Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
J Cell Biochem ; 123(10): 1663-1673, 2022 10.
Article in English | MEDLINE | ID: mdl-36271841

ABSTRACT

The multityrosine kinase inhibitor sorafenib remains an important systemic treatment option for hepatocellular carcinoma (HCC). Signaling pathways, which are targeted by sorafenib, are involved in checkpoint and DNA repair response, RAD51 being a candidate protein. Here, we aim to evaluate the effect of the human RAD51 inhibitor B02 in combination with sorafenib in human HCC cells. Impact of RAD51 expression on HCC patient survival was evaluated by an in silico approach using Human Protein Atlas dataset. Cell viability of HUH7, AKH12, AKH13, and 3P was assessed by neutral red assay. To measure the cytotoxicity, we quantified loss of membrane integrity by lactate dehydrogenase release. We also employed colony formation assay and hanging drop method to assess clonogenic and invasive ability of HCC cell lines upon sorafenib and B02 treatment. Cell cycle distribution and characterization of apoptosis was evaluated by flow cytometry. In silico approach revealed that HCC patients with higher expression of RAD51 messenger RNA had a significantly shorter overall survival. The RAD51 inhibitor B02 alone and in combination with sorafenib significantly reduced viability, colony formation ability, and invasion capacity of HCC cells. Cell cycle analysis revealed that the combination of both agents reduces the proportion of cells in the G2/M phase while leading to an accumulating in the subG1 phase. The RAD51 inhibitor B02 seems to be a promising agent for HCC treatment and enhances the antitumor effects of sorafenib in vitro.


Subject(s)
Antineoplastic Agents , Carcinoma, Hepatocellular , Liver Neoplasms , Humans , Sorafenib/pharmacology , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/drug therapy , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Cell Line, Tumor , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Apoptosis , DNA Repair , Cell Proliferation
2.
Int J Oncol ; 59(3)2021 Sep.
Article in English | MEDLINE | ID: mdl-34318902

ABSTRACT

The overexpression of chondroitin sulfate proteoglycan 4 (CSPG4) is associated with several tumor types, including malignant melanoma, squamous cell carcinoma, triple­negative breast carcinoma, oligodendrocytomas or gliomas. Due to its restricted distribution in normal tissues, CSPG4 has been considered a potential target for several antitumor approaches, including monoclonal antibody (mAb) therapies. The aim of the present study was to characterize the impact of the CSPG4­specific mAb clone 9.2.27 on its own or in combination with the commonly used BRAF­selective inhibitor, PLX4032, on different functions of melanoma cells to assess the potential synergistic effects. The BRAF V600­mutant human melanoma cell lines, M14 (CSPG4­negative) and WM164 (CSPG4­positive), were exposed to the CSPG4­specific 9.2.27 mAb and/or PLX4032. Cell viability and colony formation capacity were evaluated. A 3D­cell culture spheroid model was used to assess the invasive properties of the treated cells. In addition, flow cytometric analysis of apoptosis and cell cycle analyses were performed. Incubation of the WM164 cell line with CSPG4­specific 9.2.27 mAb decreased viability, colony formation ability and the invasive capacity of CSPG4­positive tumor cells, which was not the case for the CSPG4­negative M14 cell line. Combined treatment of the WM164 cells with 9.2.27 mAb plus PLX4032 did not exert any significant additional effect in comparison to treatment with PLX4032 alone in the clonogenic and invasion assays. M14 cell cycle distribution was not influenced by the CSPG4­specific 9.2.27 mAb. By contrast, the exposure of WM164 cells to the mAb resulted in an arrest of the cells in the S phase. Moreover, combined treatment of the WM164 cells led to a significantly increased accumulation of cells in the subG1 phase, combined with a decrease of cells in the G2/M phase. On the whole, findings of the present study indicate that the CSPG4­specific 9.2.27 mAb exerts an anti­invasive effect on CSPG4­positive melanoma spheroids, which is not enhanced by BRAF inhibition. These findings provide the basis for further investigations on the effects of anti­CSPG4­based treatments of CSPG4­positive tumors.


Subject(s)
Antibodies, Monoclonal/pharmacology , Chondroitin Sulfate Proteoglycans/metabolism , Melanoma/metabolism , Membrane Proteins/metabolism , Spheroids, Cellular/cytology , Vemurafenib/pharmacology , Cell Cycle/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Survival/drug effects , Drug Synergism , Gene Expression Regulation, Neoplastic/drug effects , Humans , Melanoma/drug therapy , Melanoma/genetics , Mutation , Proto-Oncogene Proteins B-raf/genetics , Spheroids, Cellular/drug effects , Spheroids, Cellular/metabolism , Tumor Cells, Cultured
3.
Materials (Basel) ; 14(9)2021 Apr 22.
Article in English | MEDLINE | ID: mdl-33922317

ABSTRACT

N-Heterocycles are considered as desirable scaffolds for the development of novel lead compounds for anticancer drug research. Among them, phosphorus-containing amino-derivatives play a crucial role. A series of imines and products of their further reactions with P-nucleophiles were obtained starting from vicinal bisamines. Reaction of ethylenediamine and α-carbonyl esters yielded in novel unexpected products, which structures were confirmed by crystallographic measurements. The cytotoxic activity evaluation was done on a variety of cell lines including HUH7, AKH12, DAOY, UW228-2, D283, D425, and U251. Human umbilical vein endothelial cells (HUVECs) were used as control. Two of the tested compounds, bearing TADDOL-derived, and trifluoromethyl substituents showed a significant effect on cell viability, though comparable to nonmalignant cells.

4.
Molecules ; 25(10)2020 May 18.
Article in English | MEDLINE | ID: mdl-32443610

ABSTRACT

A series of chiral sulfonamides containing the 2-azabicycloalkane scaffold were prepared from aza-Diels-Alder cycloadducts through their conversion to amines based on 2-azanorbornane or the bridged azepane skeleton, followed by the reaction with sulfonyl chlorides. The cytotoxic activity of the obtained bicyclic derivatives was evaluated using human hepatocellular carcinoma (HCC), medulloblastoma (MB), and glioblastoma (GBM) cell lines. Chosen compounds were shown to notably reduce cell viability as compared to nonmalignant cells.


Subject(s)
Alkanes/chemistry , Carcinoma, Hepatocellular/drug therapy , Liver Neoplasms/drug therapy , Sulfonamides/chemistry , Alkanes/chemical synthesis , Alkanes/pharmacology , Amines/chemical synthesis , Amines/chemistry , Amines/pharmacology , Carcinoma, Hepatocellular/pathology , Humans , Liver Neoplasms/pathology , Molecular Structure , Stereoisomerism , Sulfonamides/chemical synthesis , Sulfonamides/pharmacology
5.
Cell Death Dis ; 10(10): 696, 2019 09 20.
Article in English | MEDLINE | ID: mdl-31541075

ABSTRACT

Medulloblastoma, an embryonal tumor of the cerebellum/fourth ventricle, is one of the most frequent malignant brain tumors in children. Although genetic variants are increasingly used in treatment stratification, survival of high-risk patients, characterized by leptomeningeal dissemination, TP53 mutation or MYC amplification, is still poor. FOXM1, a proliferation-specific oncogenic transcription factor, is deregulated in various solid tumors, including medulloblastoma, and triggers cellular proliferation, migration and genomic instability. In tissue samples obtained from medulloblastoma patients, the significant upregulation of FOXM1 was associated with a loss of its putative regulating microRNA, miR-4521. To understand the underlying mechanism, we investigated the effect of miR-4521 on the expression of the transcription factor FOXM1 in medulloblastoma cell lines. Transfection of this microRNA reduced proliferation and invasion of several medulloblastoma cell lines and induced programmed cell death through activation of caspase 3/7. Further, downstream targets of FOXM1 such as PLK1 and cyclin B1 were significantly reduced thus affecting the cell cycle progression in medulloblastoma cell lines. In conclusion, a restoration of miRNA-4521 may selectively suppress the pathophysiological effect of aberrant FOXM1 expression and serve as a targeted approach for medulloblastoma therapy.


Subject(s)
Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Forkhead Box Protein M1/biosynthesis , Medulloblastoma/genetics , Medulloblastoma/metabolism , MicroRNAs/metabolism , 3' Untranslated Regions , Adolescent , Brain Neoplasms/pathology , Cell Cycle Checkpoints/physiology , Cell Line, Tumor , Cell Proliferation/physiology , Child , Child, Preschool , Down-Regulation , Female , Forkhead Box Protein M1/genetics , Forkhead Box Protein M1/metabolism , Humans , Infant , Male , Medulloblastoma/pathology , MicroRNAs/biosynthesis , MicroRNAs/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Spheroids, Cellular , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...