increase the sperm deformity rate in mice. In addition, excessive apoptosis of germ cells accompanied by the overexpression of XAF1 occurred after Roundup® exposure both in vitro and in vivo. Furthermore, the low expression of XIAP (X-linked inhibitor of apoptosis) induced by Roundup® was inversely correlated with XAF1. Moreover, the knockdown of XAF1 attenuated germ cell apoptosis, improved XIAP expression and inhibited the activation of its downstream target proteins, caspase-3 and PARP, after Roundup® exposure. Taken together, our data indicated that XAF1 plays an important role in Roundup®-induced male germ cell apoptosis. The present study suggested that Roundup® exposure has potential negative implications on male reproductive health in mammals.


Subject(s)
Apoptosis/drug effects , F-Box Proteins/biosynthesis , Germ Cells/drug effects , Glycine/analogs & derivatives , Herbicides/toxicity , Adaptor Proteins, Signal Transducing , Animals , Apoptosis Regulatory Proteins , Caspase 3/drug effects , Cell Survival/drug effects , Glycine/toxicity , Inhibitor of Apoptosis Proteins/biosynthesis , Male , Mice , Organ Size/drug effects , Poly (ADP-Ribose) Polymerase-1/drug effects , Sperm Count , Sperm Motility/drug effects , Spermatogenesis/drug effects , Spermatozoa/drug effects , Spermatozoa/ultrastructure , Glyphosate
10.
Mol Med Rep ; 18(2): 1939-1946, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29956779

ABSTRACT

Simvastatin, an inhibitor of 3­hydroxy­3-methylglutaryl­coenzyme A reductase, is been used in the clinic due to its pleiotropic effects, such as breast cancer, prostate cancer, pancreatic cancer. Simvastatin has recently been demonstrated to serve a potential role in the prophylaxis and therapeutics of a number of human cancers. The majority of reports concerning simvastatin treatment in the majority of human cancers have demonstrated that survivin is significantly decreased as a result and has been implicated in tumorigenesis. However, only a limited number of studies have investigated the use of simvastatin for the treatment of salivary gland adenoid cystic carcinoma (SACC). Therefore, this agent is a candidate for further investigation. The aim of the present study was to investigate the effects of simvastatin on the proliferation, invasion and apoptosis of the human salivary adenoid cystic carcinoma cell line, SACC­83, as well as survivin expression in the cells. The Cell Counting kit­8 assay results revealed that simvastatin inhibited the proliferation of SACC­83 cells in a dose­dependent (10 to 50 µM) and time­dependent (24 to 48 h) manner when compared with the untreated cells. Flow cytometry analysis indicated that simvastatin increased the percentage of cells in early and late apoptosis. Invasion assays revealed that simvastatin treatment inhibited the invasiveness of SACC­83 cells in a dose­dependent manner. In addition, simvastatin downregulated survivin expression in SACC­83 cells. In conclusion, simvastatin significantly inhibited the proliferation and invasion of SACC­83 cells, induced apoptosis, and reduced the expression of survivin, which suggests that simvastatin may be a novel target for SACC therapy.


Subject(s)
Carcinoma, Adenoid Cystic/metabolism , Down-Regulation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Inhibitor of Apoptosis Proteins/biosynthesis , Neoplasm Proteins/biosynthesis , Salivary Gland Neoplasms/metabolism , Simvastatin/pharmacology , Apoptosis/drug effects , Carcinoma, Adenoid Cystic/drug therapy , Carcinoma, Adenoid Cystic/pathology , Cell Line, Tumor , Humans , Neoplasm Invasiveness , Salivary Gland Neoplasms/drug therapy , Salivary Gland Neoplasms/pathology , Survivin
11.
Mol Med Rep ; 18(1): 841-847, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29767242

ABSTRACT

The aim of the present study was to investigate the influence of liposome­plasmid encoding mutant survivin­T34A (PST34A) on tumor growth in cervical cancer in vivo. Liposome­plasmid DNA encoding mutant survivin­T34A was constructed and administered via an intraperitoneal injection in mice inoculated with cervical cancer cells. Following the establishment of the tumor model, the animals were randomly divided into four groups: i) The normal saline group (NS; 100 µl sterile saline once/3 days for 15 days); ii) the 1,2­dioleoyl­3­trimethylammonium­propane (DOTAP) control (100 µg DOTAP once/3 days for 15 days); iii) the plasmid PST34A (10 µg PST34A once/3 days for 15 days); and iv) the PST34A+DOTAP (10 µg PST34A+100 µg DOTAP once/3 days for 15 days). All treatments were administered via intraperitoneal injections. Tumor growth was evaluated following injection with liposome­plasmid DNA encoding mutant survivin­T34A. Apoptosis of cells in ascitic fluid was detected by flow cytometry. The expression of Ki67 and CD34 was detected by immunohistochemical staining. Administration of liposome­plasmid complexes encoding mutant survivin­T34A inhibited tumor growth, reduced the number of tumor nodules and the volume of ascitic fluid, and decreased abdomen circumference and tumor weight. The number of Ki67­positive cells was markedly reduced in the DOTAP+PST34A group compared with the remaining groups. Flow cytometry demonstrated that the number of cells in the sub­G1 phase (apoptosis) increased in the DOTAP+PST34A group compared with all other groups. In addition, tumors in the DOTAP+PST34A group exhibited lower microvessel density compared with all other groups. In the present study, liposome­plasmid DNA encoding mutant survivin­T34A could inhibit tumor growth of cervical cancer. This inhibition may be associated with an increase in the apoptosis rate of tumor cells and a reduction in angiogenesis.


Subject(s)
Apoptosis , Inhibitor of Apoptosis Proteins , Mutation, Missense , Plasmids , Transfection/methods , Uterine Cervical Neoplasms/therapy , Female , HeLa Cells , Humans , Inhibitor of Apoptosis Proteins/biosynthesis , Inhibitor of Apoptosis Proteins/genetics , Liposomes , Plasmids/genetics , Plasmids/pharmacology , Survivin , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/metabolism , Uterine Cervical Neoplasms/pathology
12.
Cancer Biomark ; 22(1): 143-149, 2018.
Article in English | MEDLINE | ID: mdl-29562501

ABSTRACT

BACKGROUND: The potential prognostic value of survivin is variably reported depending on the gastric cancer. OBJECTIVE: Evaluation of the prognostic and predictive significance of serum survivin and its relation with survival and treatment response rates in patients with locally advanced gastric cancer (LAGC). METHODS: Serum samples were prospectively collected from 50 patients with newly diagnosed LAGC. Serum samples of 32 healthy subjects were also collected as control groups for survivin levels. Serum survivin levels were evaluated at baseline and after three cycles of neoadjuvant chemotherapy in LAGC patients. RESULTS: Median survivin level was 147 IU/L (range = 4.4-4936) at baseline and was 27 IU/L (range = 4.2-4737) after neoadjuvant chemotherapy. The difference between survivin levels of the control group (26 IU/L, range = 3.8-1430) and pre-treatment patient group was statistically significant (p< 0.001). Clinical response to mDCF regimen was classified as progressive (progressive disease) and non-progressive groups (partial response + stable disease). Baseline survivin levels were similar between patients in progressive and non-progressive groups (p= 0.55). Survivin levels were significantly reduced after chemotherapy in non-progressive group (p< 0.001). In contrast, serum survivin levels increased in a stepwise fashion from baseline to post-chemotherapy in patients with progressive disease (p= 0.06). Patients were divided into low and high survivin groups according to baseline median survivin levels. Median DFS was 12.4 and 14.6 months for low and high groups, respectively (p= 0.18). Moreover, median OS was 14.4 and 24.9 months for low and high group, respectively (p= 0.14). CONCLUSION: It can be suggested that serum survivin can be used as a predictor of response to chemotherapy- but not survival- in LAGC patients receiving neoadjuvant mDCF chemotherapy. However, large multicenter prospective studies are required to confirm these results.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biomarkers, Tumor/blood , Inhibitor of Apoptosis Proteins/blood , Stomach Neoplasms/blood , Stomach Neoplasms/drug therapy , Adult , Aged , Biomarkers, Tumor/biosynthesis , Chemotherapy, Adjuvant , Cisplatin/administration & dosage , Docetaxel , Female , Fluorouracil/administration & dosage , Humans , Inhibitor of Apoptosis Proteins/biosynthesis , Male , Middle Aged , Neoadjuvant Therapy , Stomach Neoplasms/surgery , Survivin , Taxoids/administration & dosage
13.
J Oral Pathol Med ; 47(1): 48-52, 2018 Jan.
Article in English | MEDLINE | ID: mdl-28833597

ABSTRACT

BACKGROUND: Survivin, a member of the inhibitor of apoptosis family, is overexpressed in most human tumors, but undetectable in normal adult tissues. It is a promising target molecule in cancer treatment, as interference in its function promotes apoptosis. Artepillin C, a major, biologically active ingredient of Brazilian propolis, possesses anticancer activity against several cancer cells with different tissue origins. However, little is known about its bioactivity on oral squamous cell carcinoma cells or its effect on survivin expression. The aim of this study was to investigate the cytotoxic and antisurvivin activities of artepillin C in oral squamous cell carcinoma cells. METHODS: HSC-3 human oral squamous cell carcinoma cells were treated with varying doses of artepillin C for up to 72 hours. Cell viability was measured by WST-1, and the cytotoxic effects of artepillin C on HSC-3 cells were quantified with flow cytometry. The survivin levels were determined by ELISA. RESULTS: Artepillin C exhibited dose- and time-dependent cytotoxic effects on HSC-3 cells. Flow cytometric analysis showed that 22% of untreated HSC-3 cells underwent spontaneous cell death, whereas 77.32% of the cells were killed in response to the highest dose of artepillin C at 72 hours. Survivin expression was reduced in treated cells. CONCLUSIONS: HSC-3 cells are vulnerable to artepillin C in a dose- and time-dependent manner. HSC-3 cell death induced by artepillin C, at least in part, was a result of a decrease in survivin levels.


Subject(s)
Carcinoma, Squamous Cell/drug therapy , Inhibitor of Apoptosis Proteins/drug effects , Mouth Neoplasms/drug therapy , Phenylpropionates/pharmacology , Apoptosis/drug effects , Brazil , Carcinoma, Squamous Cell/pathology , Cell Death/drug effects , Cell Line, Tumor/drug effects , Cell Survival/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Inhibitor of Apoptosis Proteins/biosynthesis , Mouth Neoplasms/pathology , Phenylpropionates/administration & dosage , Propolis/pharmacology , Survivin , Time Factors
14.
Int J Cancer ; 142(10): 2040-2055, 2018 05 15.
Article in English | MEDLINE | ID: mdl-29250796

ABSTRACT

Our recent studies demonstrate that X-linked inhibitor of apoptosis protein (XIAP) is essential for regulating colorectal cancer invasion. Here, we discovered that RhoGDIß was a key XIAP downstream effector mediating bladder cancer (BC) invasion in vitro and in vivo. We found that both XIAP and RhoGDIß expressions were consistently elevated in BCs of N-butyl-N-(4-hydroxybutyl)-nitrosamine (BBN)-treated mice in comparison to bladder tissues from vehicle-treated mice and human BCs in comparison to the paired adjacent normal bladder tissues. Knockdown of XIAP attenuated RhoGDIß expression and reduced cancer cell invasion, whereas RhoGDIß expression was attenuated in BBN-treated urothelium of RING-deletion knockin mice. Mechanistically, XIAP stabilized RhoGDIß mRNA by its positively regulating nucleolin mRNA stability via Erks-dependent manner. Moreover, ectopic expression of GFP-RhoGDIß in T24T(shXIAP) cells restored its lung metastasis in nude mice. Our results demonstrate that XIAP-regulated Erks/nucleolin/RhoGDIß axis promoted BC invasion and lung metastasis.


Subject(s)
Inhibitor of Apoptosis Proteins/biosynthesis , Lung Neoplasms/secondary , Phosphoproteins/metabolism , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , Urinary Bladder Neoplasms/pathology , X-Linked Inhibitor of Apoptosis Protein/biosynthesis , rho Guanine Nucleotide Dissociation Inhibitor beta/genetics , Animals , Cell Line, Tumor , Female , HCT116 Cells , Humans , Inhibitor of Apoptosis Proteins/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , MAP Kinase Signaling System , Mice , Mice, Inbred C57BL , Mice, Nude , Neoplasm Invasiveness , RNA, Messenger/genetics , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/metabolism , X-Linked Inhibitor of Apoptosis Protein/metabolism , rho Guanine Nucleotide Dissociation Inhibitor beta/metabolism , Nucleolin
15.
Biomed Pharmacother ; 97: 359-368, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29091885

ABSTRACT

Triple negative breast cancer is a clinically challenging subtype due to lack of biomarker for rational targeted therapy. Lipid rafts are cholesterol enriched rigid platforms, which colocalize signalling molecules of cancer progression. This study explores the effect of lipid rafts disruption by cholesterol depleting agent, MßCD on induction of apoptosis and expression of WNT receptor LRP6, survivin and common apoptotic markers in TNBC cell lines. The in vitro effect of lipid rafts disruption on viability, single cell reproductive ability, proliferation and migration were evaluated by MTT, clonogenic, BrdU incorporation and wound scratch assays, respectively. The morphological changes were assessed by tryphan blue, Wright and Giemsa staining; nuclear changes by Hoechst staining. The induction of apoptosis was evaluated by AO/EtBr staining, DNA damage and DNA fragmentation assays. Expression of Caveolin-1, LRP6, ß-Catenin, Survivin, Bcl2, BAX, Caspase-3, Ki67 and c-myc were analyzed by PCR and Western blotting techniques. The lipid raft disruption resulted in reduction of the proliferation of MDA-MB 231 and MDA-MB 468 cells by 56.3 and 42.0%; survival fraction by 54.7 and 59.4%; migration by 44.3 and 48.4%, respectively. It also induced apoptosis by causing cell shrinkage, membrane blebbing, nuclear condensation, chromatin cleavage, oligonucleotide fragmentation with an apoptotic index of 59.1 and 46.6% in MDA-MB 231 and 468 cells, respectively. Further, it downregulated the expression of caveolin-1, LRP6, ß-catenin, survivin, Bcl2, ki67, c-myc and upregulated BAX, caspase-3. The cholesterol supplementation enhanced the clonogenic potential and upregulated the expression of caveolin-1 and LRP6. The results underline a potential effect of lipid rafts disruption on induction of apoptosis in TNBC cells.


Subject(s)
Apoptosis/physiology , Gene Expression Regulation, Neoplastic , Inhibitor of Apoptosis Proteins/biosynthesis , Low Density Lipoprotein Receptor-Related Protein-6/biosynthesis , Membrane Microdomains/metabolism , Triple Negative Breast Neoplasms/metabolism , Cell Line, Tumor , Female , Humans , Inhibitor of Apoptosis Proteins/genetics , Low Density Lipoprotein Receptor-Related Protein-6/genetics , Membrane Microdomains/genetics , Survivin , Triple Negative Breast Neoplasms/genetics
16.
Stem Cells ; 36(1): 123-129, 2018 01.
Article in English | MEDLINE | ID: mdl-29067757

ABSTRACT

Although mesenchymal stromal cells (MSCs) have significant potential in cell-based therapies, little is known about the factors that regulate their functions. While exploring regulatory molecules potentially involved in MSC activities, we found that the endogenous multifunctional factor Survivin is essential for MSC survival, expansion, lineage commitment, and migration. Pharmacological or genetic blockade of Survivin expression in mouse and human bone marrow MSC enhances caspase 3 and 7 expression and reduces proliferation resulting in fewer MSC and clonogenic colony-forming unit-fibroblasts (CFU-F), whereas ectopic Survivin overexpression in MSC results in their expansion. Survivin is also required for the MSC proliferative responses to basic fibroblast growth factor and platelet derived growth factor. In a wound healing model, Survivin inhibition results in suppression of MSC migration to the wound site. In addition, loss of Survivin in MSCs compromises their hematopoiesis-supporting capacity. These results demonstrate that Survivin is a key regulator of mouse and human MSC function, and suggest that targeted modulation of Survivin in MSCs may have clinical utility to enhance MSC recovery and activity following insult or stress. Stem Cells 2018;36:123-129.


Subject(s)
Inhibitor of Apoptosis Proteins/biosynthesis , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Repressor Proteins/biosynthesis , Animals , Cell Differentiation/physiology , Cell Movement/physiology , Cell Proliferation/physiology , Humans , Inhibitor of Apoptosis Proteins/antagonists & inhibitors , Inhibitor of Apoptosis Proteins/metabolism , Mice , Mice, Inbred C57BL , Repressor Proteins/antagonists & inhibitors , Repressor Proteins/metabolism , Survivin
17.
Oncol Rep ; 38(4): 2062-2068, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28849066

ABSTRACT

The current treatment recommendation for T2-3N0M0 glottic squamous cell carcinoma (SCC) in the Nordic countries comprises of radiotherapy (RT) and chemoradiotherapy (CRT). Tumor radiosensitivity varies and another option is primary surgical treatment, which underlines the need for predictive markers in this patient population. The aim of the present study was to investigate the relation of the proteins WRAP53ß, survivin and p16INK4a to RT/CRT response and ultimate outcome of patients with T2-T3N0 glottic SCC. Protein expression was determined using immunohistochemistry on tumors from 149 patients consecutively treated with RT or CRT at Helsinki University Hospital, Karolinska University Hospital, and Linköping University Hospital during 1999-2010. Our results demonstrate a significantly better 5-year relapse-free survival, disease-free survival (DFS), disease-specific survival and overall survival of patients with T3N0 tumors treated with CRT compared with RT alone. Patients with tumors showing a cytoplasmic staining of WRAP53ß revealed significantly worse DFS compared with those with nuclear staining. For survivin, we observed a trend towards better 5-year DFS in patients with strong nuclear survivin expression compared with those with weak nuclear survivin expression (p=0.091). Eleven (7%) tumors showed p16 positivity, with predilection to younger patients, and this age group of patients with p16-positive SCC had a significantly better DFS compared with patients with p16-negative SCC. Taken together, our results highlight WRAP53ß as a potential biomarker for predicting RT/CRT response in T2-T3N0 glottic SCC. p16 may identify a small but distinct group of glottic SCC with favorable outcome. Furthermore, for T3N0 patients better outcome was observed following CRT compared to RT alone.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/therapy , Cisplatin/therapeutic use , Cyclin-Dependent Kinase Inhibitor p16/biosynthesis , Glottis/pathology , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/therapy , Inhibitor of Apoptosis Proteins/biosynthesis , Laryngeal Neoplasms/metabolism , Laryngeal Neoplasms/therapy , Telomerase/biosynthesis , Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/biosynthesis , Carcinoma, Squamous Cell/pathology , Chemoradiotherapy , Female , Glottis/drug effects , Glottis/radiation effects , Head and Neck Neoplasms/pathology , Humans , Immunohistochemistry , Laryngeal Neoplasms/pathology , Male , Middle Aged , Molecular Chaperones , Neoplasm Staging , Predictive Value of Tests , Squamous Cell Carcinoma of Head and Neck , Survivin
18.
Oncol Rep ; 38(4): 2096-2104, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28765921

ABSTRACT

Livin, a member of the inhibitor of apoptosis protein (IAP) family, is expressed at a high level in lung adenocarcinoma and influences the progression of cancer, and its response to chemotherapy and radiotherapy. Aberrant microRNA (miRNA) expression has also been associated with cancer initiation and development. However, the clinical significance of Livin and its relationship with miRNAs in lung adenocarcinoma are still unclear. In the present study, the expression level of Livin in 90 pairs of lung adenocarcinoma and their adjacent tissues were detected by immunohistochemistry staining. Spearman correlation and Kaplan-Meier, univariate and multivariate analyses were applied to evaluate the correlation between the expression of Livin and clinical characteristics. With the integration of bioinformatics analysis and dual-luciferase reporter gene assays, we identified the miRNA that can target Livin mRNA. The functional effects of miRNA-mediated Livin knockdown were assessed by Cell Counting Kit-8 (CCK-8) and apoptosis assays, and cell cycle analysis. The present study revealed that Livin was upregulated in lung adenocarcinoma tissues and may be associated with the poor prognosis in lung adenocarcinoma patients. The overexpression of Livin is partly caused by the downregulation of miR-198. Further exploration revealed that miRNA-198-mediated silencing of Livin significantly inhibited cell growth and enhanced apoptosis of A549 cells, accompanied by marked upregulation of caspase-3. Finally, we observed that the miR-198 overexpression and Livin neutralization had similar effects on improving cisplatin chemosensitivity in A549 cells. Overall, these findings suggest that Livin has the potential to become a biomarker for predicting the prognosis of lung adenocarcinoma and may provide a promising strategy for assisting chemotherapy of lung adenocarcinoma through the miR-198/Livin/caspase-3 regulatory network.


Subject(s)
Adaptor Proteins, Signal Transducing/biosynthesis , Adenocarcinoma/metabolism , Inhibitor of Apoptosis Proteins/biosynthesis , Lung Neoplasms/metabolism , MicroRNAs/metabolism , Neoplasm Proteins/biosynthesis , A549 Cells , Adaptor Proteins, Signal Transducing/genetics , Adenocarcinoma/drug therapy , Adenocarcinoma/genetics , Adenocarcinoma of Lung , Antineoplastic Agents/pharmacology , Cisplatin/pharmacology , Humans , Inhibitor of Apoptosis Proteins/genetics , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , MicroRNAs/genetics , Neoplasm Proteins/genetics , Prognosis , Up-Regulation
19.
Arch Biochem Biophys ; 636: 110-122, 2017 12 15.
Article in English | MEDLINE | ID: mdl-28803837

ABSTRACT

Cancer cells require increased aerobic glycolysis to support rapid cell proliferation. For their increased energy demands, cancer cells express glucose transporter (Glut) proteins at a high level. Glut1 is associated with basal-like breast cancer and is considered a potential therapeutic target. To investigate the possibility of Glut1 as a therapeutic target in breast cancer cells, we downregulated Glut1 in triple-negative breast cancer (TNBC) cell lines using a short hairpin system. We determined whether Glut1 silencing might enhance anti-proliferative effect of chemotherapeutic agents. Contrary to our hypothesis, ablation of Glut1 attenuated apoptosis and increased drug resistance via upregulation of p-Akt/p-GSK-3ß (Ser9)/ß-catenin/survivin. These results indicated that the potential of Glut1 as a therapeutic target should be carefully reevaluated.


Subject(s)
Drug Resistance, Neoplasm , Gene Expression Regulation, Neoplastic , Gene Silencing , Glucose Transporter Type 1/biosynthesis , Glycogen Synthase Kinase 3 beta/biosynthesis , Inhibitor of Apoptosis Proteins/biosynthesis , Proto-Oncogene Proteins c-akt/biosynthesis , Signal Transduction , Up-Regulation , beta Catenin/biosynthesis , Cell Line, Tumor , Female , Glucose Transporter Type 1/genetics , Glycogen Synthase Kinase 3 beta/genetics , Humans , Inhibitor of Apoptosis Proteins/genetics , Proto-Oncogene Proteins c-akt/genetics , Survivin , beta Catenin/genetics
20.
Anticancer Res ; 37(8): 4127-4137, 2017 08.
Article in English | MEDLINE | ID: mdl-28739697

ABSTRACT

BACKGROUND/AIM: The aim of the study was to evaluate the anti-tumor mechanism of Z-360, a gastrin/cholecystokinin-2 receptor (CCK2R) antagonist, in MIA PaCa-2 cells and in a subcutaneous xenograft mice model. MATERIALS AND METHODS: The anti-tumor effects of Z-360 and/or gemcitabine were monitored using a MIA PaCa-2 xenograft model. The effect of Z-360 on apoptosis in the model was examined by TUNEL staining and real-time PCR analysis and the effect in MIA PaCa-2 cells stably expressing human CCK2R was also evaluated by caspase-3/7 activity. RESULTS: In this xenograft model, Z-360 significantly reduced the tumor weight, increased TUNEL-positive cells and suppressed the expression of anti-apoptosis factors such as survivin, XIAP and Mcl-1, and these effects of Z-360 combined with gemcitabine were more effective. Furthermore, gastrin-17 and gastrin-34 inhibited apoptosis in vitro and Z-360 dose-dependently abrogated this effect. CONCLUSION: These results suggest that Z-360 exerts an anti-tumor effect through a reduction in anti-apoptosis factors by blocking CCK2R.


Subject(s)
Apoptosis/drug effects , Benzodiazepinones/administration & dosage , Pancreatic Neoplasms/drug therapy , Receptor, Cholecystokinin B/genetics , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Deoxycytidine/administration & dosage , Deoxycytidine/analogs & derivatives , Endopeptidases/administration & dosage , Gastrins/administration & dosage , Gene Expression Regulation, Neoplastic/drug effects , Humans , Inhibitor of Apoptosis Proteins/biosynthesis , Mice , Myeloid Cell Leukemia Sequence 1 Protein/biosynthesis , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Receptor, Cholecystokinin B/antagonists & inhibitors , Receptor, Cholecystokinin B/biosynthesis , Survivin , X-Linked Inhibitor of Apoptosis Protein/biosynthesis , Xenograft Model Antitumor Assays , Gemcitabine
SELECTION OF CITATIONS
SEARCH DETAIL
...