Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
1.
Biol. Res ; 53: 11, 2020. graf
Article in English | LILACS | ID: biblio-1100917

ABSTRACT

BACKGROUND: Melanoma is one of the major types of skin cancer. The metastatic melanoma is among the most lethal forms of malignant skin tumors. We hereby aimed to characterize a novel microRNA (miR) in the metastatic melanoma model. METHODS: First, we evaluated the expression of miR-107 in melanoma cells and tumor tissues. The comparison between primary and metastatic cancer tissues was also accessed. Next, we examined the impact of miR-107 on melanoma cell proliferation, cell cycle, colony formation, apoptotic activity, migration and matrix invasion. A downstream target of miR-107 was also predicted and validated functionally in melanoma cells. RESULTS: Our findings showed miR-107 was significantly downregulated in melanoma. Its expression was lowest in metastatic form. Over-expression of miR-107 reduced melanoma cell proliferation, migration and invasion. POU3F2 was identified as the downstream target of miR-107. Over-expression of POU3F2 antagonized miR-107-mediated inhibitory effect on melanoma cells. CONCLUSION: Our study has reported miR-107 as a novel tumor suppressive factor in the metastatic melanoma model. It has provided new avenue to manage melanoma and improve the survival rate in the advanced stage.


Subject(s)
Humans , Gene Expression Regulation, Neoplastic , Genes, Tumor Suppressor , MicroRNAs/genetics , POU Domain Factors/genetics , Melanoma/genetics , Tumor Stem Cell Assay , Cell Movement , Cell Line, Tumor , Cell Proliferation
2.
Chinese journal of integrative medicine ; (12): 116-121, 2019.
Article in English | WPRIM | ID: wpr-776636

ABSTRACT

OBJECTIVE@#To investigate the effects of ethanol extract of Patrinia scabiosaefolia (EEPS) on chemo-resistance of colorectal cancer cells (CRC) and explore the possible molecular mechanisms.@*METHODS@#5-fluorouracil (5-FU)-resistant human colorectal carcinoma cell line (HCT-8/5-FU) and its parental cells HCT-8 were treated with EEPS (0, 0.25, 0.50, 1 or 2 mg/mL), or 5-FU (0, 100, 200, 400, 800 or 1600 μmol/L). The 3-(4,5-dimethylthiazol-2-yl)-2, 5-diphenyltetrazolium bromide (MTT) assay was performed to evaluate the cell viability. Cell density was observed by phase-contrast microscope, cell counting and colony formation assay were used to determine the cell proliferation of HCT-8/5-FU cells treated with 0, 0.5, 1 or 2 mg/mL EEPS. Cell apoptosis was determined by Hoechst staining. Western-blot was performed to detect the phosphorylation of AKT as well as the protein expression level of B-cell CLL/lymphoma 2 (Bcl-2) and Bcl-2-associated X protein (Bax).@*RESULTS@#Compared with HCT-8 cells, MTT assay results indicated that HCT-8/5-FU cells were resistant to 5-FU treatment (P0.05). Moreover, compared with untreated HCT-8/5-FU cells, 1 and 2 mg/mL of EEPS treatment significantly reduced cell density, cell number, inhibited cell survival (P<0.05), and induced apoptosis in HCT-8/5-FU cells. Furthermore, 1 and 2 mg/mL of EEPS significantly decreased the phosphorylation level of p-AKT and Bcl-2 protein expression, and increased the expression of Bax protein (P<0.05).@*CONCLUSION@#EEPS is a promising therapeutic agent that may overcome chemo-resistance in cancer cells, likely through suppression of the AKT pathway and promotion of cancer cell apoptosis.


Subject(s)
Humans , Apoptosis , Cell Line, Tumor , Cell Proliferation , Cell Survival , Colorectal Neoplasms , Drug Therapy , Pathology , Drug Resistance, Neoplasm , Fluorouracil , Pharmacology , Therapeutic Uses , Patrinia , Chemistry , Phosphorylation , Proto-Oncogene Proteins c-akt , Metabolism , Signal Transduction , Tumor Stem Cell Assay , bcl-2-Associated X Protein , Metabolism
3.
Journal of Central South University(Medical Sciences) ; (12): 583-588, 2018.
Article in Chinese | WPRIM | ID: wpr-813223

ABSTRACT

To investigate the effect of manganese superoxide dismutase (MnSOD) silence on the in vitro tumorigenicity in human small cell lung cancer NCI-H446 cells and the underlying mechanisms.
 Methods: Sphere formation cells from NCI-H446 cells were obtained by suspension culture, while the expression of MnSOD and urokinase type plasminogen activator (uPAR) was analyzed by Western blot. Silence of MnSOD was performed by adenovirus infection in the second passage formation cells, and the effect of MnSOD silence on tumorigenicity in NCI-H446 cells was evaluated by sphere formation assay and soft-agar colony formation assay, while the expression of uPAR was analyzed by Western blot.
 Results: Compared with NCI-H446 cells, the sphere formation rate, colony formation rate, and the expression of MnSOD and uPAR were significantly increased in the second passage sphere formation cells in NCI-H446 cells (P<0.05). Silence of MnSOD inhibited the sphere formation rate, colony formation rate, and the expression level of uPAR in the second passage sphere formation cells in NCI-H446 cells.
 Conclusion: MnSOD may promote tumorigenicity in NCI-H446 cells by up-regulation of uPAR expression in vitro.


Subject(s)
Humans , Adenoviridae , Carcinogenesis , Cell Line, Tumor , In Vitro Techniques , Lung Neoplasms , Metabolism , RNA Interference , Receptors, Urokinase Plasminogen Activator , Genetics , Metabolism , Small Cell Lung Carcinoma , Metabolism , Spheroids, Cellular , Pathology , Superoxide Dismutase , Genetics , Metabolism , Tumor Stem Cell Assay , Up-Regulation
4.
Braz. j. med. biol. res ; 51(4): e6803, 2018. graf
Article in English | LILACS | ID: biblio-889059

ABSTRACT

Propofol is an intravenous sedative hypnotic agent of which the growth-inhibitory effect has been reported on various cancers. However, the roles of propofol in endometrial cancer (EC) remain unclear. This study aimed to explore the effects of propofol on EC in vitro and in vivo. Different concentrations of propofol were used to treat Ishikawa cells. Colony number, cell viability, cell cycle, apoptosis, migration, and invasion were analyzed by colony formation, MTT, flow cytometry, and Transwell assays. In addition, the pcDNA3.1-Sox4 and Sox4 siRNA plasmids were transfected into Ishikawa cells to explore the relationship between propofol and Sox4 in EC cell proliferation. Tumor weight in vivo was measured by xenograft tumor model assay. Protein levels of cell cycle-related factors, apoptosis-related factors, matrix metalloproteinases 9 (MMP9), matrix metalloproteinases 2 (MMP2) and Wnt/β-catenin pathway were examined by western blot. Results showed that propofol significantly decreased colony numbers, inhibited cell viability, migration, and invasion but promoted apoptosis in a dose-dependent manner in Ishikawa cells. Moreover, propofol reduced the expression of Sox4 in a dose-dependent manner. Additionally, propofol significantly suppressed the proportions of Ki67+ cells, but Sox4 overexpression reversed the results. Furthermore, in vivo assay results showed that propofol inhibited tumor growth; however, the inhibitory effect was abolished by Sox4 overexpression. Moreover, propofol inhibited Sox4 expression via inactivation of Wnt/β-catenin signal pathway. Our study demonstrated that propofol inhibited cell proliferation, migration, and invasion but promoted apoptosis by regulation of Sox4 in EC cells. These findings might indicate a novel treatment strategy for EC.


Subject(s)
Animals , Female , Apoptosis/drug effects , Endometrial Neoplasms/drug therapy , Hypnotics and Sedatives/pharmacology , Propofol/pharmacology , SOXC Transcription Factors/metabolism , beta Catenin/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Cell Survival/drug effects , Endometrial Neoplasms/pathology , Mice, Inbred BALB C , Neoplasm Invasiveness , Propofol/administration & dosage , Tumor Stem Cell Assay , Wnt Proteins/metabolism , Xenograft Model Antitumor Assays
5.
São Paulo; s.n; 20180000. 117 p.
Thesis in Portuguese | LILACS, BBO | ID: biblio-1005349

ABSTRACT

O carcinoma mucoepidermóide (CME) é o tumor mais comum entre as neoplasias malignas de glândula salivar. Recentemente, uma rara população de células com características de multipotência, autorrenovação e potencial tumorigênico, denominadas como células tronco tumorais (CTT), foi descrita no CME. As CTT são resistentes as terapias atuais, e têm sido consideradas responsáveis pela recorrência e metástase, levando a um pior prognóstico para o paciente. A descoberta de que as CTT do CME superexpressam componentes da via de sinalização mTOR, levantou a hipótese que os pacientes poderiam ser beneficiados com o uso inibidores de mTOR como terapia para eliminação das CTT. O objetivo desse estudo foi avaliar o potencial uso de inibidores de mTOR como terapia para o CME com foco em CTT, assim como, investigar o funcionamento da via de sinalização mTOR e os efeitos moleculares do tratamento com inibidores dessa via nas CTT do CME. Foi realizada imuno-histoquímica para p-mTOR e p-S6K-1 em casos de pacientes diagnosticados com CME, os resultados foram correlacionados com os dados clínicos dos pacientes e também foi realizada dupla marcação por imunofluorescência para ALDH/p-mTOR. Estudos in vitro foram realizados em 3 linhagens de CME (UM-HMC-1, -3A, -3B) e com inibidores da via de sinalização mTOR. Após exposição aos inibidores, realizou-se ensaios de western blot (proteínas da via mTOR e BMI-1), citometria de fluxo para ALDH/CD44; salisfera; e apoptose, esse último comparando com quimioterápicos utilizados atualmente. Adicionalmente, foi utilizado o silenciamento genético de mTOR para confirmar os resultados obtidos com inibidores químicos. Por fim, foram realizados ensaios in vivo com as células silenciadas e com o inibidor de mTOR tensirolimo. Os resultados evidenciaram que a via de sinalização mTOR está ativa no CME, é correlacionada com pior prognóstico clínico e está superexpressa nas CTT. O tratamento com inibidores da via mTOR levaram a diminuição da fração de CTT, devido a perda de autorrenovação e apoptose das CTT. A apoptose, junto a diminuição de p-AKT revelada por western blot, sugeriram que esteja ocorrendo inibição de mTORC2 nas CTT, um importante componente na eficácia do tratamento com inibição de mTOR no câncer. Além disso, também houve redução de vasos sanguíneos, nichos das CTT, e diminuição do crescimento tumoral com uso de inibidores ou silenciando mTOR in vivo. Coletivamente, os resultados mostraram que a inibição de mTOR foi capaz de agir nas CTT por mecanismos diretos (indução de apoptose e diminuição da autorrenovação) e indiretamente através da redução de angiogênese, sugerindo que o uso de inibidores de mTOR no tratamento do CME é uma estratégia molecular eficiente para a redução de CTT, e uma potencial terapia adjuvante.


Subject(s)
Tumor Stem Cell Assay , Carcinoma, Mucoepidermoid , Drug Therapy
6.
Braz. j. med. biol. res ; 51(1): e6822, 2018. tab, graf
Article in English | LILACS | ID: biblio-889012

ABSTRACT

Cervical cancer is a public health problem and the molecular mechanisms underlying radioresistance are still poorly understood. Here, we evaluated the modulation of key molecules involved in cell proliferation, cell cycle and DNA repair in cervical cancer cell lines (CASKI and C33A) and in malignant tissues biopsied from 10 patients before and after radiotherapy. The expression patterns of epidermal growth factor receptor (EGFR), excision repair cross-complementation group 1 (ERCC1) and p53 were evaluated in cancer cell lines by quantitative PCR and western blotting, and in human malignant tissues by immunohistochemistry. The mutation status of TP53 gene was evaluated by direct sequencing. Among cell lines, absent or weak modulations of EGFR, ERCC1 and p53 were observed after exposure to 1.8 Gy. Conversely, increased expressions of p53 (5/10 patients; P=0.0239), ERCC1 (5/10 patients; P=0.0294) and EGFR (4/10 patients; P=0.1773) were observed in malignant tissues after radiotherapy with the same radiation dose. TP53 mutations were found only in one patient. Here we show that a single dose of radiotherapy induced EGFR, ERCC1 and p53 expression in malignant tissues from cervical cancer patients but not in cancer cell lines, highlighting the gap between in vitro and in vivo experimental models. Studies on larger patient cohorts are needed to allow an interpretation that an upregulation of p53, EGFR and ERCC1 may be part of a radioresistance mechanism.


Subject(s)
Humans , Female , Adult , Middle Aged , Aged , Carcinoma, Squamous Cell/radiotherapy , Uterine Cervical Neoplasms/radiotherapy , Genes, p53/radiation effects , Genes, erbB-1/radiation effects , DNA-Binding Proteins/radiation effects , Endonucleases/radiation effects , Immunohistochemistry , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Tumor Stem Cell Assay , Blotting, Western , Prospective Studies , Cell Line, Tumor , Mutation
7.
Biol. Res ; 50: 15, 2017. graf
Article in English | LILACS | ID: biblio-838968

ABSTRACT

BACKGROUND: Ubiquitin specific peptidase 39 (USP39), an essential factor in the assembly of the mature spliceosome complex, has an aberrant expression in several cancer. However, its function and the corresponding mechanism on human osteosarcoma has not been fully explored yet. METHODS: The mRNA and DNA copies of USP39 were increased in osteosarcoma cancer tissues compared with the one in human normal tissues according to datasets from the publicly available Oncomine database. A further western blot analysis also demonstrated an aberrant endogenous expression of USP39 in three different osteosarcoma cells. Then lentivirus-mediated short hairpin RNA (shRNA) was designed to silence USP39 in human osteosarcoma cell line U2OS, which is used to test the impact of USP39-silencing on cellular proliferation, colony formation, cell cycle distribution and apoptosis. RESULTS: Knockdown of USP39 expression in U2OS cell significantly decreased cell proliferation, impaired colony formation ability. A further analysis indicated suppression of USP39 arrested cell cycle progression at G2/M phase via p21 dependent way. In addition, the results of Annexin V/7-AAD staining suggested the knockdown of USP39 could promote U2OS cell apoptosis through PARP cleavage. CONCLUSIONS: These results uncover the critical role of USP39 in regulating cancer cell mitosis and indicate USP39 is critical for osteosarcoma tumorigenesis.


Subject(s)
Humans , Osteosarcoma/enzymology , Osteosarcoma/pathology , Apoptosis , Gene Knockdown Techniques/methods , Ubiquitin-Specific Proteases/metabolism , Tumor Stem Cell Assay , Gene Expression Regulation, Neoplastic , Lentivirus , Cell Line, Tumor , Cell Proliferation , Ubiquitin-Specific Proteases/genetics , Flow Cytometry , Genetic Vectors
8.
Int. j. morphol ; 34(4): 1197-1202, Dec. 2016. ilus
Article in English | LILACS | ID: biblio-840866

ABSTRACT

Cancer has been considered as a stem cell disease. Suspension culture combined with anti-cancer drugs has recently been proposed for isolation of cancer stem cells (CSCs). In the current study, Vincristine as an anti-cancer drug combined with suspension culture was used for isolation and purification of CSCs from human breast cancer cell line (MDA-MB231). The cells were treated with different concentrations of vincristine (0, 2, 4, 6 and 8 ng/ml). Stem cells were identified with the expression of OCT4, nanog, SOX2 and nucleostemin genes by RT-PCR. Mammosphere forming unit was measured upon suspension culture containing EGF, bFGF, LIF, B27, insulin and BSA. The isolated mammospheres were investigated for CD44 expression. Results showed that 4 ng/ml of vincristine for 72 hours could be utilized as the best and most reliable dose which eliminates around 80 % of non-cancer stem cells with no destructive effect on CSCs' viability (P> 0.05). RT-PCR demonstrated that drug treated cells expressed OCT4, nanog, SOX2 and nucleostemin. Mammosphere formation unit of cells pretreated with vincristine was significantly higher than unpretreated ones (P>0.05). Immunofluorescence staining for CD44 depicted high expression of CSC marker among the isolated mammospheres. Vincristine combined with suspension culture can be considered as an appropriate method to isolate CSC.


El cáncer ha sido considerado como una enfermedad de células madre. Recientemente se ha propuesto cultivo en suspensión en combinación con medicamentos contra el cáncer para aislamiento de las células madre del cáncer (CMC). En este estudio se utilizó la vincristina como fármaco anticanceroso combinado con cultivo en suspensión para el aislamiento y purificación de las células madre cancerosas, de la línea celular de cáncer de mama humano (MDA-MB231). Las células se trataron con diferentes concentraciones de vincristina (0, 2, 4, 6 y 8 ng/ml). Las células madre se identificaron mediante la expresión de los genes OCT4, Nanog, SOX2 y nucleostemin por RT-PCR. La unidad de formación mammosphere se midió a través de cultivo en suspensión que contenía EGF, bFGF, LIF, B27, insulina y BSA. Los mammospheres aislados fueron estudiados para la expresión de CD44. Los resultados mostraron que 4 ng/ml de vincristina durante 72 horas podrían ser utilizados como la mejor y más fiable dosis que permite eliminar alrededor del 80 % de las células madre no cancerosas, sin causar un efecto destructivo sobre la viabilidad de las CMC (P> 0,05). La RT-PCR mostró que en las células tratadas con él fármaco hubo expresión de los genes OCT4, Nanog, SOX2 y nucleostemin. La unidad de formación de las células pretratadas con vincristina fue significativamente más alta que las unidades sin tratamiento previo (P>0,05). La inmunofluorescencia para CD44 muestró una alta expresión del marcador de CMC entre mammospheres aisladas. La vincristina en combinación con el cultivo en suspensión puede ser considerado como un método apropiado para aislar CMC.


Subject(s)
Humans , Female , Antineoplastic Agents, Phytogenic/pharmacology , Breast Neoplasms/pathology , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Vincristine/pharmacology , Hyaluronan Receptors/metabolism , Cell Line, Tumor , Fluorescent Antibody Technique, Indirect , Nanog Homeobox Protein , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Reverse Transcriptase Polymerase Chain Reaction , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , Staining and Labeling , Tumor Stem Cell Assay
9.
Chinese Journal of Oncology ; (12): 731-735, 2015.
Article in Chinese | WPRIM | ID: wpr-286734

ABSTRACT

<p><b>OBJECTIVE</b>To investigate whether SIS3, a specific inhibitor of Smad3 phosphorylation, can reverse the stemness of multidrug-resistant(MDR) hepatocellular carcinoma cells.</p><p><b>METHODS</b>MDR HCC Huh7.5.1/ADM cell lines were developed by exposing parental cells to stepwise increasing concentrations of ADM. CCK-8 assay was used to determine the cellular sensitivity of various anticancer drugs. Flow cytometry (FCM) was used to analyze the expression level of cancer stem cell marker CD133. Clone formation assay and mouse subcutaneous xenograft tumors were used to investigate the tumorigenicity in vitro and in vivo. Western blotting (WB) was used to analyze the changes of expressions of CD133, Smad3, Bcl-2, Bax and p-Smad3 in different conditions.</p><p><b>RESULTS</b>ADM treatment of HCC cells in vitro resulted in a development of subline, Huh7.5.1/ADM cells, with CSC phenotypes: stable MDR phenotype (besides ADMc Huh7.5.1/ADM cells were also more resistant to some other anticancer drugs including VCR, MMC and CTX ) (IC50: 0.215 ± 0.018 vs. 0.123 ± 0.004, 0.145 ± 0.009 vs. 0.014 ± 0.002, 1.021 ± 0.119 vs. 0.071 ± 0.006, 27.007 ± 1.606 vs. 1.919 ± 0.032) (unit: µg/ml) (P<0.05). Huh7.5.1/ADM cells enriched the cancer stem-like cell fraction (CD133-positive subpopulation) (76.06 ± 2.948% vs. 25.38 ± 4.349%) (P<0.05), had stronger tumorigenicity in vivo and colony formation ability, and activated the Smad3 activity. Inhibition of Smad3 activity by SIS3 decreased stemness of the Huh7.5.1/ADM cells: CD133-positive subpopulation (48.49 ± 2.304% vs. 76.06 ± 2.948%) (P<0.05); ADM IC50: (0.112 ± 0.019 vs. 0.215 ± 0.018), VCR IC50 (0.065 ± 0.013 vs. 0.145±0.009), MMC IC₅₀ (0.749 ± 0.121 vs. 1.021 ± 0.119), CTX IC50 (10.576 ± 1.248 vs. 27.007 ± 1.606) (unit: µg/ml) (P<0.05), and decreased tumorigenicity and colony formation ability.</p><p><b>CONCLUSION</b>SIS3 as a specific inhibitor of Smad3 signal is involved in the stemness of multidrug resistant hepatocellular carcinoma cells.</p>


Subject(s)
Animals , Humans , Mice , AC133 Antigen , Antibiotics, Antineoplastic , Pharmacology , Antigens, CD , Metabolism , Carcinoma, Hepatocellular , Drug Therapy , Metabolism , Pathology , Doxorubicin , Pharmacology , Drug Resistance, Neoplasm , Glycoproteins , Metabolism , Heterografts , Isoquinolines , Pharmacology , Liver Neoplasms , Drug Therapy , Metabolism , Pathology , Neoplasm Proteins , Metabolism , Neoplastic Stem Cells , Peptides , Metabolism , Proto-Oncogene Proteins c-bcl-2 , Metabolism , Pyridines , Pharmacology , Pyrroles , Pharmacology , Smad3 Protein , Metabolism , Tumor Stem Cell Assay , bcl-2-Associated X Protein , Metabolism
10.
Chinese Journal of Stomatology ; (12): 360-365, 2015.
Article in Chinese | WPRIM | ID: wpr-294703

ABSTRACT

<p><b>OBJECTIVE</b>To investigate the effect of metformin on the proliferation and cell apoptosis of oral squamous cell carcinoma (OSCC) (HSC-3, HSC-4) in vitro and in vivo.</p><p><b>METHODS</b>HSC-3, HSC-4 cells were treated with metformin at different concentration (2-50 mmol/L) for 24, 48 or 72 hours. In vitro cell proliferation ability was determined by 3-(4,5-dimethylthiazol-2-yl)-2, 5-diphenyltetrazolium bromide (MTT) assay and colony formation assay. Cell cycle progression was assessed by flow cytometry. Cell apoptosis was tested by both TdT-mediated dUTP nick-end labeling (TUNEL) assay and flow cytometry. The activation of related cell markers was examined by immunohistochemistry. Xenograft mouse model was used to demonstrate the in vivo anti-tumor effect of metformin. A total of 30 BALB/c mice were randomly divided into control groups (water + phosphate buffer saline, PBS) and treatment groups (pre-oral, oral or intraperitoneal injection). Each group had 6 mice. The tumor size was measured once every three days until endpoint (35 days). After sacrificing the mice, tumor tissue was removed, sectioning and then analyzed by TUNEL or immunohistochemistry (IHC) assays.</p><p><b>RESULTS</b>Metformin inhibited proliferation and colony formation of HSC-3, HSC-4 in a time- and dose-dependent manner. The cell proliferation was significantly reduced when treated with 5, 10, 20 and 40 mmol/L metformin for 48 and 72 hours (P < 0.05).The colony formation of OSCC cells treated with metformin for 72 hours in vitro had the same result. Treated with 2, 20 and 50 mmol/L metformin for 24 hours increased the ratio of G0/G1 1.2-1.8 fold compared with the control group on HSC-4 cell. The percentage of apoptosis cell rose from 10% (control) to around 30% (treatment) in vitro. Metformin also decreased the size of xenografts by 82.5% (pre-oral), 63.9% (oral), and 62.8% (oral or intraperitoneal injection). The percentage of apoptosis cell rose from lower than 10% (control) to 70% (pre-oral), 50% (oral), and 25% (oral or intraperitoneal injection). The percentage of PCNA positive cell was lower than 60% (control group was normalized to 100%).</p><p><b>CONCLUSIONS</b>Metformin could inhibit the growth of OSCC cell line (HSC-3, HSC-4) by reducing cell proliferation and increasing cell apoptosis in vitro and in vivo. Therefore metformin could be a potential new treatment candidate for human OSCC.</p>


Subject(s)
Animals , Humans , Mice , Apoptosis , Carcinoma, Squamous Cell , Drug Therapy , Pathology , Cell Cycle , Physiology , Cell Line, Tumor , Cell Proliferation , Flow Cytometry , Formazans , In Vitro Techniques , Metformin , Pharmacology , Mice, Inbred BALB C , Mouth Neoplasms , Drug Therapy , Pathology , Random Allocation , Tetrazolium Salts , Time Factors , Tumor Stem Cell Assay
11.
Braz. j. med. biol. res ; 47(12): 1044-1049, 12/2014. graf
Article in English | LILACS | ID: lil-727666

ABSTRACT

Protein phosphatase magnesium/manganese-dependent 1D (PPM1D) is a p53-induced phosphatase that functions as a negative regulator of stress response pathways and has oncogenic properties. However, the functional role of PPM1D in bladder cancer (BC) remains largely unknown. In the present study, lentivirus vectors carrying small hairpin RNA (shRNA) targeting PPM1D were used to explore the effects of PPM1D knockdown on BC cell proliferation and tumorigenesis. shRNA-mediated knockdown of PPM1D significantly inhibited cell growth and colony forming ability in the BC cell lines 5637 and T24. Flow cytometric analysis showed that PPM1D silencing increased the proportion of cells in the G0/G1 phase. Downregulation of PPM1D also inhibited 5637 cell tumorigenicity in nude mice. The results of the present study suggest that PPM1D plays a potentially important role in BC tumorigenicity, and lentivirus-mediated delivery of shRNA against PPM1D might be a promising therapeutic strategy for the treatment of BC.


Subject(s)
Animals , Humans , Male , Phosphoprotein Phosphatases/physiology , RNA Interference/physiology , RNA, Small Interfering/pharmacology , Urinary Bladder Neoplasms/pathology , Cell Line, Tumor , Carcinogenesis/drug effects , Cell Proliferation/drug effects , Down-Regulation , Flow Cytometry , Gene Knockdown Techniques , Genetic Vectors , Lentivirus/genetics , Mice, Inbred BALB C , Mice, Nude , Phosphoprotein Phosphatases/genetics , Real-Time Polymerase Chain Reaction , Stress, Physiological/genetics , Tumor Stem Cell Assay , Urinary Bladder Neoplasms/therapy
12.
Journal of Huazhong University of Science and Technology (Medical Sciences) ; (6): 621-625, 2014.
Article in English | WPRIM | ID: wpr-351028

ABSTRACT

Tumors are believed to consist of a heterogeneous population of tumor cells originating from rare cancer stem cells (CSCs). However, emerging evidence suggests that tumor may also originate from non-CSCs. To support this viewpoint, we are here to present definitive evidence indicating that the number of tumorigenic tumor cells is greater than that of CSCs in tumor, and tumor can also derive from non-CSCs. To achieve this, an idealized mathematical model was employed in the present study and theoretical calculation revealed that non-CSCs could initiate the occurrence of tumor if their proliferation potential was adequate. Further, experimental studies demonstrated that 17.7%, 38.6% and 5.2% of tumor cells in murine B16 solid melanoma, H22 hepatoma and Lewis lung carcinoma, respectively, were potentially tumorigenic. Thus, based on the aforementioned findings, we propose that the scarce CSCs, if exist, are not the sole source of a tumor.


Subject(s)
Animals , Algorithms , Carcinogenesis , Pathology , Carcinoma, Lewis Lung , Pathology , Cell Differentiation , Cell Line, Tumor , Cell Proliferation , Liver Neoplasms, Experimental , Pathology , Melanoma, Experimental , Pathology , Mice, Inbred BALB C , Mice, Inbred C57BL , Models, Biological , Neoplasms, Experimental , Pathology , Neoplastic Stem Cells , Pathology , Time Factors , Tumor Stem Cell Assay , Methods
13.
Chinese journal of integrative medicine ; (12): 36-42, 2014.
Article in English | WPRIM | ID: wpr-267187

ABSTRACT

<p><b>OBJECTIVE</b>To determine the antiproliferative activity of Rubus parvifolius L. (RP) extract, its medicinal serum and RP total saponins (RPTS) against K562 cells in vitro and in vivo.</p><p><b>METHODS</b>Nude mice models bearing leukemia tumors were treated with different concentrations of RP extract. The size, weight and histopathological change of leukemic tumors were determined. Semi-solid agar culture and methylthiazolyl tetrazolium (MTT) assay were used to determine in vitro the inhibition of colony formation and proliferation of K562 cells respectively by different concentrations of RP medicinal serum and RPTS.</p><p><b>RESULTS</b>RP extract had a tumor inhibition rate of 84.8% when administered to mice at a dose of 1.0 g/day of crude RP root equivalent. Semi-solid agar culture of K562 cells in the presence of 20% (v/v) of RP medicinal serum and 150 mg/L RPTS demonstrated a 50.8% and 100% inhibition of the colony forming unit (CFU)-K562, respectively. The same doses of RP medicinal serum and RPTS showed a proliferation inhibition of 31.4% and 86.3%, respectively against K562 cells in MTT assay.</p><p><b>CONCLUSION</b>RP extract and RPTS show effective antiproliferative activity against myeloid leukemia cells in vitro and in vivo.</p>


Subject(s)
Animals , Humans , Mice , Agar , Cell Proliferation , Chromatography, High Pressure Liquid , K562 Cells , Leukemia , Drug Therapy , Pathology , Mice, Inbred BALB C , Mice, Nude , Plant Extracts , Pharmacology , Therapeutic Uses , Rosaceae , Chemistry , Saponins , Pharmacology , Therapeutic Uses , Subcutaneous Tissue , Pathology , Tumor Stem Cell Assay , Xenograft Model Antitumor Assays
14.
São Paulo; s.n; s.n; 2014. 158 p. ilus, graf, tab.
Thesis in Portuguese | LILACS | ID: biblio-881898

ABSTRACT

Células tumorais têm sua proliferação e mobilidade modificada por diversos fatores de crescimento, citocinas e mediadores inflamatórios, dentre os quais a amilóide sérica A (SAA). Estudos prévios do nosso grupo mostraram o efeito direto da SAA em processos de proliferação, migração e invasão de células de glioblastoma multiforme (GBM), A172 e T98G. Neste estudo nós complementamos resultados prévios de migração e invasão; avaliamos SAA como possível indutora de moléculas importantes para a invasividade do tumor, como as MMP-2 e -9 e ROS; realizamos ensaio clonogênico com a intenção de investigar uma possível contribuição da rSAA no estágio inicial de desenvolvimento do tumor; avaliamos o impacto da hipóxia na expressão dos diferente genes da SAA; estimulamos as células com indutores hepáticos clássicos da SAA e analisamos a possibilidade destes induzirem os diferentes genes da SAA em células tumorais; avaliamos possíveis receptores e vias de sinalizações envolvidas nos processos de proliferação, migração e invasão. Construímos knockdowns (KDs) dos genes da SAA de fase aguda (SAA1 e 2) e constitutiva (SAA4) e avaliamos a função de cada um deles para a morfologia e para os processos de proliferação, migração e invasão de GBM. Por fim investigamos SAA como possível biomarcadora de gliomas em amostras clínicas. Nossos resultados sugerem que rSAA afetou a atividade das MMP-2 e -9 e a produção de ROS em ambos GBM, mas não se mostrou clonogênica. As citocinas IL-6, TNF-α e IL-1ß, mas não a hipóxia, foram capazes de induzir os diferentes genes da SAA. A adição de rSAA às culturas celulares estimulou a transrição dos diferentes genes da SAA, sugerindo a ativação de mecanismos intracelulares retroalimentados. Efeitos pró-tumorais da rSAA parecem ser viabilizados via RAGE, enquanto efeitos anti-tumorais parecem ser induzidos via TLR-4. Pela primeira vez mostramos que SAA induz aumento de RAGE. KDs da SAA inibiram proliferação, migração e invasão, sugerindo que SAA seja um produto tumoral importante para a manutenção do fenótipo invasivo de GBM. A adição de SAA exógena reverteu grande parte dos efeitos nas células T98G KD, enquanto células A172 KD responderam parcialmente à rSAA. KDs da SAA sugerem a mesma como mantenedora da morfologia das células de GBM. De maneira inédita mostramos que o gene SAA4 até então descrito como um gene constitutivo de função desconhecida é importante para a proliferação, migração e invasão de GBM. Nós especulamos que os efeitos diferenciados induzidos por rSAA nos GBM estejam associados à natureza multiligante da SAA e às diferenças genéticas dos GBM. Pacientes com GBM apresentaram aumento significativo na transcrição e expressão de SAA1 no tecido tumoral, bem como aumento sérico de SAA. A correlação na expressão de SAA1 com moléculas importantes para progressão tumoral, como CXCR4, CXCR7, CD163 e HIF-1α também a identificam como uma proteína associada à malignidade


Tumor cells have their proliferation and migration modified by several growth factors, cytokines and inflammatory mediators, such as serum amyloid A (SAA). Previous studies from our group showed the direct effect of SAA on proliferation, migration and invasion of glioblastoma multiforme (GBM) cells, A172 and T98G. In this study we complemented previous migration and invasion data; evaluated SAA as possible inducer of MMP-2, -9 and ROS; performed clonogenic assay to investigate a possible contribution of rSAA in the early stage of tumor development; evaluated the impact of hypoxia on the expression of different genes of SAA; stimulated the cells with classics inducers of hepatic SAA and analyzed the possibility of these different genes to induce SAA in tumor cells; evaluated possible receptors and signaling pathways involved in proliferation, migration and invasion processes. SAA knockdowns (KDs) were made for acute phase (SAA1 and 2) and constitutive protein (SAA4) and evaluated their role in cell proliferation, migration, morfology and invasion. Finally it was investigated SAA as a possible biomarker of glioma grade in clinical samples. Our results suggest that rSAA affects MMP-2 and -9 activity and ROS production in both GBM, but did not affect clonogenicity. IL-6, TNF-α and IL-1ß, but not hypoxia, were able to induce SAA expression. rSAA addition to cell cultures stimulated transcription of the three different SAA genes, suggesting the activation of intracellular feedback mechanisms. Pro-tumor effects of rSAA seem to occur via RAGE and anti-tumor effects appear to be induced via TLR-4. This was de first time that induction of RAGE triggered by rSAA was shown. Proliferation, migration and invasion were inhibited in SAA KDs, suggesting that SAA is an important tumoral product for the maintenance of the invasive phenotype of GBM. The addition of exogenous SAA largely reversed the effects on SAA KDs T98G cells, whereas SAA KDs A172 cells partially responded to the rSAA. The findings with SAA KDs suggest that SAA affect cell morphology. Another new contribution from our study was that SAA4, a constitutive gene with unknown function, was important for the proliferation, migration and invasion of GBM and it can be induced by rSAA, IL-6, TNF-α and IL-1ß. We speculate that the different effects induced by rSAA in GBM are associated with the affinity of SAA to different receptors and the different genetic backgrounds of GBM. Patients with GBM showed a significant increase in the transcription and expression of SAA1 in tumor tissue as well as increased serum SAA. The correlation between the expression of SAA1 with important molecules for tumor progression, such as CXCR4, CXCR7, CD163 and HIF-1α also identified SAA as a protein associated with malignancy


Subject(s)
Serum Amyloid A Protein/analysis , Glioblastoma/metabolism , Tumor Stem Cell Assay/methods , Cell Hypoxia , Cell Movement , Colony-Forming Units Assay , Cell Proliferation , Gene Knockdown Techniques/methods
15.
Chinese journal of integrative medicine ; (12): 119-126, 2013.
Article in English | WPRIM | ID: wpr-347115

ABSTRACT

<p><b>OBJECTIVE</b>To investigate the effects of serum containing Chinese medicine (CM) Sanpi Pingwei (, SPPW) formula on the proliferation and apoptosis of human SGC-7901 cells and the possible mechanism.</p><p><b>METHODS</b>Serum containing CM SPPW formula (SPPW serum) was prepared by a serum pharmacology method. Human SGC-7901 cells were incubated with SPPW serum at three different concentrations and with the anticancer drug 5-fluorouracil (5-FU), respectively. Cell proliferation was assessed by MTT assay, and cell apoptosis was detected by flow cytometry assay. Real-time quantitative polymerase chain reaction (RT-PCR) and Western blot assay were employed to confirm the expressions of Bcl-2, Bax and p53 in SGC-7901 cells at mRNA and protein levels, respectively.</p><p><b>RESULTS</b>SPPW serum suppressed the proliferation of SGC-7901 cells in a time- and dose-dependent manner. The colony forming rate of negative control was 48.2%, while those in the three SPPW serum groups and the 5-FU group decreased significantly (P<0.01). The number of colony forming units in the SPPW high dosage group was significantly smaller than that in the 5-FU group (P<0.01). MTT assay showed that SPPW serum restrained the proliferation of SGC-7901 cells, and the inhibition rate increased significantly in a dose-dependent manner. Annexin V/PI Assay suggested that SPPW serum induced the apoptosis of SGC-7901 cells significantly. RT-PCR and western blot assay indicated that SPPW serum upregulated the protein and mRNA expression levels of Bax and p53 in SGC-7901 cells, but downregulated the protein and mRNA expressions of Bcl-2.</p><p><b>CONCLUSIONS</b>SPPW formula inhibits the proliferation of SGC-7901 cells in vitro and induces the cell apoptosis. It plays an anticancer role by regulating the expressions of Bax, p53 and Bcl-2 in SGC-7901 cells.</p>


Subject(s)
Animals , Humans , Rats , Apoptosis , Blotting, Western , Cell Line, Tumor , Cell Proliferation , Drugs, Chinese Herbal , Pharmacology , Fluorouracil , Pharmacology , Gene Expression Regulation, Neoplastic , RNA, Messenger , Genetics , Metabolism , Rats, Sprague-Dawley , Serum , Chemistry , Tumor Stem Cell Assay , Tumor Suppressor Protein p53 , Genetics , Metabolism , bcl-2-Associated X Protein , Genetics , Metabolism
16.
Chinese Journal of Cancer ; (12): 205-212, 2013.
Article in English | WPRIM | ID: wpr-295869

ABSTRACT

The discovery of induced pluripotent stem cells(iPSCs) is a promising advancement in the field of regenerative medicine. Previous studies have indicated that the teratoma-forming propensity of iPSCs is variable; however, the relationship between tumorigenic potential and genomic instability in human iPSCs (HiPSCs) remains to be fully elucidated. Here, we evaluated the malignant potential of HiPSCs by using both colony formation assays and tumorigenicity tests. We demonstrated that HiPSCs formed tumorigenic colonies when grown in cancer cell culture medium and produced malignancies in immunodeficient mice. Furthermore, we analyzed genomic instability in HiPSCs using whole-genome copy number variation analysis and determined that the extent of genomic instability was related with both the cells' propensity to form colonies and their potential for tumorigenesis. These findings indicate a risk for potential malignancy of HiPSCs derived from genomic instability and suggest that quality control tests, including comprehensive tumorigenicity assays and genomic integrity validation, should be rigorously executed before the clinical application of HiPSCs. In addition, HiPSCs should be generated through the use of combined factors or other approaches that decrease the likelihood of genomic instability.


Subject(s)
Animals , Humans , Mice , Carcinogenesis , Cells, Cultured , DNA Copy Number Variations , Genomic Instability , Induced Pluripotent Stem Cells , Cell Biology , Metabolism , Transplantation , Mice, SCID , NIH 3T3 Cells , Octamer Transcription Factor-3 , Metabolism , Teratocarcinoma , Teratoma , Tumor Stem Cell Assay
17.
Experimental & Molecular Medicine ; : e3-2013.
Article in English | WPRIM | ID: wpr-81324

ABSTRACT

MicroRNAs (miRNAs) participate in diverse biological functions and carcinogenesis by inhibiting specific gene expression. We previously reported that suppression of adenine nucleotide translocase 2 (ANT2) by using the short hairpin RNA (shRNA) approach has an antitumor effect in several cancer cells. We here examined the influence of ANT2 on expression of miRNAs in hepatocellular carcinoma (HCC) to further elucidate the tumor-suppressive mechanism of ANT2 shRNA. We first carried out screening for miRNAs, whose expression is regulated by ANT2 suppression in the Hep3B HCC cell line using miRNA microarrays. Validation of candidate miRNAs was done by incorporating clinical samples, and their effects on the tumorigenesis of HCC were studied in vitro and in vivo. miR-636 was one of the miRNAs whose expression was highly upregulated by ANT2 suppression in miRNA microarray analysis, as confirmed by real-time reverse transcription-polymerase chain reaction. Notably, miR-636 was markedly downregulated in HCC tissues compared with matched non-neoplastic liver in clinical samples. Restoration of miR-636 in Hep3B cells led to significant reduction of cell proliferation and colony formation. miR-636 restoration resulted in a decreased level of Ras, one of the putative targets of miR-636, and inactivation of its signaling pathway. Moreover, tumorigenesis was efficiently suppressed by miR-636 in an in vivo tumor xenograft model of HCC. The data suggest that miR-636 might function as a tumor suppressor miRNA affecting HCC tumorigenesis via downregulation of Ras, and that ANT2 suppression by shRNA could exert an anticancer effect by restoring miR-636 expression in HCC.


Subject(s)
Animals , Humans , Mice , Adenine Nucleotide Translocator 2/metabolism , Carcinoma, Hepatocellular/genetics , Cell Line, Tumor , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Down-Regulation/genetics , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Liver Neoplasms/genetics , Mice, Inbred BALB C , Mice, Nude , MicroRNAs/genetics , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , RNA, Small Interfering/metabolism , Signal Transduction/genetics , Transcription, Genetic , Tumor Stem Cell Assay , Up-Regulation/genetics , ras Proteins/genetics
18.
Asian Pacific Journal of Tropical Medicine ; (12): 645-650, 2012.
Article in English | WPRIM | ID: wpr-819603

ABSTRACT

OBJECTIVE@#To explore the role of proto-oncogene Pim-1 in the proliferation and migration of nasopharyngeal carcinoma (NPC) cells.@*METHODS@#Pim-1 expressions in NPC cell lines CNE1, CNE1-GL, CNE-2Z and C666-1 were examined by RT-PCR, western blotting and immunoflucesence, respectively. After CNE1, CNE1-GL and C666-1 cells were treated with different concentrations of Pim-1 special inhibitor, quercetagetin, the cell viability, colony formation rate and migration ability were analyzed.@*RESULTS@#Pim-1 expression was negative in well-differentiated CNE1 cells, whereas expressed weakly positive in poor-differentiated CNE-2Z cells and strongly positive in undifferentiated C666-1 cells. Interestingly, CNE1-GL cells that derived from CNE1 transfected with an Epstein Barr virus latent membrane protein-1 over-expression plasmid displayed stronger expression of Pim-1. Treatment of CNE1-GL and C666-1 cells with quercetagetin significantly decreased the cell viability, colony formation rate and migration ability but not the CNE1 cells.@*CONCLUSIONS@#These findings suggest that Pim-1 overexpression contributes to NPC proliferation and migration, and targeting Pim-1 may be a potential treatment for anti-Pim-1-expressed NPCs.


Subject(s)
Humans , Antineoplastic Agents , Pharmacology , Biomarkers, Tumor , Metabolism , Blotting, Western , Carcinoma , Cell Line, Tumor , Cell Movement , Cell Proliferation , Cell Survival , Chromones , Pharmacology , Flavones , Fluorescent Antibody Technique, Indirect , Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms , Metabolism , Pathology , Proto-Oncogene Proteins c-pim-1 , Metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tumor Stem Cell Assay
19.
São Paulo; s.n; s.n; 2012. 140 p. tab, graf, ilus.
Thesis in Portuguese | LILACS | ID: biblio-846867

ABSTRACT

Um passo limitante no desenvolvimento de fármacos para terapias do câncer está na descoberta de vulnerabilidades específicas de células tumorais que sirvam à identificação de alvos moleculares apropriados à intervenção farmacológica. Esta é a motivação central desta tese, cuja abordagem experimental focaliza a ação oncogênica das proteínas Ras. Amplificação ou mutação ativadora nos proto-oncogenes ras estão entre as alterações genéticas mais frequentes em cânceres. Essas lesões genéticas aparecem na origem etiológica de múltiplas formas de fenótipos malignos. Mas, essas lesões oncogênicas também conferem susceptibilidades letais às células malignamente transformadas frente a determinados agentes que não interferem significativamente nas funções vitais de células normais. Nos últimos anos nosso laboratório vem estudando os mecanismos moleculares da ação antiproliferativa do fator de crescimento FGF2 (Fibroblast Growth Factor2) e do éster de forbol PMA (Phorbol-12-Myristate-13-Acetate) em linhagens de células murinas malignas dependentes de ras oncogênico. Nesta tese investigamos quanto de nossas observações anteriores com células murinas são aplicáveis a células humanas. Nesse sentido focalizamos a linhagem HaCaT de queratinócitos humanos imortalizados e seus subclones malignizados por expressão ectópica de H-RasV12; além disso, numa triagem inicial também examinamos treze linhagens celulares humanas derivadas de tumores naturais portadores de mutação ativadora em H-Ras, N-Ras ou K-Ras. Nossos resultados mostram que os queratinócitos da linhagem parental HaCaT expressam receptores de FGFs e respondem mitogenicamente tanto a FGF2 como a PMA; portanto, ambos FGF2 e PMA são benéficos aos queratinócitos HaCaT. Por outro lado, o FGF2 mostrou-se citotóxico para subclones HaCaT que expressam H-RasV12 induzível, mas sublinhagens HaCaT com expressão constitutiva de H-RasV12 mostraram-se resistentes à ação citotóxica de FGF2. Diferentemente de FGF2, PMA bloqueou a proliferação de sublinhagens clonais HaCaT-H-RasV12 em ambos substrato sólido e suspensão de agarose e, também, reduziu a estratificação dos queratinócitos HaCaT-H-RasV12 em culturas organotípicas. PMA foi citotóxico e não citostático, pois induziu morte apoptótica sem causar arresto em nenhuma fase específica do ciclo celular. Em HaCaT parental, PMA induziu aumento transitório dos níveis intracelulares de espécies reativas de oxigênio (ROS), mas nos queratinócitos HaCaT-H-RasV12, PMA causou aumentos mais altos e persistentes de ROS, o que promove forte estresse oxidativo, provavelmente responsável pela toxidez deste ester de forbol. Entre as treze linhagens celulares humanas malignas com H-Ras, N-Ras ou K-Ras mutados, onze foram vulneráveis à ação citotóxica de PMA; mas apenas uma delas, a linhagem de tumor urotelial UM-UC-3, foi sensível ao efeito anti-proliferativo de FGF2. Em conclusão, células malignas humanas com Ras mutado parecem superar rapidamente uma possível toxidez de FGF2, mas não ultrapassam a toxidez causada por PMA


A challenge in drug development for cancer therapy is the discovering of molecular targets suitable for pharmacological interference. This challenge was the main motivation of the present thesis. Amplification or activating mutation in ras proto-oncogenes are among the most frequent genetic lesions in human cancer. Actually, mutated Ras onco-proteins are in the etiological roots of multiple malignant phenotypes; however these onco-proteins also cause specific lethal vulnerabilities even in robust malignant cells. Recently, our laboratory reported that malignant murine cell lines dependent on oncogenic Ras are prone to toxicity initiated by FGF2 (Fibroblast Growth Factor 2) and PMA (Phorbol-12-Myristate-13-Acetate), which are not harmful to normal cells. This cytotoxicity of FGF2 and PMA very likely follows different molecular mechanisms, which, however, are not yet completely understood. The aim of this thesis was to investigate whether these vulnerabilities found in murine malignant cells were also valid for human malignant cell lines dependent on oncogenic Ras. To this end the experimental approach was focused on the HaCaT cell line of immortalized human keratinocytes and its sublines transformed by H-RasV12 ectopic expression. In addition thirteen human cell lines derived from natural tumor carrying mutated H-Ras, N-Ras or K-Ras oncogenes were also screened. The results showed that HaCaT keratinocytes express FGF receptors and respond mitogenically to both FGF2 and PMA. On the other hand, FGF2 was cytotoxic to HaCaT subclones expressing inducible H-RasV12. But, HaCaT sublines constitutively expressing H-RasV12 were resistant to FGF2 toxicity. However, PMA was toxic to all HaCaT-H-RasV12 sublines, inhibiting proliferation in both solid substrate and agarose suspension cultures and, also reducing stratification in organotypic cultures. Furthermore, in HaCaT-H-RasV12 sublines, but not in the parental HaCaT line, PMA caused a persistently high increase in intracellular levels of reactive oxygen species (ROS) and concomitantly induced apoptosis. Moreover, eleven of the thirteen human tumor cell lines with mutated H-Ras, N-Ras or K-Ras, were growth inhibited by PMA, whereas only one of them was inhibited by FGF2, the urothelial tumor cell line UM-UC-3. In conclusion, human malignant cells driven by Ras oncogenes very likely rapidly overcome FGF2 toxicity, whereas they remain stably vulnerable to PMA cytotoxicity


Subject(s)
Carcinoma, Islet Cell , Health Vulnerability , Oxidative Stress/genetics , ras Proteins/analysis , Cytotoxins , Fibroblast Growth Factor 2/analysis , Flow Cytometry/methods , Fluorescent Antibody Technique/methods , Immunoprecipitation/statistics & numerical data , Neoplasms , Phorbol Esters/analysis , Tumor Cells, Cultured , Tumor Stem Cell Assay/instrumentation
20.
Biol. Res ; 45(3): 297-305, 2012. ilus
Article in English | LILACS | ID: lil-659287

ABSTRACT

Prostate cancer (PCa) is the most frequently diagnosed malignancy in men worldwide. Chemotherapy response is very poor and resistance to hormone-based treatments is frequent in advances stages. Recently, tumor-initiating cells or cancer stem cells (CSCs) have been identified in several cancers, including PCa. These cells are thought to be responsible for therapy resistance, relapse and metastasis. In the present work, enriched populations of CSCs were obtained using a mixed procedure that included differential clone-forming ability, sphere growing induction (prostatospheres) and magnetic-associated cell sorting (MACS). Also, stem marker expression was determined in PCa biopsies of different histological grades and metastasis samples. The signature for stem markers of the isolated CSCs was CD133+/CD44+/ABCG2+/ CD24-. Expression of stem markers (CD133, CD44, and ABCG2) was higher in medium Gleason biopsies than in lower and higher grades, and lymph-node and bone metastasis samples. These results suggest that the CSCs in PCa reach an important number in medium Gleason grades, when the tumor is still confined into the gland. At this stage, the surgical treatment is usually with curative intention. However, an important percentage of patients relapse after treatment. Number and signature of CSCs may be a prognosis factor for PCa recurrence.


Subject(s)
Humans , Male , Antigens, CD/analysis , Gene Expression Regulation, Neoplastic , Neoplastic Stem Cells/pathology , Prostatic Neoplasms/genetics , Biopsy , Bone Neoplasms/secondary , Cell Separation , Immunohistochemistry , Lymphatic Metastasis/pathology , Neoplasm Grading , Neoplasm Metastasis , Neoplasm Recurrence, Local , Prognosis , Prostatic Neoplasms/pathology , Tumor Stem Cell Assay , Biomarkers, Tumor/analysis
SELECTION OF CITATIONS
SEARCH DETAIL