Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Acta Histochem ; 125(1): 151975, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36455340

ABSTRACT

BACKGROUND: The motor ability of cancer cells to cross the basement membrane contributes to their implantation in a new location. Metastasis is a significant factor that worsens the prognosis of cancer patients. Thus, reducing cell invasiveness is an important aspect of anticancer therapy, also in bladder cancer treatment. MATERIAL: The study material was the T24 cell line of human urinary bladder cancer. The migratory potential of the cells and the effect of the treatment with individually doses and synergistic combination of doxorubicin and metformin in the 500:1 ratio for 24 h were analyzed. RESULTS: The results obtained show a compound-initiated decrease in the motor abilities of bladder cancer cells compared to controls. A decrease in the rate of colony formation was observed, as well as inhibition of migration through inserts. The visualized reorganization of the vimentin and actin networks confirms the drug-initiated limitation of the metastatic potential of T24 cells. CONCLUSION: According to our knowledge, we are the first to show, that combination of doxorubicin and metformin also worth considering in the treatment of bladder cancer. We showed that simultaneous administration of these cytostatic enhances the antiproliferative effect of drugs, but also limits cells' migratory potential.


Subject(s)
Metformin , Urinary Bladder Neoplasms , Humans , Metformin/pharmacology , Doxorubicin/pharmacology , Urinary Bladder Neoplasms/drug therapy , Cell Line, Tumor , Cell Movement , Cell Proliferation
2.
Int J Mol Sci ; 22(23)2021 Nov 26.
Article in English | MEDLINE | ID: mdl-34884588

ABSTRACT

Metastasis is one of the most urgent issues in breast cancer patients. One of the factors necessary in the migration process is the remodeling of the extracellular matrix (ECM). Metalloproteinases (MMPs) can break down the elements of the ECM, which facilitates cell movement. Many highly aggressive tumors are characterized by high levels of MMPs. In the case of breast cancer, the association between MMP-9 and the migration potential and invasiveness of cells has been demonstrated. In addition, reports indicating increased migration of breast cancer cells after the administration of the commonly used cytostatic cyclophosphamide (CP) are particularly disturbing. Hence, our research aimed to assess the effect of CP treatment on MDA-MB-231 and MCF-7 cells and how this response is influenced by the downregulation of the MMP-9 level. The obtained results suggest that CP causes a decrease in the survival of breast cancer cells of various invasiveness, and the downregulation of MMP-9 enhances this effect, mainly by inducing apoptosis. Moreover, in the group of MMP-9 siRNA-transfected CP-treated cells, a more severe reduction in invasion and migration of cells of both lines was observed, as indicated by the migration and invasion transwell assays and Wound healing assay. Hence, we suggest that CP alone may not result in satisfactory therapeutic effects. On the other hand, the use of combination therapy targeting MMP-9, together with the CP, could improve the effectiveness of the treatment. Additionally, we confirmed a relationship between the levels of MMP-9 and cytokeratin 19 (CK19).


Subject(s)
Breast Neoplasms/pathology , Cell Movement , Cyclophosphamide/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Matrix Metalloproteinase 9/chemistry , Antineoplastic Agents, Alkylating/pharmacology , Apoptosis , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Cycle , Cell Proliferation , Female , Humans , Keratin-19/genetics , Keratin-19/metabolism , Prognosis , Tumor Cells, Cultured
3.
Cancer Manag Res ; 13: 91-106, 2021.
Article in English | MEDLINE | ID: mdl-33447082

ABSTRACT

PURPOSE: Metastasis remains a serious clinical problem in which epithelial-to-mesenchymal transition is strictly involved. The change of cell phenotype is closely related to the dynamics of the cytoskeleton. Regarding the great interest in microfilaments, the manipulation of ABPs (actin-binding proteins) appears to be an interesting treatment strategy. MATERIAL: The research material was the highly aggressive A549 cells with FHOD1 (F FH1/FH2 domain-containing protein 1) downregulation. The metastatic potential of the cells and the sensitivity to treatment with alkaloids (piperlongumine, sanguinarine) were analyzed. RESULTS: In comparison to A549 cells with naïve expression of FHOD1, those after manipulation were characterized by a reduced migratory potential. The obtained results were associated with microfilaments and vimentin reorganization induced by the manipulation of FHOD1 together with alkaloids treatment. The result was also an increase in the percentage of late apoptotic cells. CONCLUSION: Downregulation of FHOD1 induced reorganization of microfilament network followed by the reduction in the metastatic potential of the A549 cells, as well as their sensitization to selected compounds. The presented results and the analysis of clinical data indicate the possibility of transferring research from the basic level to in vivo models in the context of manipulation of ABPs as a new therapeutic target in oncology.

4.
Acta Histochem ; 122(7): 151625, 2020 Oct.
Article in English | MEDLINE | ID: mdl-33066845

ABSTRACT

The aim of this investigation was to determine the effect of doxorubicin on F-actin rearrangement and ß-catenin and cofilin-1 in a rat glioma C6 cell line in combination with changes in their morphology and ultrastructure. The experimental material constituted rat glioma C6 cell line. The cells were incubated with sublethal doses of doxorubicin in the concentration of 50, 100 and 200 nM. The blue trypan dye method was used to determine the number of dead cells. Morphological and ultrastructural changes in the cells were evaluated using light and transmission electron microscope, respectively. In order to determine the rearrangements and level of expression of F-actin, ß-catenin and cofilin-1 they were analyzed using a fluorecence microscope. In turn, cell death and cell cycle were evaluated by Guava 6HT-2 L Cytometer. The performed experiments showed a dose-dependent decrease in the survival of C6 cells after treatment with doxorubicin. The analysis of cell death showed a dose-dependent increase in the population of apoptotic and necrotic cells. These results were confirmed by microscopy observation. The changes in morphology, ultrastructure, and rearrangements of F-actin, ß-catenin and cofilin-1 were also observed. The results obtained in the study showed that sublethal concentrations of doxorubicin influenced the structure of F-actin and other proteins involved in cell-cell interactions. Moreover, mitotic catastrophe may preceding apoptosis, what suggest the cytotoxic effect of low dose of doxorubicin. Furthermore, our results confirmed the multi-dimensional mechanism of DOX action in tumor cells.


Subject(s)
Cell Cycle/drug effects , Cell Death/drug effects , Doxorubicin/pharmacology , Glioma/drug therapy , Actins/metabolism , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Glioma/ultrastructure
5.
Cells ; 9(10)2020 10 06.
Article in English | MEDLINE | ID: mdl-33036298

ABSTRACT

The actin cytoskeleton plays a crucial role in many cellular processes while its reorganization is important in maintaining cell homeostasis. However, in the case of cancer cells, actin and ABPs (actin-binding proteins) are involved in all stages of carcinogenesis. Literature has reported that ABPs such as SATB1 (special AT-rich binding protein 1), WASP (Wiskott-Aldrich syndrome protein), nesprin, and villin take part in the initial step of carcinogenesis by regulating oncogene expression. Additionally, changes in actin localization promote cell proliferation by inhibiting apoptosis (SATB1). In turn, migration and invasion of cancer cells are based on the formation of actin-rich protrusions (Arp2/3 complex, filamin A, fascin, α-actinin, and cofilin). Importantly, more and more scientists suggest that microfilaments together with the associated proteins mediate tumor vascularization. Hence, the presented article aims to summarize literature reports in the context of the potential role of actin and ABPs in all steps of carcinogenesis.


Subject(s)
Actins/metabolism , Carcinogenesis/metabolism , Microfilament Proteins/metabolism , Cell Movement , Humans
6.
Molecules ; 25(13)2020 Jul 03.
Article in English | MEDLINE | ID: mdl-32635287

ABSTRACT

BACKGROUND: Cancers are one of the leading causes of deaths nowadays. The development of new treatment schemes for oncological diseases is an interesting direction in experimental medicine. Therefore, the evaluation of the influence of two alkaloids-piperlongumine (PL), sanguinarine (SAN) and their combination-on the basic life processes of the A549 cell line was considered reasonable. METHODS: The aim was achieved by analyzing the cytotoxic effects of PL and SAN and their combination in the ratio of 4:1 on the induction of cell death, changes in the distribution of cell cycle phases, reorganization of cytoskeleton and metastatic potential of A549 cells. The versatility of the applied concentration ratio was evaluated in terms of other cancer cell lines: MCF-7, H1299 and HepG2. RESULTS: The results obtained from the MTT assay indicated that the interaction between the alkaloids depends on the concentration and type of cells. Additionally, the compounds and their combination did not exhibit a cytotoxic effect against normal cells. The combined effects of PL and SAN increased apoptosis and favored metastasis inhibition. CONCLUSION: Selected alkaloids exhibit a cytotoxic effect on A549 cells. In turn, treatment with the combination of PL and SAN in a 4:1 ratio indicates a synergistic effect and is associated with an increase in the level of reactive oxygen species (ROS).


Subject(s)
Benzophenanthridines/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Dioxolanes/pharmacology , Drug Synergism , Isoquinolines/pharmacology , Lung Neoplasms/drug therapy , Anti-Infective Agents/pharmacology , Apoptosis , Carcinoma, Non-Small-Cell Lung/pathology , Cell Cycle , Cell Movement , Cell Proliferation , Humans , Lung Neoplasms/pathology , Neoplasm Invasiveness , Reactive Oxygen Species/metabolism , Tumor Cells, Cultured
7.
Oncol Rep ; 43(3): 765-772, 2020 03.
Article in English | MEDLINE | ID: mdl-32020229

ABSTRACT

Cyclin F is a non­canonical cyclin which is a part of the SKP1­CUL1­F­box protein (SCF) E3 ubiquitin­protein ligase complex. Cyclin F is responsible for target recognition, ubiquitination, and degradation of various molecular targets. This protein also controls genome stability through the degradation of ribonucleotide reductase subunit M2 (RRM2). In the present study, the difference between cyclin F expression in cell lines derived from primary and metastatic melanoma, A375 and RPMI­7951, respectively, were investigated using a western blot analysis and flow cytometry assays. A decrease in cyclin F expression in the A375 cells and an increase in RPMI­7951 cells after cisplatin treatment were observed. These changes may be related to a mutation in p53 in the RPMI­7951 cell line. Flow cytometry was conducted to observe that the RPMI­7951 cell line exhibited greater susceptibility to cisplatin, associated with lack of proper cell cycle control. Therefore, it is possible that cyclin F may modulate drug response in melanoma. The presented data describe cyclin F as a new potential factor that contributes to drug resistance in melanoma patients.


Subject(s)
Cisplatin/pharmacology , Cyclins/genetics , Melanoma/drug therapy , Ribonucleoside Diphosphate Reductase/genetics , Cell Line, Tumor , Cisplatin/adverse effects , Drug Resistance, Neoplasm/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Genomic Instability/drug effects , Humans , Melanoma/genetics , Melanoma/pathology , Neoplasm Metastasis , Proteolysis/drug effects , Ubiquitination/genetics
8.
Int J Oncol ; 55(6): 1261-1274, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31638196

ABSTRACT

The present study aimed to evaluate the cellular and molecular effects of low concentrations of the flavonoid, fisetin, on K562 human chronic myeloid leukemia cells, in the context of both potential anti­proliferative and anti­metastatic effects. Thiazolyl blue tetrazolium bromide assay, Trypan blue exclusion assay, Annexin V/propidium iodide test, cell cycle analysis, Transwell migration and invasion assays, the fluorescence staining of ß­catenin and F­actin as well as reverse transcription­quantitative polymerase chain reaction were performed to achieve the research goal. Furthermore, the nature of the interaction between fisetin and arsenic trioxide in the K562 cells was analyzed according to the Chou­Talalay median­effect method. We found that low concentrations of fisetin had not only a negligible effect on the viability and apoptosis of the K562 cells, but also modulated the mRNA levels of selected metastatic­related markers, accompanied by an increase in the migratory and invasive properties of these cancer cells. Although some markers of cell death were significantly elevated in response to fisetin treatment, these were counterbalanced through anti­apoptotic and pro­survival signals. With decreasing concentrations of fisetin and arsenic trioxide, the antagonistic interactions between the 2 agents increased. On the whole, the findings of this study suggest that careful consideration should be taken when advising cancer patients to take fisetin as a dietary supplement and when considering fisetin as a potential candidate for the treatment of chronic myeloid leukemia. Further more detailed studies are required to confirm our findings.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Arsenic Trioxide/pharmacology , Cell Proliferation/drug effects , Flavonoids/pharmacology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis/drug effects , Apoptosis/genetics , Arsenic Trioxide/therapeutic use , Cell Movement/drug effects , Cell Movement/genetics , Cell Survival/drug effects , Cell Survival/genetics , Dose-Response Relationship, Drug , Drug Antagonism , Drug Screening Assays, Antitumor , Drug Synergism , Flavonoids/therapeutic use , Flavonols , Gene Expression Regulation, Neoplastic/drug effects , Humans , K562 Cells , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
9.
Cancers (Basel) ; 11(8)2019 Aug 20.
Article in English | MEDLINE | ID: mdl-31434275

ABSTRACT

Currently, autophagy in the context of cancer progression arouses a lot of controversy. It is connected with the possibility of switching the nature of this process from cytotoxic to cytoprotective and vice versa depending on the treatment. At the same time, autophagy of cytoprotective character may be one of the factors determining multidrug resistance, as intensification of the process is observed in patients with poorer prognosis. The exact mechanism of this relationship is not yet fully understood; however, it is suggested that one of the elements of the puzzle may be a cytoskeleton. In the latest literature reports, more and more attention is paid to the involvement of actin in the autophagy. The role of this protein is linked to the formation of autophagosomes, which are necessary element of the process. However, based on the proven effectiveness of manipulation of the actin pool, it seems to be an attractive alternative in breaking autophagy-dependent multidrug resistance in cancer.

10.
Acta Histochem ; 121(6): 724-731, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31262517

ABSTRACT

Oxymatrine is the alkaloid derived from the root of Sophora species. This compound is proven to exhibit anti-viral, anti-asthmatic, anti-fibrotic and anti-inflammatory properties. Additionally, oxymatrine is able to promote cancer cells apoptosis and inhibit their proliferation. The aim of this study was to present the influence of oxymatrine on non-small cell lung cancer cells. The results indicate, that this agent induces dose-dependent cell death mainly through ER stress-induced apoptosis pathway. We also suggest that the oxymatrine reduces the metastatic potential by inhibition of the EMT process, as A549 cells treated with chosen doses of the compound were characterized by a decrease in the expression of the N-cadherin, vimentin and the elevation of E-cadherin level. Moreover, the study broadens the knowledge on so far poorly understood aspect of the influence of oxymatrine on the cytoskeleton structure.


Subject(s)
Alkaloids/pharmacology , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Proliferation/drug effects , Endoplasmic Reticulum Stress/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Lung Neoplasms/metabolism , Neoplasm Proteins/biosynthesis , Quinolizines/pharmacology , A549 Cells , Carcinoma, Non-Small-Cell Lung/pathology , Epithelial-Mesenchymal Transition , Humans , Lung Neoplasms/pathology
11.
Int J Oncol ; 54(3): 1099-1111, 2019 03.
Article in English | MEDLINE | ID: mdl-30569147

ABSTRACT

Malignant glioma is the most common type of brain cancer with poor prognosis. Surgical resection, chemotherapy and radiotherapy are the main therapeutic options; however, in addition to their insufficient efficacy, they are associated with the pain experienced by patients. To relieve pain, local anesthetics, such as lidocaine can be used. In the present study, the effects of lidocaine on the C6 rat glioma cell line were investigated. An MTT assay and Annexin V/propidium iodide analysis indicated the increase in the percentage of apoptotic and necrotic cells in response to lidocaine. Furthermore, light microscopy analysis on the ultrastructural level presented the occurrence of vacuole­like structures associated with autophagy, which was supported by the analysis of autophagy markers (microtubule­associated protein 1A/1B­light chain 3, acridine orange and Beclin­1). Additionally, reorganization of the cytoskeleton was observed following treatment with lidocaine, which serves an important role in the course of autophagy. To determine the nature of autophagy, an inhibitor, bafilomycin A1 was applied. This compound suppressed the fusion of autophagosomes with lysosomes and increased the percentage of apoptotic cells. These results demonstrated that lidocaine may induce cytoprotective autophagy and that manipulation of this process could be an alternative therapeutic strategy in the treatment of cancer.


Subject(s)
Autophagy/drug effects , Brain Neoplasms/pathology , Glioma/pathology , Lidocaine/pharmacology , Animals , Beclin-1/genetics , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Cell Cycle/drug effects , Cell Death/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Cytoskeleton/drug effects , Glioma/genetics , Glioma/metabolism , Microtubule-Associated Proteins/genetics , Rats
12.
Onco Targets Ther ; 11: 8275-8292, 2018.
Article in English | MEDLINE | ID: mdl-30538497

ABSTRACT

PURPOSE: The aim of the study was to evaluate the effect of piperlongumine (2 and 4 µM) on endothelial EA.hy926 and lung adenocarcinoma A549 cells with regulated expression of profilin-1 (PFN1). MATERIAL AND METHODS: The cytotoxicity of alkaloid was evaluated by MTT assay, while cell death was assessed using double staining with annexin V and propidium iodide. Subsequently, the level of PFN1 1) upregulation in EA.hy926 endothelial cells and 2) downregulation in A549 lung adenocarcinoma cells. The next step was the analysis of the effect of PFN1 manipulation on cytoskeletal proteins. RESULTS: The results showed that piperlongumine may inhibit proliferation of EA.hy926 and A549 cell lines and also induce cell death in a dose-dependent manner. Furthermore, endothelial cells with PFN1 overexpression showed lower sensitivity to alkaloid and strengthening of cell-cell interactions. In the case of A549 cells, loss of PFN1 expression resulted in a lower percentage of early apoptotic cells, reorganization of F-actin and vimentin network, and reduction of migratory potential. CONCLUSION: We suggest that upregulation of PFN1 in endothelial cell line may stabilize the cell junctions. In turn, PFN1 downregulation in A549 cells probably suppresses cell migration and sensitizes cells to anticancer agents.

13.
Int J Mol Sci ; 19(3)2018 Feb 27.
Article in English | MEDLINE | ID: mdl-29495431

ABSTRACT

The identification and development of new agents with a therapeutic potential as well as novel drug combinations are gaining the attention of scientists and clinicians as a plausible approach to improve therapeutic regimens for chemoresistant tumors. We have recently reported that the flavonoid fisetin (FIS), at physiologically attainable concentrations, acts synergistically with clinically achievable doses of paclitaxel (PTX) to produce growth inhibitory and pro-death effects on A549 human non-small cell lung cancer (NSCLC) cells. To further investigate a potential therapeutic efficacy of the combination of fisetin with paclitaxel, we decided to assess its impact on metastatic capability of A549 cells as well as its toxicity toward normal human lung fibroblast. Cell viability, cell migration, and invasion were measured by thiazolyl blue tetrazolium bromide (MTT) assay, wound healing assay, and Transwell chamber assay, respectively. The expression of metastasis-related genes was assessed with quantitative reverse transcriptase real-time polymerase chain reaction (qRT-PCR). Actin and vimentin filaments were examined under the fluorescence microscope. The combination of FIS and PTX significantly reduced cancer cell migration and invasion, at least partially, through a marked rearrangement of actin and vimentin cytoskeleton and the modulation of metastasis-related genes. Most of these effects of the combination treatment were significantly greater than those of individual agents. Paclitaxel alone was even more toxic to normal cells than the combination of this drug with the flavonoid, suggesting that FIS may provide some protection against PTX-mediated cytotoxicity. The combination of FIS and PTX is expected to have a synergistic anticancer efficacy and a significant potential for the treatment of NSCLC, however, further in vitro and in vivo studies are required to confirm this preliminary evidence.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Non-Small-Cell Lung/pathology , Flavonoids/pharmacology , Lung Neoplasms/pathology , Paclitaxel/pharmacology , A549 Cells , Actins/metabolism , Apoptosis/drug effects , Biomarkers , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Synergism , Flavonols , Gene Expression Profiling , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Neoplasm Metastasis , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Vimentin/metabolism
14.
Adv Clin Exp Med ; 27(3): 367-378, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29533537

ABSTRACT

BACKGROUND: Niacinamide is a stable and water-soluble form of vitamin B3, a valuable and versatile cosmetic ingredient, which is well absorbed and tolerated by the skin. A large body of literature has reported on the antioxidant and cell repair properties of niacinamide. Therefore, it has been shown to be useful in the protection of the skin against ultraviolet B (UVB) radiation and free radicals. Despite numerous hypotheses on the mechanism of vitamin B3, its protective effects have not yet been fully elucidated. OBJECTIVES: The aim of the study was to determine the protective effects of niacinamide on CHO AA8 cell line against UVB radiation. We assessed the following factors: cell death, cell cycle phase distributions, reorganization of main cytoskeletal proteins, such as F-actin, vimentin and ß-tubulin, and also alterations at the ultrastructural level. MATERIAL AND METHODS: The material used for our research was Chinese hamster ovary cell line (CHO AA8). We used 4 research groups: 1) control cells; 2) cells treated with niacinamide; 3) cells exposed to UV radiation; and 4) cells co-incubated with niacinamide and next exposed to ultraviolet. The cell death and cell cycle were evaluated by a Tali' based-image cytometer. A fluorescence microscope was used to assess the reorganization of cytoskeletal proteins, whereas a transmission electron microscope enabled the evaluation of the alterations at the ultrastructural level of cells. RESULTS: We showed that UV-induced apoptosis and cell cycle distributions during treatment with niacinamide resulted in a non-statistical significance in cell survival and no significant changes in the morphology and cytoskeleton in comparison to the control group. In turn, a combination of both factors led to an increase in the population of live cells and a decreased level of apoptotic cells in comparison to UV-exposed cells. CONCLUSIONS: Our results confirmed the harmful effects of UV radiation on CHO AA8 cell line. Furthermore, niacinamide can protect cells against these factors, and the mechanism of action may be related to the stabilization of the cell cytoskeleton.


Subject(s)
CHO Cells/drug effects , Niacinamide/pharmacology , Protective Agents/pharmacology , Ultraviolet Rays , Vitamin B Complex/pharmacology , Animals , Cricetinae , Cricetulus , Cytoskeletal Proteins , Niacinamide/administration & dosage , Protective Agents/administration & dosage , Vitamin B Complex/administration & dosage
15.
Exp Ther Med ; 13(3): 1057-1063, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28450942

ABSTRACT

Pseudoexfoliation syndrome (PEX) is an age-associated, sight disorder affecting elastic fibers in the eye and visceral organs but its exact etiology remains unknown. The purpose of the current study was to determine the morphology and ultrastructure of lens epithelial cells (LECs), and to use immunohistochemistry to examine localization of microsomal glutathione S-transferase 1 (MGST1) and clusterin. Anterior lens capsules were obtained from 24 patients (13 PEX and 11 controls) who underwent phacoemulsification. Immunohistochemistry was performed, using antibodies against MGST1 and clusterin, to determine their expression. The morphology and ultrastructure of LECs were evaluated by light and transmission electron microscopy, respectively. The PEX LECs were characterized by significantly lower MGST1 (P=0.0001) and clusterin expression (P=0.0005) compared with the control group patients. PEX LECs were also observed to have significantly increased thickness compared with the control group patients (P=0.0002). The current findings suggest that low MGST1 and clusterin expression levels may be an early clinical indicator of PEX, and that oxidative stress may serve an important role, but that the specific etiology of this disease has yet to be revealed.

16.
Acta Histochem ; 119(2): 99-112, 2017 Mar.
Article in English | MEDLINE | ID: mdl-27887793

ABSTRACT

To our knowledge, this study is the first to investigate the effect of the dietary flavonoid quercetin on the main cytoskeletal elements, namely microfilaments, microtubules and vimentin intermediate filaments, as well as cytoskeleton-driven processes in A549 non-small cell lung cancer cells. The methyl-thiazol-diphenyl-tetrazolium assay, annexin V/propidium iodide test, electron microscopic examination, cell cycle analysis based on DNA content, real-time PCR assays, in vitro scratch wound-healing assay, fluorescence staining of F-actin, ß-tubulin and vimentin were performed to assess the effects of quercetin on A549 cells. Our results showed that quercetin triggered BCL2/BAX-mediated apoptosis, as well as necrosis and mitotic catastrophe, and inhibited the migratory potential of A549 cells. The disassembling effect of quercetin on microfilaments, microtubules and vimentin filaments along with its inhibitory impact on vimentin and N-cadherin expression might account for the decreased migration of A549 cells in response to quercetin treatment. We also suggest that the possible mechanism underlying quercetin-induced mitotic catastrophe involves the perturbation of mitotic microtubules leading to monopolar spindle formation, and, consequently, to the failure of cytokinesis. We further propose that cytokinesis failure could also be a result of the depletion of actin filaments by quercetin. These findings are important to our further understanding of the detailed mechanism of the antitumor activity of quercetin and render this flavonoid a potentially useful candidate for combination therapy with conventional antimicrotubule drugs, nucleic acid-directed agents or novel cytoskeletal-directed agents.


Subject(s)
Antineoplastic Agents/pharmacology , Microtubules/drug effects , Quercetin/pharmacology , A549 Cells , Apoptosis/drug effects , Carcinoma, Non-Small-Cell Lung/drug therapy , Cell Movement/drug effects , Cell Survival/drug effects , Cytoskeletal Proteins/metabolism , Drug Screening Assays, Antitumor , Humans , Lung Neoplasms/drug therapy , Microtubules/metabolism , Tubulin Modulators/pharmacology
17.
Oncol Rep ; 35(5): 3084-93, 2016 May.
Article in English | MEDLINE | ID: mdl-26934847

ABSTRACT

Numerous studies on the biological mechanism of breast cancer have identified a number of potential therapeutic molecular targets. In this context, one type of potential candidates appears to be agents that target the actin cytoskeleton of cancer cells or regulate actin cytoskeleton dynamics. The aim of the present study was to study the impact of altered actin transport between the cytoplasm and nucleus by the downregulation of importin-9 (IPO9) in breast adenocarcinoma MCF-7 cells exposed to an apoptosis-inducing combination of garlic-derived S-allyl-L-cysteine sulfoxide (alliin) and paclitaxel (PTX). The expression of IPO9 was downregulated by the transfection of non-aggressive breast cancer MCF-7 cells with siRNA against IPO9. The altered expression of IPO9 and cofilin-1 (CFL1) was examined using western blotting. Moreover, the effect of the downregulation of IPO9 on cell death induced by the combination of PTX and alliin was also investigated. The alterations of IPO9 and CFL1 levels were also related with F-actin organizational changes and F-actin fluorescence intensity in the nuclear/perinuclear area of the cells. The results presented here indicate that alliin and PTX act synergistically to promote and potentiate apoptosis in MCF-7 cells. Furthermore, using RNA interference technique, we showed that downregulation of IPO9 expression was correlated with a significant reduction in the apoptotic cell population as well as with a decrease in F-actin content in whole cells, and in the cortical and nuclear/perinuclear areas of the cells. Simultaneously, the downregulation of IPO9 was also accompanied by the increased post-translational expression of CFL1. Furthermore, the data obtained in the present study allow us to conclude that CFL1 itself does not translocate actin into the cell nucleus but this transport requires the functional expression of IPO9.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Cysteine/analogs & derivatives , Karyopherins/metabolism , Paclitaxel/pharmacology , Actins/metabolism , Active Transport, Cell Nucleus , Apoptosis/drug effects , Breast Neoplasms/drug therapy , Cofilin 1/metabolism , Cysteine/pharmacology , Down-Regulation , Drug Screening Assays, Antitumor , Drug Synergism , Female , Garlic/chemistry , Humans , Karyopherins/genetics , MCF-7 Cells
18.
Cancer Cell Int ; 16: 10, 2016.
Article in English | MEDLINE | ID: mdl-26884726

ABSTRACT

BACKGROUND: The use of the dietary polyphenols as chemosensitizing agents to enhance the efficacy of conventional cytostatic drugs has recently gained the attention of scientists and clinicians as a plausible approach for overcoming the limitations of chemotherapy (e.g. drug resistance and cytotoxicity). The aim of this study was to investigate whether a naturally occurring diet-based flavonoid, fisetin, at physiologically attainable concentrations, could act synergistically with clinically achievable doses of paclitaxel to produce growth inhibitory and/or pro-death effects on A549 non-small cell lung cancer cells, and if it does, what mechanisms might be involved. METHODS: The drug-drug interactions were analyzed based on the combination index method of Chou and Talalay and the data from MTT assays. To provide some insights into the mechanism underlying the synergistic action of fisetin and paclitaxel, selected morphological, biochemical and molecular parameters were examined, including the morphology of cell nuclei and mitotic spindles, the pattern of LC3-II immunostaining, the formation of autophagic vacuoles at the electron and fluorescence microscopic level, the disruption of cell membrane asymmetry/integrity, cell cycle progression and the expression level of LC3-II, Bax, Bcl-2 and caspase-3 mRNA. RESULTS: Here, we reported the first experimental evidence for the existence of synergism between fisetin and paclitaxel in the in vitro model of non-small cell lung cancer. This synergism was, at least partially, ascribed to the induction of mitotic catastrophe. The switch from the cytoprotective autophagy to the autophagic cell death was also implicated in the mechanism of the synergistic action of fisetin and paclitaxel in the A549 cells. In addition, we revealed that the synergism between fisetin and paclitaxel was cell line-specific as well as that fisetin synergizes with arsenic trioxide, but not with mitoxantrone and methotrexate in the A549 cells. CONCLUSIONS: Our results provide rationale for further testing of fisetin in the combination with paclitaxel or arsenic trioxide to obtain detailed insights into the mechanism of their synergistic action as well as to evaluate their toxicity towards normal cells in an animal model in vivo. We conclude that this study is potentially interesting for the development of novel chemotherapeutic approach to non-small cell lung cancer.

19.
Acta Histochem ; 118(3): 225-35, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26805581

ABSTRACT

The aim of the study was to estimate the effect of tropomyosin-1-based structural stabilization of F-actin in transformed human alveolar epithelial line H1299 cells subjected to high oxidative stress induced by cigaret smoke extract. We demonstrated here that cigaret smoke extract induces cell shrinking and detachment as a consequence of F-actin cytoskeleton degradation in H1299 cells not overexpressing tropomyosin-1. Furthermore, the treatment of these cells with cigaret smoke extract resulted in the loss of peripheral localization of ZO-1 and initiated apoptosis. In contrast, structural stabilization of F-actin, by overexpression of tropomyosin-1, preserved cell to cell interactions through the attenuation of cortical actin organization into thin fibers and thus protected these cells against oxidative stress-induced degradation of actin cytoskeleton and cell death. In conclusion, we suggest that structural stabilization of thin cortical F-actin fibers increases link between tight junctions proteins and actin cytoskeleton and thus protects H1299 cells against cigaret smoke extract.


Subject(s)
Actins/metabolism , Alveolar Epithelial Cells/metabolism , Plant Extracts/toxicity , Tropomyosin/physiology , Alveolar Epithelial Cells/drug effects , Cell Line, Transformed , Gene Expression , Humans , Intercellular Junctions/metabolism , Oxidative Stress , Plant Extracts/chemistry , Protective Factors , Protein Stability , Proteolysis , Smoke , Nicotiana/chemistry , Zonula Occludens-1 Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...