Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.878
Filter
1.
Med Oncol ; 40(7): 196, 2023 Jun 07.
Article in English | MEDLINE | ID: mdl-37284891

ABSTRACT

Anaplastic thyroid cancer (ATC) represents the type with the worst prognosis among thyroid cancers. In ATC with a highly invasive phenotype, selective targeting of TERT with BIBR1532 may be a goal-driven approach to preserving healthy tissues. In present study, it was aimed to investigate the effects of treatment of SW1736 cells with BIBR1532 on apoptosis, cell cycle progression, and migration. The apoptotic effect of BIBR1532 on SW1736 cells was examined using the Annexin V method, the cytostatic effect using cell cycle test, migration properties using wound healing assay. Gene expression differences were determined by real-time qRT-PCR and differences in protein level by ELISA test. BIBR1532-treated SW1736 cells had 3.1-fold increase in apoptosis compared to their untreated counterpart. There was 58.1% arrest in the G0/G1 phase and 27.6% arrest in the S phase of the cell cycle in untreated group, treatment with BIBR1532 increased cell population in G0/G1 phase to 80.9% and decreased in S phase to 7.1%. Treatment with the TERT inhibitor resulted in a 50.8% decrease in cell migration compared to the untreated group. After BIBR1532 treatment of SW1736 cells, upregulation of BAD, BAX, CASP8, CYCS, TNFSF10, CDKN2A genes, and downregulation of BCL2L11, XIAP, CCND2 genes were detected. BIBR1532 treatment resulted in an increase in BAX and p16 proteins, and a decrease in concentration of BCL-2 protein compared to untreated group. Targeting TERT with BIBR1532 as a mono drug or using of BIBR1532 at "priming stage" prior to chemotherapy treatment in ATC may present a novel and promising treatment strategy.


Subject(s)
Antineoplastic Agents , Apoptosis , Cell Cycle , Cell Movement , Enzyme Inhibitors , Telomerase , Thyroid Carcinoma, Anaplastic , Thyroid Neoplasms , Humans , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Apoptosis/genetics , Cell Cycle/drug effects , Cell Cycle/genetics , Cell Line, Tumor , Cell Movement/drug effects , Enzyme Inhibitors/pharmacology , Telomerase/antagonists & inhibitors , Thyroid Carcinoma, Anaplastic/drug therapy , Thyroid Carcinoma, Anaplastic/genetics , Thyroid Carcinoma, Anaplastic/pathology , Thyroid Neoplasms/drug therapy , Thyroid Neoplasms/genetics , Thyroid Neoplasms/pathology , Cell Cycle Checkpoints/drug effects , Cell Cycle Checkpoints/genetics , Resting Phase, Cell Cycle/drug effects , G1 Phase/drug effects , Gene Expression Regulation, Neoplastic/drug effects
2.
Sci Rep ; 12(1): 3093, 2022 02 23.
Article in English | MEDLINE | ID: mdl-35197531

ABSTRACT

Development of chemotherapy has led to a high survival rate of cancer patients; however, the severe side effects of anticancer drugs, including organ hypoplasia, persist. To assume the side effect of anticancer drugs, we established a new ex vivo screening model and described a method for suppressing side effects. Cyclophosphamide (CPA) is a commonly used anticancer drug and causes severe side effects in developing organs with intensive proliferation, including the teeth and hair. Using the organ culture model, we found that treatment with CPA disturbed the growth of tooth germs by inducing DNA damage, apoptosis and suppressing cellular proliferation and differentiation. Furthermore, low temperature suppressed CPA-mediated inhibition of organ development. Our ex vivo and in vitro analysis revealed that low temperature impeded Rb phosphorylation and caused cell cycle arrest at the G1 phase during CPA treatment. This can prevent the CPA-mediated cell damage of DNA replication caused by the cross-linking reaction of CPA. Our findings suggest that the side effects of anticancer drugs on organ development can be avoided by maintaining the internal environment under low temperature.


Subject(s)
Antineoplastic Agents/adverse effects , Cyclophosphamide/adverse effects , Temperature , Cell Cycle Checkpoints/drug effects , Cell Proliferation/drug effects , DNA Damage/drug effects , DNA Replication/drug effects , G1 Phase/drug effects , Humans , Models, Biological , Organ Culture Techniques
3.
BMC Plant Biol ; 22(1): 46, 2022 Jan 22.
Article in English | MEDLINE | ID: mdl-35065609

ABSTRACT

BACKGROUND: Land plants respond to drought and salinity by employing multitude of sophisticated mechanisms with physiological and developmental consequences. Abscisic acid-mediated signaling pathways have evolved as land plant ancestors explored their habitats toward terrestrial dry area, and now play major roles in hyperosmotic stress responses in flowering plants. Green algae living in fresh water habitat do not possess abscisic acid signaling pathways but need to cope with increasing salt concentrations or high osmolarity when challenged with adverse aquatic environment. Hyperosmotic stress responses in green algae are largely unexplored. RESULTS: In this study, we characterized hyperosmotic stress-induced cytoskeletal responses in Chlamydomonas reinhardtii, a fresh water green algae. The Chlamydomonas PROPYZAMIDE-HYPERSENSITEVE 1 (PHS1) tubulin kinase quickly and transiently phosphorylated a large proportion of cellular α-tubulin at Thr349 in G1 phase and during mitosis, which resulted in transient disassembly of microtubules, when challenged with > 0.2 M sorbitol or > 0.1 M NaCl. By using phs1 loss-of-function algal mutant cells, we demonstrated that transient microtubule destabilization by sorbitol did not affect cell growth in G1 phase but delayed mitotic cell cycle progression. Genome sequence analyses indicate that PHS1 genes evolved in ancestors of the Chlorophyta. Interestingly, PHS1 genes are present in all sequenced genomes of freshwater Chlorophyta green algae (including Chlamydomonas) but are absent in some marine algae of this phylum. CONCLUSION: PHS1-mediated tubulin phosphorylation was found to be partly responsible for the efficient stress-responsive mitotic delay in Chlamydomonas cells. Ancient hyperosmotic stress-triggered cytoskeletal remodeling responses thus emerged when the PHS1 tubulin kinase gene evolved in freshwater green algae.


Subject(s)
Chlamydomonas reinhardtii/physiology , Microtubules/metabolism , Osmotic Pressure/physiology , Plant Proteins/metabolism , Tubulin/metabolism , Cell Culture Techniques/methods , Cell Division , Chlamydomonas reinhardtii/cytology , Chlamydomonas reinhardtii/drug effects , Chlorophyta/genetics , G1 Phase/drug effects , Mitosis/drug effects , Phosphorylation , Plant Proteins/genetics , Protein Kinases/genetics , Protein Kinases/metabolism , Salt Stress , Sorbitol/pharmacology , Threonine
4.
J Inorg Biochem ; 226: 111663, 2022 01.
Article in English | MEDLINE | ID: mdl-34801972

ABSTRACT

Three neutral Pt(II) complexes containing 1-Methylimidazole and the antifungal imidazolyl drugs Clotrimazole and Bifonazole have been prepared. The general formula of the new derivatives is [Pt(κ2-(C^N)Cl(L)], where C^N stands for ppy = 2-phenylpyridinate, and L = 1-Methylimidazole (MeIm) for [Pt-MeIm]; L = Clotrimazole (CTZ) for [Pt-CTZ] and L = Bifonazole (BFZ) for [Pt-BFZ]). The complexes have been completely characterized in solution and the crystal structures of [Pt-BFZ] and [Pt-CTZ] have been resolved. Complexes [Pt-MeIm] and [Pt-BFZ] present higher cytotoxicity than cisplatin in SW480 (colon adenocarcinoma), A549 (lung adenocarcinoma) and A2780 (ovarian cancer) cell lines. [Pt-MeIm] shows the highest accumulation in A549 cells, in agreement with its inability to interact with serum albumin. By contrast, [Pt-CTZ] and [Pt-BFZ] interact with serum proteins, a fact that reduces their bioavailability. The strongest interaction with bovine serum albumin (BSA) is found for [Pt-BFZ], which is the least internalized inside the cells. All the complexes are able to covalently interact with DNA. The most cytotoxic complexes, [Pt-MeIm] and [Pt-BFZ] induce cellular accumulation in G0/G1 and apoptosis by a similar pathway, probably involving a reactive oxygen species (ROS) generation mechanism. [Pt-BFZ] turns out to be the most efficient complex regarding ROS generation and causes mitochondrial membrane depolarization, whereas [Pt-MeIm] induces the opposite effect, hyperpolarization of the mitochondrial membrane. On the contrary, the least cytotoxic complex, [Pt-CTZ] cannot block the cell cycle or generate ROS and the mechanism by which it induces apoptosis could be a different one.


Subject(s)
Antifungal Agents , Antineoplastic Agents , Coordination Complexes , Neoplasms , Platinum , A549 Cells , Antifungal Agents/chemical synthesis , Antifungal Agents/chemistry , Antifungal Agents/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Coordination Complexes/chemical synthesis , Coordination Complexes/chemistry , Coordination Complexes/pharmacology , G1 Phase/drug effects , Humans , Neoplasms/drug therapy , Neoplasms/metabolism , Platinum/chemistry , Platinum/pharmacology , Resting Phase, Cell Cycle/drug effects
5.
Bioengineered ; 12(2): 12812-12820, 2021 12.
Article in English | MEDLINE | ID: mdl-34895043

ABSTRACT

Osteoarthritis (OA) is a cartilage degenerative disease commonly observed in the elderly population and significantly impacts the normal life of OA patients. It has been reported that the development of pathological cell senescence in chondrocytes is involved in the pathogenesis of OA. Celecoxib is a common non-steroidal anti-inflammatory drug, and it has been recently reported to exert therapeutic effects on OA. However, its underlying mechanism is still unclear. The present study intends to explore its mechanism and provide fundamental evidence for the application of Celecoxib in the treatment of clinical OA. Tumor necrosis factor-α (TNF-α) was utilized to establish an in vitro model of chondrocytes senescence. The elevated reactive oxygen species (ROS) generation, increased cell cycle arrest in G0/G1 phase, reduced telomerase activity, and upregulated senescence-associatedß-galactosidase (SA-ß-Gal) staining were all observed in TNF-α-treated chondrocytes, which were then dramatically reversed by 10 and 20 µM Celecoxib. In addition, the upregulated DNA damage biomarkers, p-ATM, and p-CHK2, observed in TNF-α-treated chondrocytes were significantly downregulated by 10 and 20 µM Celecoxib. Lastly, the expression level of p21 and p53 was greatly elevated in chondrocytes by stimulation with TNF-α which was then pronouncedly repressed by treatment with Celecoxib. Taken together, our data reveal that Celecoxib ameliorated TNF-α-induced cellular senescence in human chondrocytes.


Subject(s)
Celecoxib/pharmacology , Cellular Senescence/drug effects , Chondrocytes/pathology , Tumor Necrosis Factor-alpha/toxicity , Ataxia Telangiectasia Mutated Proteins/metabolism , Celecoxib/chemistry , Cell Cycle Checkpoints/drug effects , Cell Death/drug effects , Cell Line , Checkpoint Kinase 2/metabolism , Chondrocytes/drug effects , Chondrocytes/metabolism , Cyclin-Dependent Kinase Inhibitor p21/metabolism , G1 Phase/drug effects , Humans , Phosphorylation/drug effects , Reactive Oxygen Species/metabolism , Resting Phase, Cell Cycle/drug effects , Telomerase/metabolism , Tumor Suppressor Protein p53/metabolism
6.
Mol Cell ; 81(24): 5007-5024.e9, 2021 12 16.
Article in English | MEDLINE | ID: mdl-34767771

ABSTRACT

As cells enter mitosis, chromatin compacts to facilitate chromosome segregation yet remains transcribed. Transcription supercoils DNA to levels that can impede further progression of RNA polymerase II (RNAPII) unless it is removed by DNA topoisomerase 1 (TOP1). Using ChIP-seq on mitotic cells, we found that TOP1 is required for RNAPII translocation along genes. The stimulation of TOP1 activity by RNAPII during elongation allowed RNAPII clearance from genes in prometaphase and enabled chromosomal segregation. Disruption of the TOP1-RNAPII interaction impaired RNAPII spiking at promoters and triggered defects in the post-mitotic transcription program. This program includes factors necessary for cell growth, and cells with impaired TOP1-RNAPII interaction are more sensitive to inhibitors of mTOR signaling. We conclude that TOP1 is necessary for assisting transcription during mitosis with consequences for growth and gene expression long after mitosis is completed. In this sense, TOP1 ensures that cellular memory is preserved in subsequent generations.


Subject(s)
Cell Proliferation , Chromatin Assembly and Disassembly , Colorectal Neoplasms/enzymology , DNA Topoisomerases, Type I/metabolism , G1 Phase , Mitosis , RNA Polymerase II/metabolism , Transcription, Genetic , Cell Proliferation/drug effects , Chromatin Immunoprecipitation Sequencing , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , DNA Topoisomerases, Type I/genetics , G1 Phase/drug effects , Gene Expression Regulation, Neoplastic , HCT116 Cells , Humans , MTOR Inhibitors/pharmacology , Mitosis/drug effects , RNA Polymerase II/genetics
7.
Int J Mol Sci ; 22(21)2021 Oct 27.
Article in English | MEDLINE | ID: mdl-34769053

ABSTRACT

The potent splicing inhibitor spliceostatin A (SSA) inhibits cell cycle progression at the G1 and G2/M phases. We previously reported that upregulation of the p27 cyclin-dependent kinase inhibitor encoded by CDKN1B and its C-terminal truncated form, namely p27*, which is translated from CDKN1B pre-mRNA, is one of the causes of G1 phase arrest caused by SSA treatment. However, the detailed molecular mechanism underlying G1 phase arrest caused by SSA treatment remains to be elucidated. In this study, we found that SSA treatment caused the downregulation of cell cycle regulators, including CCNE1, CCNE2, and E2F1, at both the mRNA and protein levels. We also found that transcription elongation of the genes was deficient in SSA-treated cells. The overexpression of CCNE1 and E2F1 in combination with CDKN1B knockout partially suppressed G1 phase arrest caused by SSA treatment. These results suggest that the downregulation of CCNE1 and E2F1 contribute to the G1 phase arrest induced by SSA treatment, although they do not exclude the involvement of other factors in SSA-induced G1 phase arrest.


Subject(s)
Cell Cycle Checkpoints/drug effects , Cyclin E/genetics , E2F1 Transcription Factor/genetics , G1 Phase/drug effects , Oncogene Proteins/genetics , Pyrans/pharmacology , Spiro Compounds/pharmacology , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p27/genetics , Down-Regulation/drug effects , HeLa Cells , Humans , RNA, Messenger/genetics
8.
Anticancer Res ; 41(11): 5461-5468, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34732415

ABSTRACT

BACKGROUND/AIM: This study aimed to assess the effects of telmisartan (TEL), a potential antitumor agent, and its mechanism of action in the regulation of apoptosis, autophagy, and cell cycle in scirrhous gastric cancer (SGC). MATERIALS AND METHODS: The effect of TEL on the viability and chromatin condensation of OCUM-2M and OCUM-12 cells was assessed. Protein expression and the cell cycle were analysed using western blotting and flow cytometry, respectively. RESULTS: TEL inhibited cell proliferation in a dose-dependent manner and increased chromatin condensation and autophagy marker LC3-II levels in OCUM-12 cells. TEL also increased the proportion of cells in the G0/G1 phase transition. CONCLUSION: Apoptosis and autophagy are partially involved in the inhibitory effect of TEL on cell proliferation. Additionally, TEL caused G0/G1 cell cycle arrest. Therefore, TEL could be a promising treatment for SGC.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , G1 Phase Cell Cycle Checkpoints/drug effects , G1 Phase/drug effects , Resting Phase, Cell Cycle/drug effects , Stomach Neoplasms/drug therapy , Telmisartan/pharmacology , Apoptosis/drug effects , Autophagy/drug effects , Cell Line, Tumor , Humans , Microtubule-Associated Proteins/metabolism , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology
9.
PLoS One ; 16(9): e0257984, 2021.
Article in English | MEDLINE | ID: mdl-34570813

ABSTRACT

Prostate cancer is the second leading cause of cancer related death in American men. Several therapies have been developed to treat advanced prostate cancer, but these therapies often have severe side effects. To improve the outcome with fewer side effects we focused on the furanocoumarin bergamottin, a natural product found in grapefruit juice and a potent CYP3A inhibitor. Our recent studies have shown that CYP3A5 inhibition can block androgen receptor (AR) signaling, critical for prostate cancer growth. We observed that bergamottin reduces prostate cancer (PC) cell growth by decreasing both total and nuclear AR (AR activation) reducing downstream AR signaling. Bergamottin's role in reducing AR activation was confirmed by confocal microscopy studies and reduction in prostate specific antigen (PSA) levels, which is a marker for prostate cancer. Further studies revealed that bergamottin promotes cell cycle block and accumulates G0/G1 cells. The cell cycle block was accompanied with reduction in cyclin D, cyclin B, CDK4, P-cdc2 (Y15) and P-wee1 (S642). We also observed that bergamottin triggers apoptosis in prostate cancer cell lines as evident by TUNEL staining and PARP cleavage. Our data suggests that bergamottin may suppress prostate cancer growth, especially in African American (AA) patients carrying wild type CYP3A5 often presenting aggressive disease.


Subject(s)
Androgen Receptor Antagonists/therapeutic use , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Cytochrome P-450 CYP3A Inhibitors/therapeutic use , Furocoumarins/therapeutic use , G1 Phase/drug effects , Prostatic Neoplasms/drug therapy , Resting Phase, Cell Cycle/drug effects , Blotting, Western , Cell Fractionation , Cell Line, Tumor , Citrus paradisi/chemistry , Down-Regulation , Fruit and Vegetable Juices/analysis , Humans , Male , Microscopy, Confocal , Receptors, Androgen/drug effects
10.
Cytokine ; 148: 155700, 2021 12.
Article in English | MEDLINE | ID: mdl-34560609

ABSTRACT

Transforming growth factor (TGFß) is known to play a major role in establishment and maintenance of endometriosis as reported by our group earlier, the underlying mechanism remains to be explored. We deciphered the involvement of TAK1 in TGFß1- induced cellular responses and delineated the signaling mechanism in human endometriotic cells. The endometriotic cells showed elevated expression of TGFß1 signaling-effector molecules. TGFß1 exposure to endometriotic cells induced the expression of the downstream target molecules indicating that TGFß1 is implicated in the commencement ofTAK1/NFκB-p65/Smad7 cascade. The silencing of TAK1 in endometriotic cells attenuated the TGFß1 -induced NFκB transcriptional activation and nuclear translocation of NFκB-p65 subunit. The pharmacological inhibition of NFκB by QNZ or knockdown of TAK1 reduced the expression of Smad7 and Cox2. The knockdown of TAK1 in endometriotic cells showed G1 phase cell-cycle arrest and showed low BrdU-incorporation in the presence of TGFß1. The inhibition of TAK1 attenuated the TGFß1 signaling activation indicating that TAK1 is a crucial mediator for TGFß1 action in endometriotic cells. The exposure of endometriotic cells to TAK1 inhibitor, celastrol caused activation of caspase-3 and -9 that led to PARP cleavage and induced apoptosis. Simultaneously, autophagy occurred in celastrol-treated and TAK1-silenced cells as was evidenced by the formation of autophagosome and the increased expression of autophagic markers. Thus, TAK1 activation appears to protect the growth of endometriotic cells by suppressing the cell death process. Overall, our study provided the evidence that of TAK1 significant in the endometriotic cell regulation and mediates a functional cross-talk between TGFß1 and NFκB-p65 that promotes the growth and inflammatory response in endometriotic cells.


Subject(s)
Autophagy , Endometriosis/metabolism , Endometriosis/pathology , MAP Kinase Kinase Kinases/antagonists & inhibitors , NF-kappa B/metabolism , Signal Transduction , Smad7 Protein/metabolism , Transforming Growth Factor beta1/metabolism , Apoptosis/drug effects , Autophagy/drug effects , Cell Cycle Checkpoints/drug effects , Cell Death/drug effects , Cell Line , Cell Proliferation/drug effects , Cells, Cultured , Cyclooxygenase 2/metabolism , Endometrium/pathology , Female , G1 Phase/drug effects , Gene Knockdown Techniques , Humans , Models, Biological , Pentacyclic Triterpenes/pharmacology , Signal Transduction/drug effects , Stromal Cells/drug effects , Stromal Cells/metabolism
11.
Int J Biol Sci ; 17(12): 3224-3238, 2021.
Article in English | MEDLINE | ID: mdl-34421361

ABSTRACT

Mechanisms of breast cancer progression and invasion, often involve alteration of hormonal signaling, and upregulation and/or activation of signal transduction pathways that input to cell cycle regulation. Herein, we describe a rationally designed first-in-class novel small molecule inhibitor for targeting oncogenic and hormonal signaling in ER-positive breast cancer. BC-N102 treatment exhibits dose-dependent cytotoxic effects against ER+ breast cancer cell lines. BC-N102 exhibited time course- and dose-dependent cell cycle arrest via downregulation of the estrogen receptor (ER), progesterone receptor (PR), androgen receptor (AR), phosphatidylinositol 3-kinase (PI3K), phosphorylated (p)-extracellular signal-regulated kinase (ERK), p-Akt, CDK2, and CDK4 while increasing p38 mitogen-activated protein kinase (MAPK), and mineralocorticoid receptor (MR) signaling in breast cancer cell line. In addition, we found that BC-N102 suppressed breast cancer tumorigenesis in vivo and prolonged the survival of animals. Our results suggest that the proper application of BC-N102 may be a beneficial chemotherapeutic strategy for ER+ breast cancer patients.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Cell Cycle Checkpoints/drug effects , Cell Cycle Proteins/metabolism , G1 Phase/drug effects , Receptors, Estrogen/metabolism , Resting Phase, Cell Cycle/drug effects , Animals , Biomarkers, Tumor/genetics , Blotting, Western , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Division , Cell Line, Tumor , Cyclin-Dependent Kinase 2/genetics , Cyclin-Dependent Kinase 4/genetics , Female , Flow Cytometry , Gene Expression Regulation, Neoplastic/physiology , Humans , Maximum Tolerated Dose , Mice , Mice, Nude , Xenograft Model Antitumor Assays
12.
Biomed Pharmacother ; 139: 111663, 2021 Jul.
Article in English | MEDLINE | ID: mdl-34243605

ABSTRACT

Isolongifolanone is a high value-added sustainable natural product. Recent studies have demonstrated that isolongifolanone possesses anticancer activities. In this study, a series of novel pyrazole ring-containing isolongifolanone derivatives was designed, synthesized, and their anti-proliferative activities in three cancer cell lines were evaluated. Among them, compound 3b exhibited strongest antiproliferative ability on MCF-7 cancer cells and induced the generation of intracellular ROS and mitochondrial depolarization. More importantly, compound 3b still maintained antitumor activity in MCF-7 3D culture systems. The study on molecular mechanism suggested that compound 3b induced apoptosis via activation of caspase-3 and PARP, also via decreasing of Bcl-2 and increasing of Bax and p53. Moreover, compound 3b down-regulated the level of CDK2, a crucial cyclin-dependent kinase which is necessary for the progression of the cells out of the G1 phase of the cell cycle. Docking results showed that compound 3b could bind well with CDK2 by forming hydrogen bonds with amino acid residues (LYS89 and HIS84). These results suggested that compound 3b could be taken as a lead compound for anticancer agents.


Subject(s)
Antineoplastic Agents/pharmacology , Cyclin-Dependent Kinase 2/metabolism , Pyrazoles/pharmacology , Sesquiterpenes/pharmacology , A549 Cells , Amino Acids/metabolism , Apoptosis/drug effects , Caspase 3/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Down-Regulation/drug effects , G1 Phase/drug effects , HeLa Cells , Humans , MCF-7 Cells , Mitochondria/drug effects , Mitochondria/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Tumor Suppressor Protein p53/metabolism
13.
Mol Med Rep ; 23(4)2021 04.
Article in English | MEDLINE | ID: mdl-34223631

ABSTRACT

Acute myelogenous leukemia (AML) is frequently accompanied by a poor prognosis. The majority of patients with AML will experience recurrence due to multiple drug resistance. Our previous study reported that targeting the mTOR pathway may increase cell sensitivity to doxorubicin (Doxo) and provide an improved therapeutic approach to leukemia. However, the effect and mechanism of action of NVP­BEZ235 (BEZ235), a dual inhibitor of PI3K/mTOR, on Doxo­resistant K562 cells (K562/A) is yet to be elucidated. Therefore, the aim of the present study was to investigate the effects of BEZ235 on K562/A cell proliferation. K562/A cells was investigated using CCK­8, flow cytometry and western blotting, following BEZ235 treatment. It was observed that BEZ235 significantly decreased the viability of K562/A cells. In addition, BEZ235 arrested K562/A cells at the G0/G1 phase, and reduced the protein expression levels of CDK4, CDK6 and cyclin D1. Apoptotic cells were more frequently detected in K562/A cells treated with BEZ235 compared with the control group (12.97±0.91% vs. 7.37±0.42%, respectively; P<0.05). Cells treated with BEZ235 exhibited downregulation of Bcl­2 and upregulation of Bax. Furthermore, BEZ235 treatment markedly decreased the activation of the PI3K/AKT/mTOR pathway and its downstream effectors. Thus, these results demonstrated that BEZ235 inhibited cell viability, induced G0/G1 arrest and increased apoptosis in K562/A cells, suggesting that BEZ235 may reverse Doxo resistance in leukemia cells. Therefore, targeting the PI3K/mTOR pathway may be of value as a novel therapeutic approach to leukemia.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Drug Resistance, Neoplasm/drug effects , Imidazoles/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors/pharmacology , Quinolines/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Apoptosis/drug effects , Cell Survival/drug effects , Down-Regulation/drug effects , Doxorubicin/pharmacology , G1 Phase/drug effects , Humans , K562 Cells , Phosphatidylinositol 3-Kinases/genetics , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Resting Phase, Cell Cycle/drug effects , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism
14.
Biomed Res Int ; 2021: 9981815, 2021.
Article in English | MEDLINE | ID: mdl-34307685

ABSTRACT

BACKGROUND: Colorectal cancer (CRC) is among the top three gastrointestinal malignancy in morbidity and mortality. The abnormal activation of Wnt/ß-catenin pathway is considered to be a key factor in the occurrence and development of CRC. Novel inhibitor discovery against key factor in WNT pathway is important for CRC treatment and prevention. METHODS: Cell proliferation was detected after hydroxyphenyl butanone treatment in human colorectal cancer HCT116, LOVO, and normal colonic epithelial NCM460 cells. Colony formation, cell invasion ability, and cell cycle were detected with and without GSK-3ß knockdown. RESULTS: Hydroxyphenyl butanone induces cycle arresting on G1-S phase of colorectal cancer cell line through GSK3ß in Wnt/ß-catenin pathway and inhibits malignant biological manifestations of cell proliferation, colony formation, and invasion. The inhibition in the high concentration group is stronger than that in the low concentration group, and the antitumor effect is different for different tumor cells. Under the same concentration of natural hydroxyphenyl butanone, the inhibition on normal colonic epithelial cells is significantly lower than that on tumor cells. The natural hydroxyphenyl butanone with medium and low concentration could promote the proliferation of normal colonic epithelial cells. CONCLUSION: This study illustrated natural hydroxyphenyl butanone as new inhibitor of GSK3ß and revealed the mechanisms underlying the inhibitory effects in colorectal cancer.


Subject(s)
Butanones/pharmacology , Cell Cycle Checkpoints/drug effects , Colorectal Neoplasms/enzymology , Glycogen Synthase Kinase 3 beta/antagonists & inhibitors , Cell Line, Tumor , Cell Proliferation/drug effects , G1 Phase/drug effects , Glycogen Synthase Kinase 3 beta/metabolism , Humans , Neoplasm Invasiveness , Plant Extracts/pharmacology , Rubus/chemistry , S Phase/drug effects , Tumor Stem Cell Assay , Wnt Signaling Pathway/drug effects
15.
Cancer Lett ; 519: 304-314, 2021 10 28.
Article in English | MEDLINE | ID: mdl-34329742

ABSTRACT

ß-Lapachone is a classic quinone-containing antitumor NQO1-bioactivatable drug that directly kills NQO1-overexpressing cancer cells. However, the clinical applications of ß-lapachone are primarily limited by its high toxicity and modest lethality. To overcome this side effect and expand the therapeutic utility of ß-lapachone, we demonstrate the effects of a novel combination therapy including ß-lapachone and the proliferating cell nuclear antigen (PCNA) inhibitor T2 amino alcohol (T2AA) on various NQO1+ cancer cells. PCNA has DNA clamp processivity activity mediated by encircling double-stranded DNA to recruit proteins involved in DNA replication and DNA repair. In this study, we found that compared to monotherapy, a nontoxic dose of the T2AA synergized with a sublethal dose of ß-lapachone in an NQO1-dependent manner and that combination therapy prevented DNA repair, increased double-strand break (DSB) formation and promoted programmed necrosis and G1 phase cell cycle arrest. We further determined that combination therapy enhanced antitumor efficacy and prolonged survival in Lewis lung carcinoma (LLC) xenografts model. Our findings show novel evidence for a new therapeutic approach that combines of ß-lapachone treatment with PCNA inhibition that is highly effective in treating NQO1+ solid tumor cells.


Subject(s)
Antineoplastic Agents/pharmacology , DNA Damage/drug effects , NAD(P)H Dehydrogenase (Quinone)/metabolism , Naphthoquinones/pharmacology , Neoplasms/drug therapy , Oxidative Stress/drug effects , Proliferating Cell Nuclear Antigen/metabolism , Animals , Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Survival/drug effects , DNA Repair/drug effects , Female , G1 Phase/drug effects , Humans , MCF-7 Cells , Mice , Mice, Inbred C57BL , Neoplasms/genetics , Reactive Oxygen Species/metabolism
16.
Int J Mol Sci ; 22(10)2021 May 17.
Article in English | MEDLINE | ID: mdl-34067547

ABSTRACT

Resveratrol (RSV) is a natural compound that displays several pharmacological properties, including anti-cancer actions. However, its clinical application is limited because of its low solubility and bioavailability. Here, the antiproliferative and anti-inflammatory activity of a series of phenylacetamide RSV derivatives has been evaluated in several cancer cell lines. These derivatives contain a monosubstituted aromatic ring that could mimic the RSV phenolic nucleus and a longer flexible chain that could confer a better stability and bioavailability than RSV. Using MTT assay, we demonstrated that most derivatives exerted antiproliferative effects in almost all of the cancer cell lines tested. Among them, derivative 2, that showed greater bioavailability than RSV, was the most active, particularly against estrogen receptor positive (ER+) MCF7 and estrogen receptor negative (ER-) MDA-MB231 breast cancer cell lines. Moreover, we demonstrated that these derivatives, particularly derivative 2, were able to inhibit NO and ROS synthesis and PGE2 secretion in lipopolysaccharide (LPS)-activated U937 human monocytic cells (derived from a histiocytoma). In order to define the molecular mechanisms underlying the antiproliferative effects of derivative 2, we found that it determined cell cycle arrest at the G1 phase, modified the expression of cell cycle regulatory proteins, and ultimately triggered apoptotic cell death in both breast cancer cell lines. Taken together, these results highlight the studied RSV derivatives, particularly derivative 2, as promising tools for the development of new and more bioavailable derivatives useful in the treatment of breast cancer.


Subject(s)
Breast Neoplasms/metabolism , Resveratrol/pharmacology , Apoptosis/drug effects , Biological Availability , Breast/pathology , Breast Neoplasms/drug therapy , Cell Cycle/drug effects , Cell Cycle Checkpoints/drug effects , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , G1 Phase/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , MCF-7 Cells , Resveratrol/analogs & derivatives
17.
Proc Natl Acad Sci U S A ; 118(24)2021 06 15.
Article in English | MEDLINE | ID: mdl-34099549

ABSTRACT

CAP1 (Cyclase-Associated Protein 1) is highly conserved in evolution. Originally identified in yeast as a bifunctional protein involved in Ras-adenylyl cyclase and F-actin dynamics regulation, the adenylyl cyclase component seems to be lost in mammalian cells. Prompted by our recent identification of the Ras-like small GTPase Rap1 as a GTP-independent but geranylgeranyl-specific partner for CAP1, we hypothesized that CAP1-Rap1, similar to CAP-Ras-cyclase in yeast, might play a critical role in cAMP dynamics in mammalian cells. In this study, we report that CAP1 binds and activates mammalian adenylyl cyclase in vitro, modulates cAMP in live cells in a Rap1-dependent manner, and affects cAMP-dependent proliferation. Utilizing deletion and mutagenesis approaches, we mapped the interaction of CAP1-cyclase with CAP's N-terminal domain involving critical leucine residues in the conserved RLE motifs and adenylyl cyclase's conserved catalytic loops (e.g., C1a and/or C2a). When combined with a FRET-based cAMP sensor, CAP1 overexpression-knockdown strategies, and the use of constitutively active and negative regulators of Rap1, our studies highlight a critical role for CAP1-Rap1 in adenylyl cyclase regulation in live cells. Similarly, we show that CAP1 modulation significantly affected cAMP-mediated proliferation in an RLE motif-dependent manner. The combined study indicates that CAP1-cyclase-Rap1 represents a regulatory unit in cAMP dynamics and biology. Since Rap1 is an established downstream effector of cAMP, we advance the hypothesis that CAP1-cyclase-Rap1 represents a positive feedback loop that might be involved in cAMP microdomain establishment and localized signaling.


Subject(s)
Adenylyl Cyclases/metabolism , Cytoskeletal Proteins/metabolism , Amino Acid Sequence , Animals , Catalytic Domain , Cell Line , Cell Proliferation/drug effects , Cell Survival/drug effects , Cyclic AMP/metabolism , Cytoskeletal Proteins/chemistry , Down-Regulation/drug effects , Enzyme Activation/drug effects , G1 Phase/drug effects , Isoenzymes/metabolism , Protein Binding/drug effects , Rats , S Phase/drug effects , Thyrotropin/pharmacology , rap1 GTP-Binding Proteins/metabolism
18.
Cell Mol Life Sci ; 78(15): 5827-5846, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34155535

ABSTRACT

Stromal interaction molecule 1 (STIM1) and the ORAI1 calcium channel mediate store-operated calcium entry (SOCE) and regulate a multitude of cellular functions. The identity and function of these proteins in thyroid cancer remain elusive. We show that STIM1 and ORAI1 expression is elevated in thyroid cancer cell lines, compared to primary thyroid cells. Knock-down of STIM1 or ORAI1 attenuated SOCE, reduced invasion, and the expression of promigratory sphingosine 1-phosphate and vascular endothelial growth factor-2 receptors in thyroid cancer ML-1 cells. Cell proliferation was attenuated in these knock-down cells due to increased G1 phase of the cell cycle and enhanced expression of cyclin-dependent kinase inhibitory proteins p21 and p27. STIM1 protein was upregulated in thyroid cancer tissue, compared to normal tissue. Downregulation of STIM1 restored expression of thyroid stimulating hormone receptor, thyroid specific proteins and increased iodine uptake. STIM1 knockdown ML-1 cells were more susceptible to chemotherapeutic drugs, and significantly reduced tumor growth in Zebrafish. Furthermore, STIM1-siRNA-loaded mesoporous polydopamine nanoparticles attenuated invasion and proliferation of ML-1 cells. Taken together, our data suggest that STIM1 is a potential diagnostic and therapeutic target for treatment of thyroid cancer.


Subject(s)
Cell Proliferation/genetics , Neoplasm Proteins/genetics , Stromal Interaction Molecule 1/genetics , Thyroid Epithelial Cells/pathology , Thyroid Gland/pathology , Thyroid Neoplasms/genetics , Thyroid Neoplasms/pathology , Adolescent , Adult , Aged , Aged, 80 and over , Animals , Calcium Channels/genetics , Calcium Signaling/drug effects , Calcium Signaling/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Female , G1 Phase/drug effects , G1 Phase/genetics , Humans , Indoles/administration & dosage , Male , Middle Aged , Nanoparticles/administration & dosage , ORAI1 Protein/genetics , Polymers/administration & dosage , RNA, Small Interfering/administration & dosage , Thyroid Epithelial Cells/drug effects , Thyroid Gland/drug effects , Thyroid Neoplasms/drug therapy , Up-Regulation/drug effects , Up-Regulation/genetics , Young Adult , Zebrafish
19.
Drug Deliv ; 28(1): 963-972, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34036867

ABSTRACT

PD-1/PD-L1 blocking therapy has become one of the most promising methods in the field of tumor treatment. However, it encounters the challenge of immune escape due to the exhaustion of T cells. Studies have shown that the epigenetic regulation drug histone deacetylase inhibitor (HDACi) may be able to reverse exhausted T cells by changing the epigenetic transcription program. Therefore, the combination of epigenetic therapy and PD-1/PD-L1 blockade therapy is expected to reverse the immune escape, whereas the overriding goal should aim at the spontaneous release and synergy of PD-1/PD-L1 blocking siRNA and HDACi. In this study, we develop PDDS{polyethylene glycol-b-asparaginate(diethylenetriamine-vorinostat), (PEG-b-P[Asp(DET-SAHA)n] PPDS)}encapsulating siRNA-PD-L1to provide micelles siRNA-PD-L1-loaded micelles (siRNA@PPDS). Transmission electron microscope (TEM) images demonstrate that siRNA@PPDS micelles presented spherical morphology with a size of about 120 nm; hydrodynamic data analysis indicates pH sensitivity of siRNA@PPDS micelles. The experiments reveal that siRNA@PPDS micelles could be well uptaken by the tumor cells to silence the expression of PD-L1 protein in a dose-dependent manner; compared with the free SAHA, the SAHA-loaded micelles PPDS show higher cytotoxicity to induce tumor cell apoptosis and block cell cycle in G1 phase on melanoma-bearing mice, siRNA@PPDS has shown outstanding inhibition of tumor growth and pulmonary metastasis. By comprehensively activating the immune system, lysosome activable polymeric vorinostat encapsulating PD-L1KD for the combination therapy of PD-L1-KD and HDACIs can be an effective strategy to reverse the unresponsiveness of immune checkpoint inhibitors and a promising treatment to inhibit tumor growth, recurrence, and metastasis in clinic.


Subject(s)
Antineoplastic Agents/pharmacology , Histone Deacetylase Inhibitors/pharmacology , Micelles , Programmed Cell Death 1 Ligand 2 Protein/antagonists & inhibitors , RNA, Small Interfering/pharmacology , Vorinostat/pharmacology , Animals , Antineoplastic Agents/administration & dosage , Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , Cell Survival , Chemistry, Pharmaceutical , Dose-Response Relationship, Drug , Drug Carriers/chemistry , Drug Liberation , Epigenesis, Genetic , G1 Phase/drug effects , Histone Deacetylase Inhibitors/administration & dosage , Hydrogen-Ion Concentration , Melanoma, Experimental , Mice , Mice, Inbred C57BL , Microscopy, Electron, Transmission , Particle Size , Polyethylene Glycols/chemistry , RNA, Small Interfering/administration & dosage , Vorinostat/administration & dosage , Xenograft Model Antitumor Assays
20.
Biomed Pharmacother ; 140: 111690, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34004513

ABSTRACT

BACKGROUND: Oxidative stress is a significant feature in the pathomechanism of neurodegenerative diseases. Thus, the search for an effective and safe novel antioxidant agent with neuroprotective properties has increased the interest in medicinal plant products as a bioactive phytochemical source. However, little is known about the potential effects of the medically important Glaucium corniculatum as a natural antioxidant. OBJECTIVE: In the present study, it was aimed to investigate the anti-oxidative, anti-apoptotic, and cell cycle regulatory mechanisms underlying the neuroprotective effects of alkaloid extracts (chloroform, methanol, and water) from G. corniculatum, which was profiled for major alkaloid/alkaloids, against H2O2-induced neuronal damage in differentiated PC12 cells. MATERIALS AND METHODS: The profiles of the alkaloid extracts were analyzed by GC-MS. The effects of the alkaloid extracts on intracellular ROS production, level of apoptotic cells, and cell cycle dysregulation were analyzed by flow cytometry; the effects on mRNA expression of apoptosis-related genes were also analyzed by qRT-PCR. RESULTS: The same alkaloid components, allocryptopine, tetrahydropalmatine, and tetrahydroberberine N-oxide were obtained in all three solvents, but the ratios of the components differed according to the solvents. Allocryptopine was determined to be the major alkaloid ingredient in the alkaloid extracts, with the highest amount of allocryptopine (497 µg/mg) being found in the chloroform alkaloid extract (CAE) (*p < 0.05). The best results were obtained from CAE, which has the highest amount of allocryptopine among alkaloid extracts in all studies. CAE suppressed intracellular ROS production (5.7-fold), percentage of apoptotic cells (3.0-fold), and cells in the sub G1 phase (6.8-fold); additionally, it increased cells in the G1 phase (1.5-fold) (**p < 0.01). CAE remarkably reduced the expressions of Bax, Caspase-9/-3 mRNA (2.4-3.5-fold) while increasing the expression of Bcl-2 mRNA (3.0-fold) (*p < 0.05). CONCLUSIONS: Our results demonstrated that alkaloid extracts from G. corniculatum, which contain allocryptopine, tetrahydropalmatine, and tetrahydroberberine N-oxide suppressed oxidative stress-induced neuronal apoptosis, possibly by suppressing the mitochondrial apoptotic pathway and regulating the cell cycle. These results are the first report that related alkaloids have played a neuroprotective role by regulating multiple mechanisms. Thus, our study indicated that these alkaloids especially allocryptopine could offer an efficient and novel strategy to explore novel drugs for neuroprotection and cognitive improvement.


Subject(s)
Alkaloids/pharmacology , Berberine Alkaloids/pharmacology , Neurons/drug effects , Neuroprotective Agents/pharmacology , Oxidative Stress/drug effects , Plant Extracts/pharmacology , Animals , Antioxidants/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , G1 Phase/drug effects , Hydrogen Peroxide/pharmacology , Mitochondria/drug effects , Mitochondria/metabolism , Neurons/metabolism , PC12 Cells , Papaveraceae/chemistry , Rats , Reactive Oxygen Species/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...