Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 59
Filter
1.
Br J Pharmacol ; 181(22): 4546-4570, 2024 Nov.
Article in English | MEDLINE | ID: mdl-39081110

ABSTRACT

BACKGROUND AND PURPOSE: Gastrointestinal tumours overexpress voltage-gated calcium (CaV3) channels (CaV3.1, 3.2 and 3.3). CaV3 channels regulate cell growth and apoptosis colorectal cancer. Gossypol, a polyphenolic aldehyde found in the cotton plant, has anti-tumour properties and inhibits CaV3 currents. A systematic study was performed on gossypol blocking mechanism on CaV3 channels and its potential anticancer effects in colon cancer cells, which express CaV3 isoforms. EXPERIMENTAL APPROACH: Transcripts for CaV3 proteins were analysed in gastrointestinal cancers using public repositories and in human colorectal cancer cell lines HCT116, SW480 and SW620. The gossypol blocking mechanism on CaV3 channels was investigated by combining heterologous expression systems and patch-clamp experiments. The anti-tumoural properties of gossypol were estimated by cell proliferation, viability and cell cycle assays. Ca2+ dynamics were evaluated with cytosolic and endoplasmic reticulum (ER) Ca2+ indicators. KEY RESULTS: High levels of CaV3 transcripts correlate with poor prognosis in gastrointestinal cancers. Gossypol blockade of CaV3 isoforms is concentration- and use-dependent interacting with the closed, activated and inactivated conformations of CaV3 channels. Gossypol and CaV3 channels down-regulation inhibit colorectal cancer cell proliferation by arresting cell cycles at the G0/G1 and G2/M phases, respectively. CaV3 channels underlie the vectorial Ca2+ uptake by endoplasmic reticulum in colorectal cancer cells. CONCLUSION AND IMPLICATIONS: Gossypol differentially blocked CaV3 channel and its anticancer activity was correlated with high levels of CaV3.1 and CaV3.2 in colorectal cancer cells. The CaV3 regulates cell proliferation and Ca2+ dynamics in colorectal cancer cells. Understanding this blocking mechanism maybe improve cancer therapies.


Subject(s)
Calcium Channel Blockers , Calcium Channels, T-Type , Cell Proliferation , Colonic Neoplasms , Gossypol , Humans , Gossypol/pharmacology , Gossypol/analogs & derivatives , Colonic Neoplasms/drug therapy , Colonic Neoplasms/pathology , Colonic Neoplasms/metabolism , Calcium Channel Blockers/pharmacology , Cell Proliferation/drug effects , Calcium Channels, T-Type/metabolism , Calcium Channels, T-Type/genetics , G1 Phase Cell Cycle Checkpoints/drug effects , Calcium/metabolism , Cell Line, Tumor , Resting Phase, Cell Cycle/drug effects , Antineoplastic Agents/pharmacology
2.
Chem Biol Interact ; 344: 109535, 2021 Aug 01.
Article in English | MEDLINE | ID: mdl-34051208

ABSTRACT

Imatinib, a specific Bcr-Abl tyrosine kinase inhibitor, is the most commonly used drug in the treatment of chronic myeloid leukemia. However, optimal response is not achieved in up to 33% of patients. Therefore, development of novel therapeutic strategies for chronic myeloid leukemia is critical. Betulinic (1) and ursolic (2) acids are natural pentacyclic triterpenes that exhibit antileukemic activities. In this study, we evaluated the effects of pharmacomodulations at the C-3 position of the triterpene moiety of betulinic and ursolic acids on their activity against K562 leukemia cells. Six new derivatives (1a-2c) were synthesized and evaluated for pro-apoptotic and anti-proliferative effects in mammalian and leukemic cells. 2c derivative containing an amine group at the C-3 position of ursolic acid was the most active against leukemia cells with an IC50 value of 5.2 µM after 48 h of treatment. 2c did not exhibit cytotoxic effects against VERO and HepG2 cells and human lymphocytes, showing a good selectivity index for cancer over normal cells. Induced cell death by apoptosis via caspases 3 and 8, and also caused cell cycle arrest as evidenced by accumulation of cells in the G1 phase and decreased cell population in the G2 phase. Furthermore, co-treatment of 2c with imatinib, the chemotherapy drug most commonly used to treat leukemia, resulted in a synergistic effect. Our findings provide a strong rationale for further investigation of combination therapy using the 2c derivative and imatinib in pre-clinical studies.


Subject(s)
Antineoplastic Agents/pharmacology , Imatinib Mesylate/pharmacology , Triterpenes/pharmacology , Animals , Antineoplastic Agents/chemical synthesis , Apoptosis/drug effects , Caspase 3/metabolism , Caspase 8/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Chlorocebus aethiops , Drug Screening Assays, Antitumor , Drug Synergism , G1 Phase Cell Cycle Checkpoints/drug effects , G2 Phase Cell Cycle Checkpoints/drug effects , Humans , Triterpenes/chemical synthesis , Vero Cells , Ursolic Acid
3.
Cell Death Dis ; 12(2): 194, 2021 02 18.
Article in English | MEDLINE | ID: mdl-33602932

ABSTRACT

Malignant transformation involves an orchestrated rearrangement of cell cycle regulation mechanisms that must balance autonomic mitogenic impulses and deleterious oncogenic stress. Human papillomavirus (HPV) infection is highly prevalent in populations around the globe, whereas the incidence of cervical cancer is 0.15%. Since HPV infection primes cervical keratinocytes to undergo malignant transformation, we can assume that the balance between transforming mitogenic signals and oncogenic stress is rarely attained. We showed that highly transforming mitogenic signals triggered by HRasG12V activity in E6E7-HPV-keratinocytes generate strong replication and oxidative stresses. These stresses are counteracted by autophagy induction that buffers the rapid increase of ROS that is the main cause of genotoxic stress promoted by the oncoprotein. As a result, autophagy creates a narrow window of opportunity for malignant keratinocytes to emerge. This work shows that autophagy is crucial to allow the transition of E6E7 keratinocytes from an immortalized to a malignant state caused by HRasG12V.


Subject(s)
Alphapapillomavirus/pathogenicity , Autophagy , Cell Transformation, Viral , DNA Damage , Keratinocytes/virology , Papillomavirus Infections/virology , Proto-Oncogene Proteins p21(ras)/metabolism , Uterine Cervical Neoplasms/virology , Alphapapillomavirus/genetics , Alphapapillomavirus/metabolism , Cell Line , Cell Proliferation , Female , G1 Phase Cell Cycle Checkpoints , Host-Pathogen Interactions , Humans , Keratinocytes/metabolism , Keratinocytes/pathology , Mitosis , Oncogene Proteins, Viral/genetics , Oncogene Proteins, Viral/metabolism , Oxidative Stress , Papillomavirus E7 Proteins/genetics , Papillomavirus E7 Proteins/metabolism , Papillomavirus Infections/metabolism , Papillomavirus Infections/pathology , Proto-Oncogene Proteins p21(ras)/genetics , Repressor Proteins/genetics , Repressor Proteins/metabolism , Signal Transduction , Uterine Cervical Neoplasms/metabolism , Uterine Cervical Neoplasms/pathology
4.
Clin Transl Oncol ; 23(3): 612-619, 2021 Mar.
Article in English | MEDLINE | ID: mdl-32710211

ABSTRACT

BACKGROUND: Glioblastoma (GB) remains an incurable and deadly brain malignancy that often proves resistant to upfront treatment with temozolomide. Nevertheless, temozolomide remains the most commonly prescribed FDA-approved chemotherapy for GB. The DNA repair protein methylguanine-DNA methyl transferase (MGMT) confers resistance to temozolomide. Unsurprisingly temozolomide-resistant tumors tend to possess elevated MGMT protein levels or lack inhibitory MGMT promotor methylation. In this study, cultured human temozolomide resistance GB (43RG) cells were introduced to the MGMT inhibitor O6-benzylguanine combined with temozolomide and either LY2835219 (CDK 4/6 inhibitor) or LY2157299 (TGF-ßRI inhibitor) seeking to overcome GB treatment resistance. METHODS: Treatment effects were assessed using MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) assay, western blot, cell viability, and cell cycle progression. RESULTS: Our in vitro study demonstrated that sequential treatment of O6-Benzylguanine with either LY2385219 or LY2157299-enhanced temozolomide enhanced sensitivity in MGMT+ 43RG cells. Importantly, normal human neurons and astrocytes remained impervious to the drug therapies under these conditions. Furthermore, LY2835219 has additional anti-proliferative effects on cell cycling, including induction of an RB-associated G (1) arrest via suppression of cyclin D-CDK4/6-Rb pathway. LY2157299 enhances anti-tumor effect by disrupting TGF-ß-dependent HIF-1α signaling and by activating both Smad and PI3K-AKT pathways towards transcription of S/G2 checkpoints. CONCLUSION: This study establishes the groundwork for the development of a combinatorial pharmacologic approach by using either LY2385219 or LY2157299 inhibitor plus O6-Benzylguanine to augment temozolomide response in temozolomide-resistant GB cells.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , Brain Neoplasms/drug therapy , Cyclin-Dependent Kinase 4/antagonists & inhibitors , Cyclin-Dependent Kinase 6/antagonists & inhibitors , DNA Modification Methylases/antagonists & inhibitors , DNA Repair Enzymes/antagonists & inhibitors , Glioblastoma/drug therapy , Receptor, Transforming Growth Factor-beta Type I/antagonists & inhibitors , Temozolomide/pharmacology , Tumor Suppressor Proteins/antagonists & inhibitors , Aminopyridines/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Astrocytes/drug effects , Benzimidazoles/pharmacology , Brain Neoplasms/enzymology , Cell Cycle/drug effects , Cell Survival/drug effects , Cells, Cultured , Cyclin D/antagonists & inhibitors , Drug Resistance, Neoplasm/drug effects , G1 Phase Cell Cycle Checkpoints , Glioblastoma/enzymology , Guanine/analogs & derivatives , Guanine/pharmacology , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/drug effects , Neurons/drug effects , Phosphatidylinositol 3-Kinases/drug effects , Pyrazoles/pharmacology , Quinolines/pharmacology , Smad Proteins/drug effects
5.
Clin Transl Oncol ; 23(4): 856-865, 2021 Apr.
Article in English | MEDLINE | ID: mdl-32880047

ABSTRACT

BACKGROUND: Deregulated expression of cell cycle regulators p27 and p16 is associated with cancer progression. p27kip1 and p16INKa are a cyclin dependent kinase inhibitor whose major target is the cyclinE/CDK2 and cyclinD/CDK4/6 complex, respectively, that governs cell cycle transition from late G1 to S phase. METHODS: We recruited biopsies of a total of 84 subjects including 72 primary tumor biopsies from histopathologically proven gastric carcinoma, 8 adjacent controls and 12 independent controls. We used gastric cancer cell line, AGS, for validation of our data. Expression profiling at transcript level was done by semi-quantitative RT-PCR and at proteome level by immunohistochemistry and immunofluorescence. Receiver operator characteristics analysis was done for determining the diagnostic utility of p27 and p16 with respect to the sensitivity and specificity. RESULTS: We demonstrate that p27 and p16 are frequently over expressed in early stages of gastric carcinoma. Our semi-quantitative data show a significant upregulation of p27 (Mean ± SEM, 0.4771 ± 0.0895; p = 0.0001) and p16 (Mean ± SEM, 0.4676 ± 0.04305; p = 0.0001) at mRNA level. Concordant to semi-quantitative data, immunohistochemistry data also showed a significant upregulation of p27 (Mean ± SEM, 196.4 ± 10.84; p < 0.0001) and p16 (Mean ± SEM, 100.4 ± 23.71; p < 0.0001) at protein level. CONCLUSIONS: The present study showed that the significant upregulation of p27 and p16 were associated with early events in gastric carcinogenesis. Our data suggests that clinical correlation of these differentially expressed genes may be useful as diagnostic biomarkers for early detection of gastric carcinoma and promising therapeutics target for GC patients.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p16/metabolism , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Stomach Neoplasms/metabolism , Adult , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Disease Progression , Early Detection of Cancer , Female , Fluorescent Antibody Technique , G1 Phase Cell Cycle Checkpoints , Humans , Immunohistochemistry , Male , Middle Aged , RNA, Messenger/metabolism , ROC Curve , Reverse Transcriptase Polymerase Chain Reaction , S Phase Cell Cycle Checkpoints , Sensitivity and Specificity , Stomach Neoplasms/diagnosis , Stomach Neoplasms/pathology , Up-Regulation , Young Adult
6.
Clin Transl Oncol ; 23(2): 222-228, 2021 Feb.
Article in English | MEDLINE | ID: mdl-32594303

ABSTRACT

PURPOSE: The current study aims to explore the effects of CDKN2A on cell proliferation and cycle, and investigate the underlying mechanisms. METHODS: Expression of CDKN2A in cervical cancer cell lines was evaluated by real-time quantitative PCR (RT-qPCR) and western blotting. Apoptotic rate was detected by Annexin V assay. MTT assay, Transwell assay and cell cycle assay kit were applied to examine the effect of CDKN2A on cell viability, invasion and cell cycle. Co-immunoprecipitation and western blotting were devoted to explore the mechanism by which CDKN2A contributes to cell function. RESULTS: CDKN2A was expressed at a low level in cervical cancer cell lines. Overexpression of CDKN2A inhibited cell proliferation and invasion, and caused cell cycle arrest in the G1 phase. CDKN2A mediates the AKT-mTOR signaling pathway by suppressing lactate dehydrogenase (LDHA). Taken together, our data revealed that CDKN2A can be applied as a therapeutic target for the treatment of cervical cancer in future. CONCLUSIONS: CDKN2A inhibits cell proliferation and invasion in cervical cancer through LDHA-mediated AKT-mTOR pathway.


Subject(s)
Cell Proliferation/physiology , Cyclin-Dependent Kinase Inhibitor p16/metabolism , L-Lactate Dehydrogenase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases/metabolism , Uterine Cervical Neoplasms/metabolism , Apoptosis/physiology , Cell Line, Tumor , Cell Survival/physiology , Down-Regulation/physiology , Female , G1 Phase Cell Cycle Checkpoints , HeLa Cells , Humans , Immunoprecipitation , Neoplasm Invasiveness , Uterine Cervical Neoplasms/pathology
7.
Arch Biochem Biophys ; 696: 108654, 2020 12 15.
Article in English | MEDLINE | ID: mdl-33130087

ABSTRACT

ß-Sitosterol (ßSito) is the most abundant phytosterol found in vegetable oils, grains such as wheat, beans, and corn, and in many phytosterol-enriched foods. It is prone to oxidation by reactive oxygen species, such as ozone, leading to the formation of oxyphytosterols. A better understanding regarding the biological effects and mechanism of action of oxyphytosterols is required since the beneficial and adverse side effects of these compounds on human health remain highly controversial. In this work, we investigated the biological effects of ß-Secosterol (ßSec), a new oxyphytosterol generated by the reaction of ßSito with ozone. Treatment of HepG2 cells with ßSito or ßSec (0.1-100 µM) for 24, 48, and 72 h induced a dose-dependent reduction of cell viability in the MTT assay, with ßSec showing higher efficacy than ßSito. However, ßSec presented a lower potency than ßSito, showing IC50 = 37.32 µM, higher than ßSito (IC50 = 0.23 µM) at 48 h. Cell cycle analyses by flow cytometry showed a slight decrease of G0/G1 phase with ßSito 0.5 µM, but a significant cell cycle arrest at the G0/G1 phase in the treatment for 48 h with ßSec 20 µM (62.69 ± 2.15%, p < 0.05) and ßSec 40 µM (66.96 ± 5.39%, p < 0.0001) when compared to control (56.97 ± 2.60%). No suggestion of apoptosis was indicated by flow cytometry data. Also, ßSec (20 and 40 µM) reduced the mitotic index. In the laser scanning confocal microscopy analysis no alterations in cell morphology were observed with ßSito (0.5 µM). Nevertheless, round-shaped cells, abnormal nuclear morphology with shrinkage, and formation of microtubules clusters were observed in the treatment with ßSec, indicating a disruption in the microtubules network organization. N-acetyl-l-cysteine was not able to inhibit any of these cellular effects, indicating a lack of involvement of oxidative stress in the mechanism of action of ßSec. Although not further investigated in this study, it was discussed the hypothesis that covalent adduct formation with lysine residues of proteins, could play an important role in the biological effects elicited by ßSec. Elucidation of the primary cellular processes induced by ßSec provides the essential knowledge to be aware of its potential adverse side effects or therapeutic use of this oxyphytosterol.


Subject(s)
Sitosterols/pharmacology , Acetylcysteine/pharmacology , Cell Nucleus/drug effects , Cell Survival/drug effects , G1 Phase Cell Cycle Checkpoints/drug effects , Hep G2 Cells , Humans , Microtubules/drug effects , Mitotic Index , Oxidative Stress/drug effects , Ozone/chemistry , Sitosterols/chemical synthesis , Sitosterols/chemistry
8.
An Acad Bras Cienc ; 92(4): e20191533, 2020.
Article in English | MEDLINE | ID: mdl-33237139

ABSTRACT

Seseli L. is an important genus of the Apiaceae family, with a large number of aromatic species. It is used in traditional medicine extensively, but there is quite limited information on their phytochemicals and biological activities. Seseli petraeum M. Bieb. grows in Northern Anatolia, and there are no phytochemical studies on this species. In the present study, we aimed to investigate the effect of the extracts of S. petraeum on A549 lung cancer cell proliferation. For this purpose, the antiproliferative effect was determined via MTT assay, and the extracts obtained from the root of S. petraeum showed a significant inhibitory effect on cell proliferation. The hexane extract of the root exhibited potent inhibition on A549 cancer cell growth at the 24th hour with 3.432 mg/mL IC50 value. The results also showed that the hexane extract had displayed cytotoxic effect through an arrest at the G0/G1 phase of the cell cycle and induced apoptosis as well as DNA damage of A549 cells. Consequently, this study demonstrated the antiproliferative potential of the extracts from S. petraeum, especially hexane extract from the roots. Further studies are required to identify the mechanisms underlying these effects.


Subject(s)
Apiaceae , Lung Neoplasms , A549 Cells , Apoptosis , Cell Cycle , Cell Line, Tumor , Cell Proliferation , G1 Phase Cell Cycle Checkpoints , Humans , Lung Neoplasms/drug therapy , Plant Extracts/pharmacology
9.
Inorg Chem ; 59(20): 15004-15018, 2020 Oct 19.
Article in English | MEDLINE | ID: mdl-32997499

ABSTRACT

In this paper, a series of new ruthenium complexes of the general formula [Ru(NS)(dpphpy)(dppb)]PF6 (Ru1-Ru3), where dpphpy = diphenyl-2-pyridylphosphine, NS ligands = 2-thiazoline-2-thiol (tzdt, Ru1), 2-mercaptopyrimidine (pySm, Ru2), and 4,6-diamino-2-mercaptopyrimidine (damp, Ru3), and dppb = 1,4-bis(diphenylphosphino)butane, were synthesized and characterized by elemental analysis, spectroscopic techniques (IR, UV/visible, and 1D and 2D NMR), and X-ray diffraction. In the characterization, the correlation between the phosphorus atoms and their respective aromatic hydrogen atoms of the compounds in the assignment stands outs, by 1H-31P HMBC experiments. The compounds show anticancer activities against A549 (lung) and MDA-MB-231 (breast) cancer cell lines, higher than the clinical drug cisplatin. All of the complexes are more cytotoxic against the cancer cell lines than against the MRC-5 (lung) and MCF-10A (breast) nontumorigenic human cell lines. For A549 tumor cells, cell cycle analysis upon treatment with Ru2 showed that it inhibits the mitotic phase because arrest was observed in the Sub-G1 phase. Additionally, the compound induces cell death by an apoptotic pathway in a dose-dependent manner, according to annexin V-PE assay. The multitargeted character of the compounds was investigated, and the biomolecules were DNA, topoisomerase IB, and proteasome, as well as the fundamental biomolecule in the pharmacokinetics of drugs, human serum albumin. The experimental results indicate that the complexes do not target DNA in the cells. At low concentrations, the compounds showed the ability to partially inhibit the catalytic activity of topoisomerase IB in the process of relaxation of the DNA plasmid. Among the complexes assayed in cultured cells, complex Ru3 was able to diminish the proteasomal chymotrypsin-like activity to a greater extent.


Subject(s)
Antineoplastic Agents/pharmacology , Coordination Complexes/pharmacology , DNA Topoisomerases, Type I/metabolism , Proteasome Inhibitors/pharmacology , Topoisomerase I Inhibitors/pharmacology , Antineoplastic Agents/chemical synthesis , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Coordination Complexes/chemical synthesis , Drug Screening Assays, Antitumor , G1 Phase Cell Cycle Checkpoints/drug effects , Humans , Phosphines/chemical synthesis , Phosphines/pharmacology , Proteasome Inhibitors/chemical synthesis , Ruthenium/chemistry , Sulfhydryl Compounds/chemical synthesis , Sulfhydryl Compounds/pharmacology , Topoisomerase I Inhibitors/chemical synthesis
10.
DNA Repair (Amst) ; 96: 102971, 2020 12.
Article in English | MEDLINE | ID: mdl-32987354

ABSTRACT

How a cell determines a given phenotype upon damaged DNA is an open problem. Cell fate decisions happen at cell cycle checkpoints and it is becoming clearer that the p53 pathway is a major regulator of cell fate decisions involving apoptosis or senescence upon DNA damage, especially at G1/S. However, recent results suggest that this pathway is also involved in autophagy induction upon DNA damage. To our knowledge, in this work we propose the first model of the DNA damage-induced G1/S checkpoint contemplating the decision between three phenotypes: apoptosis, senescence, and autophagy. The Boolean model is proposed based on experiments with U87 glioblastoma cells using the transfection of miR-16 that can induce a DNA damage response. The wild-type case of the model shows that DNA damage induces the checkpoint and the coexistence of the three phenotypes (tristable dynamics), each with a different probability. We also predict that the positive feedback involving ATM, miR-16, and Wip1 has an influence on the tristable state. The model predictions were compared to experiments of gain and loss of function in other three different cell lines (MCF-7, A549, and U2OS) presenting agreement. For p53-deficient cell lines such as HeLa, H1299, and PC-3, our model contemplates the experimental observation that the alternative AMPK pathway can compensate this deficiency. We conclude that at the G1/S checkpoint the p53 pathway (or, in its absence, the AMPK pathway) can regulate the induction of different phenotypes in a stochastic manner in the U87 cell line and others.


Subject(s)
Autophagy , DNA Damage , G1 Phase Cell Cycle Checkpoints , Models, Genetic , Signal Transduction , Tumor Suppressor Protein p53/metabolism , Apoptosis , Ataxia Telangiectasia Mutated Proteins/metabolism , Cellular Senescence , Gene Regulatory Networks , Glioblastoma/genetics , Glioblastoma/metabolism , Humans , MicroRNAs/metabolism , Protein Phosphatase 2C/metabolism , Tumor Cells, Cultured
11.
Molecules ; 25(17)2020 Aug 28.
Article in English | MEDLINE | ID: mdl-32872363

ABSTRACT

In research on natural molecules with cytotoxic activity that can be used for the development of new anticancer agents, the cytotoxic activity of hexane, chloroform, and methanol extracts from the roots of Acacia schaffneri against colon, lung, and skin cancer cell lines was explored. The hexane extract showed the best activity with an average IC50 of 10.6 µg mL-1. From this extract, three diterpenoids, phyllocladan-16α,19-diol (1), phyllocladan-16α-ol (2), and phylloclad-16-en-3-ol (3), were isolated and characterized by their physical and spectroscopic properties. Diterpenoids 1 and 2 were tested against the same cancer cell lines, as well as their healthy counterparts, CCD841 CoN, MRC5, and VH10, respectively. Compound 1 showed moderate activity (IC50 values between 24 and 70 µg mL-1), although it showed a selective effect against cancer cell lines. Compound 2 was practically inactive. The cytotoxicity mechanism of 1 was analyzed by cell cycle, indicating that the compound induces G0/G1 cell cycle arrest. This effect might be generated by DNA alkylation damage. In addition, compound 1 decreased migration of HT29 cells.


Subject(s)
Fabaceae/chemistry , Plant Extracts/chemistry , Plant Extracts/pharmacology , Plant Roots/chemistry , Cell Line, Tumor , Cell Movement , Cell Proliferation/drug effects , Cell Survival/drug effects , Chemical Fractionation , G1 Phase Cell Cycle Checkpoints/drug effects , Humans , Magnetic Resonance Spectroscopy , Molecular Structure , Plant Extracts/isolation & purification
12.
Toxins (Basel) ; 12(10)2020 09 24.
Article in English | MEDLINE | ID: mdl-32987941

ABSTRACT

Canine and human osteosarcomas (OSA) share similarities. Novel therapies are necessary for these tumours. The Bacillus anthracis toxin was reengineered to target and kill cells with high expressions of matrix metalloproteinases (MMPs) and urokinase plasminogen activator (uPA). Since canine OSA express MMPs and uPA, we assessed whether the reengineered toxin could show efficacy against these tumours. Two OSA cell lines (canine D17 and human MG63) and a non-neoplastic canine osteoblastic cell line (COBS) were used. Cells were treated with different concentrations of the reengineered anthrax toxin and cell viability was quantified using MTT assay. The cell cycle, apoptosis, and necrosis were analysed by flow cytometry. The wound-healing assay was performed to quantify the migration capacity of treated cells. D17 and MG63 cells had significantly decreased viability after 24 h of treatment. Cell cycle analysis revealed that OSA cells underwent apoptosis when treated with the toxin, whereas COBS cells arrested in the G1 phase. The wound-healing assay showed that D17 and MG63 cells had a significantly reduced migration capacity after treatment. These results point for the first time towards the in vitro inhibitory effects of the reengineered anthrax toxin on OSA cells; this reengineered toxin could be further tested as a new therapy for OSA.


Subject(s)
Antigens, Bacterial/pharmacology , Antineoplastic Agents/pharmacology , Bacterial Toxins/pharmacology , Bone Neoplasms/drug therapy , Osteosarcoma/drug therapy , Adolescent , Animals , Antigens, Bacterial/genetics , Apoptosis/drug effects , Bacterial Toxins/genetics , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Cell Line, Tumor , Cell Movement/drug effects , Dogs , Dose-Response Relationship, Drug , Female , G1 Phase Cell Cycle Checkpoints/drug effects , Humans , Inhibitory Concentration 50 , Male , Matrix Metalloproteinases/metabolism , Membrane Proteins/metabolism , Neoplasm Invasiveness , Osteosarcoma/metabolism , Osteosarcoma/pathology , Protein Engineering
13.
Inorg Chem ; 59(7): 5072-5085, 2020 Apr 06.
Article in English | MEDLINE | ID: mdl-32208661

ABSTRACT

In this study, half-sandwich Ru(II) complexes containing acylthiourea ligands of the general type [Ru(η6-p-cymene)(PPh3)(S)Cl]PF6 (1m-6m) and [Ru(η6-p-cymene)(PPh3)(S-O)]PF6 (1b-6b) where S/S-O = N',N'-disubstituted acylthiourea were synthesized and characterized (via elemental analyses, IR spectroscopy, 1H NMR spectroscopy, 13C{1H} NMR spectroscopy, and X-ray diffractometry), and their cytotoxic activity was evaluated. The different coordination modes of the acylthiourea ligands, monodentately via S (1m-6m) and bidentately via S,O (1b-6b), to ruthenium were modulated from different synthetic routes. The cytotoxicity of the complexes was evaluated in five human cell lines (DU-145, A549, MDA-MB-231, MRC-5, and MCF-10A) by MTT assay. The IC50 values for prostate cancer cells (2.89-7.47 µM) indicated that the complexes inhibited cell growth, but that they were less cytotoxic than cisplatin (2.00 µM). Unlike for breast cancer cells (IC50 = 0.28-0.74 µM) and lung cancer cells (IC50 = 0.51-1.83 µM), the complexes were notably more active than the reference drug, and a remarkable selectivity index (SI 4.66-19.34) was observed for breast cancer cells. Based on both the activity and selectivity, complexes 5b and 6b, as well as their respective analogous complexes in the monodentate coordination 5m and 6m, were chosen for further investigation in the MDA-MB-231 cell line. These complexes not only induced morphology changes but also were able to inhibit colony formation and migration. In addition, the complexes promoted cell cycle arrest at the sub-G1 phase inducing apoptosis. Interaction studies by viscosity measurements, gel electrophoresis, and fluorescence spectroscopy indicated that the complexes interact with the DNA minor groove and exhibit an HSA binding affinity.


Subject(s)
Antineoplastic Agents/pharmacology , Coordination Complexes/pharmacology , Thiourea/analogs & derivatives , Thiourea/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/metabolism , Apoptosis/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Coordination Complexes/chemical synthesis , Coordination Complexes/metabolism , DNA/metabolism , Drug Screening Assays, Antitumor , G1 Phase Cell Cycle Checkpoints/drug effects , Humans , Ligands , Molecular Structure , Ruthenium/chemistry , Serum Albumin, Human/metabolism , Thiourea/metabolism
14.
Chem Biol Interact ; 311: 108789, 2019 Sep 25.
Article in English | MEDLINE | ID: mdl-31401089

ABSTRACT

The cytotoxicity of a dinuclear imine-copper (II) complex 2, and its analogous mononuclear complex 1, toward different melanoma cells, particularly human SKMEL-05 and SKMEL-147, was investigated. Complex 2, a tyrosinase mimic, showed much higher activity in comparison to complex 1, and its reactivity was verified to be remarkably activated by UVB-light, while the mononuclear compound showed a small or negligible effect. Further, a significant dependence on the melanin content in the tumor cells, both from intrinsic pigmentation or stimulated by irradiation, was observed in the case of complex 2. Similar tests with keratinocytes and melanocytes indicated a much lower sensitivity to both copper (II) complexes, even after exposition to UV light. Clonogenic assays attested that the fractions of melanoma cells survival were much lower under treatment with complex 2 compared to complex 1, both with or without previous irradiation of the cells. The process also involves generation of reactive oxygen species (ROS), as verified by EPR spectroscopy, and by using fluorescence indicators. Autophagic assays indicated a remarkable formation of cytoplasmic vacuoles in melanomas treated with complex 2, while this effect was not observed in similar treatment with complex 1. Monitoring of specific protein LC3 corroborated the simultaneous occurrence of autophagy. A balance interplay between different modes of cell death, apoptosis and autophagy, occurs when melanomas were treated with the dinuclear complex 2, in contrast to the mononuclear complex 1. These results pointed out to different mechanisms of action of such complexes, depending on its nuclearity.


Subject(s)
Coordination Complexes/chemistry , Copper/chemistry , Imines/chemistry , Monophenol Monooxygenase/metabolism , Animals , Autophagy/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/radiation effects , Coordination Complexes/metabolism , Coordination Complexes/pharmacology , Electron Spin Resonance Spectroscopy , G1 Phase Cell Cycle Checkpoints/drug effects , G1 Phase Cell Cycle Checkpoints/radiation effects , Humans , Melanins/metabolism , Melanoma/metabolism , Melanoma/pathology , Mice , Mitochondria/drug effects , Mitochondria/metabolism , Reactive Oxygen Species/metabolism , Tubulin/metabolism , Ultraviolet Rays
15.
Chembiochem ; 20(18): 2390-2401, 2019 09 16.
Article in English | MEDLINE | ID: mdl-31026110

ABSTRACT

Class 1 myosins (Myo1s) were the first unconventional myosins identified and humans have eight known Myo1 isoforms. The Myo1 family is involved in the regulation of gene expression, cytoskeletal rearrangements, delivery of proteins to the cell surface, cell migration and spreading. Thus, the important role of Myo1s in different biological processes is evident. In this study, we have investigated the effects of pentachloropseudilin (PClP), a reversible and allosteric potent inhibitor of Myo1s, on angiogenesis. We demonstrated that treatment of cells with PClP promoted a decrease in the number of vessels. The observed inhibition of angiogenesis is likely to be related to the inhibition of cell proliferation, migration and adhesion, as well as to alteration of the actin cytoskeleton pattern, as shown on a PClP-treated HUVEC cell line. Moreover, we also demonstrated that PClP treatment partially prevented the delivery of integrins to the plasma membrane. Finally, we showed that PClP caused DNA strand breaks, which are probably repaired during the cell cycle arrest in the G1 phase. Taken together, our results suggest that Myo1s participate directly in the angiogenesis process.


Subject(s)
Actin Cytoskeleton/drug effects , Angiogenesis Inhibitors/pharmacology , Cell Cycle/drug effects , Hydrocarbons, Chlorinated/pharmacology , Integrins/metabolism , Pyrroles/pharmacology , Angiogenesis Inhibitors/toxicity , Cell Adhesion/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , G1 Phase Cell Cycle Checkpoints/drug effects , Human Umbilical Vein Endothelial Cells , Humans , Hydrocarbons, Chlorinated/toxicity , Integrins/genetics , Myosin Type I/metabolism , Pyrroles/toxicity , RNA, Messenger/metabolism
16.
J Inorg Biochem ; 195: 1-12, 2019 06.
Article in English | MEDLINE | ID: mdl-30861423

ABSTRACT

Some metallodrugs that exhibit interesting biological activity contain transition metals such as ruthenium, and have been extensively exploited because of their antiparasitic potential. In previous study, we reported the remarkable anti-Leishmania activity of precursor cis-[RuIICl2(dppm)2], where dppm = bis(diphenylphosphino)methane, and new ruthenium(II) complexes, cis-[RuII(η2-O2CC10H13)(dppm)2]PF6 (bbato), cis-[RuII(η2-O2CC7H7S)(dppm)2]PF6 (mtbato) and cis-[RuII(η2-O2CC7H7O2)(dppm)2]PF6 (hmxbato) against some Leishmania species. In view of the promising activity of the hmxbato complex against Leishmania (Leishmania) amazonensis promastigotes, the present work investigated the possible parasite death mechanism involved in the action of this hmxbato and its precursor. We report, for the first time, that hmxbato and precursor promoted an increase in reactive oxygen species production, depolarization of the mitochondrial membrane, DNA fragmentation, formation of a pre-apoptotic peak, alterations in parasite morphology and formation of autophagic vacuoles. Taken together, our results suggest that these ruthenium complexes cause parasite death by apoptosis. Thus, this work provides relevant knowledge on the activity of ruthenium(II) complexes against L. (L.) amazonensis. Such information will be essential for the exploitation of these complexes as future candidates for cutaneous leishmaniasis treatment.


Subject(s)
Apoptosis/drug effects , Coordination Complexes/pharmacology , Leishmania/drug effects , Reactive Oxygen Species/metabolism , Trypanocidal Agents/pharmacology , Cell Proliferation/drug effects , DNA Fragmentation/drug effects , DNA, Protozoan/drug effects , G1 Phase Cell Cycle Checkpoints/drug effects , Membrane Potential, Mitochondrial/drug effects , Mitochondria/drug effects , Ruthenium/chemistry
17.
Cell Physiol Biochem ; 48(1): 397-408, 2018.
Article in English | MEDLINE | ID: mdl-30016791

ABSTRACT

BACKGROUND/AIMS: Compared with non-obese individuals, obese individuals commonly store more vitamin D in adipose tissue. VDR expression in adipose tissue can influence adipogenesis and is therefore a target pathway deserving further study. This study aims to assess the role of 1,25(OH)2D3 in human preadipocyte proliferation and differentiation. METHODS: RTCA, MTT, and trypan blue assays were used to assess the effects of 1,25(OH)2D3 on the viability, proliferation, and adipogenic differentiation of SGBS cells. Cell cycle and apoptosis analyses were performed with flow cytometry, triglycerides were quantified, and RT-qPCR was used to assess gene expression. RESULTS: We confirmed that the SGBS cell model is suitable for studying adipogenesis and demonstrated that the differentiation protocol induces cell maturation, thereby increasing the lipid content of cells independently of treatment. 1,25(OH)2D3 treatment had different effects according to the cell stage, indicating different modes of action driving proliferation and differentiation. In preadipocytes, 1,25(OH)2D3 induced G1 growth arrest at both tested concentrations without altering CDKN1A gene expression. Treatment with 100 nM 1,25(OH)2D3 also decreased MTT absorbance and the lipid concentration. Moreover, increased normalized cell index values and decreased metabolic activity were not induced by proliferation or apoptosis. Exposure to 100 nM 1,25(OH)2D3 induced VDR, CEBPA, and CEBPB expression, even in the preadipocyte stage. During adipogenesis, 1,25(OH)2D3 had limited effects on processes such as VDR and PPARG gene expression, but it upregulated CEBPA expression. CONCLUSIONS: We demonstrated for the first time that 1,25(OH)2D3 induces changes in preadipocytes, including VDR expression and growth arrest, and increases the lipid content in adipocytes treated for 16 days. Preadipocytes are important cells in adipose tissue homeostasis, and understanding the role of 1,25(OH)2D3 in adipogenesis is a crucial step in ensuring adequate vitamin D supplementation, especially for obese individuals.


Subject(s)
Adipogenesis/drug effects , Cell Proliferation/drug effects , Vitamin D/analogs & derivatives , Adipocytes/cytology , Adipocytes/drug effects , Adipocytes/metabolism , CCAAT-Enhancer-Binding Protein-beta/genetics , CCAAT-Enhancer-Binding Protein-beta/metabolism , CCAAT-Enhancer-Binding Proteins/genetics , CCAAT-Enhancer-Binding Proteins/metabolism , Cell Line , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , G1 Phase Cell Cycle Checkpoints/drug effects , Humans , PPAR gamma/genetics , PPAR gamma/metabolism , Receptors, Calcitriol/genetics , Receptors, Calcitriol/metabolism , Up-Regulation/drug effects , Vitamin D/pharmacology
18.
Cell Cycle ; 17(14): 1721-1744, 2018.
Article in English | MEDLINE | ID: mdl-29995582

ABSTRACT

Human pluripotent stem cells (hPSCs), including embryonic and induced pluripotent stem cells (hESCs and hiPSCs) show unique cell cycle characteristics, such as a short doubling time due to an abbreviated G1 phase. Whether or not the core cell cycle machinery directly regulates the stemness and/or the differentiation potential of hPSCs remains to be determined. To date, several scenarios describing the atypical cell cycle of hPSCs have been suggested, and therefore there is still controversy over how cyclins, master regulators of the cell cycle, are expressed and regulated. Furthermore, the cell cycle profile and the expression pattern of major cyclins in hESCs-derived neuroprogenitors (NP) have not been studied yet. Therefore, herein we characterized the expression pattern of major cyclins in hPSCs and NP. We determined that all studied cyclins mRNA expression levels fluctuate along cell cycle. Particularly, after a thorough analysis of synchronized cell populations, we observed that cyclin E1 mRNA levels increased sharply in G1/S concomitantly with cyclin E1 protein accumulation in hPSCs and NP. Additionally, we demonstrated that cyclin E1 mRNA expression levels involves the activation of MEK/ERK pathway and the transcription factors c-Myc and E2Fs in hPSCs. Lastly, our results reveal that proteasome mediates the marked down-regulation (degradation) of cyclin E1 protein observed in G2/M by a mechanism that requires a functional CDK2 but not GSK3ß activity. ABBREVIATIONS: hPSCs: human pluripotent stem cells; hESCs: human embryonic stem cells; hiPSCs: human induced pluripotent stem cells; NP: neuroprogenitors; HF: human foreskin fibroblasts; MEFs: mouse embryonic fibroblasts; iMEFs: irradiated mouse embryonic fibroblasts; CDKs: cyclindependent kinases; CKIs: CDK inhibitors; CNS: central nervous system; Oct-4: Octamer-4; EB: embryoid body; AFP: Alpha-fetoprotein; cTnT: Cardiac Troponin T; MAP-2: microtubule-associated protein; TUJ-1: neuron-specific class III ß-tubulin; bFGF: basic fibroblastic growth factor; PI3K: Phosphoinositide 3-kinase; KSR: knock out serum replacement; CM: iMEF conditioned medium; E8: Essential E8 medium.


Subject(s)
Cyclin E/genetics , Gene Expression Regulation , Neurons/cytology , Neurons/metabolism , Oncogene Proteins/genetics , Pluripotent Stem Cells/cytology , Cell Proliferation , Cells, Cultured , Cyclin E/metabolism , E2F Transcription Factors/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Fibroblasts/metabolism , G1 Phase Cell Cycle Checkpoints , G2 Phase , Humans , Mitogen-Activated Protein Kinase Kinases/metabolism , Mitosis , Neural Stem Cells/metabolism , Oncogene Proteins/metabolism , Pluripotent Stem Cells/metabolism , Proto-Oncogene Proteins c-myc/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism
19.
Sci Rep ; 8(1): 5405, 2018 03 29.
Article in English | MEDLINE | ID: mdl-29599445

ABSTRACT

One of the most important mechanisms for repairing double-strand breaks (DSBs) in model eukaryotes is homologous recombination (HR). Although the genes involved in HR have been found in Trypanosoma brucei and studies have identified some of the proteins that participate in this HR pathway, the recruitment kinetics of the HR machinery onto DNA during DSB repair have not been clearly elucidated in this organism. Using immunofluorescence, protein DNA-bound assays, and DNA content analysis, we established the recruitment kinetics of the HR pathway in response to the DSBs generated by ionizing radiation (IR) in procyclic forms of T. brucei. These kinetics involved the phosphorylation of histone H2A and the sequential recruitment of the essential HR players Exo1, RPA, and Rad51. The process of DSB repair took approximately 5.5 hours. We found that DSBs led to a decline in the G2/M phase after IR treatment, concomitant with cell cycle arrest in the G1/S phase. This finding suggests that HR repairs DSBs faster than the other possible DSB repair processes that act during the G1/S transition. Taken together, these data suggest that the interplay between DNA damage detection and HR machinery recruitment is finely coordinated, allowing these parasites to repair DNA rapidly after DSBs during the late S/G2 proficient phases.


Subject(s)
Homologous Recombination/radiation effects , Radiation, Ionizing , Trypanosoma brucei brucei/metabolism , DNA Fragmentation/radiation effects , G1 Phase Cell Cycle Checkpoints/radiation effects , Histones/metabolism , Phosphorylation/radiation effects , Protozoan Proteins/metabolism , Recombinational DNA Repair/radiation effects , Replication Protein A/genetics , Replication Protein A/metabolism , S Phase Cell Cycle Checkpoints/radiation effects , Trypanosoma brucei brucei/radiation effects
20.
Oncology ; 94(6): 383-393, 2018.
Article in English | MEDLINE | ID: mdl-29539615

ABSTRACT

OBJECTIVE: Ewing sarcoma (ES) is a type of childhood cancer probably arising from stem mesenchymal or neural crest cells. The epidermal growth factor receptor (EGFR) acts as a driver oncogene in many types of solid tumors. However, its involvement in ES remains poorly understood. METHODS: Human SK-ES-1 and RD-ES ES cells were treated with EGF, the EGFR inhibitor tyrphostin (AG1478), or phosphoinositide 3-kinase (PI3K) or extracellular-regulated kinase (ERK)/mitogen-activated kinase (MAPK) inhibitors. Cell proliferation survival, cycle, and senescence were analyzed. The protein content of possible targets of EGFR manipulation was measured by Western blot. RESULTS: Cell proliferation and survival were increased by EGF and inhibited by AG1478. The EGFR inhibitor also altered the cell cycle, inducing arrest in G1 and increasing the sub-G1 population, reduced polyploidy and increased the population of senescent cells. In addition, AG1478 reduced the levels of phosphorylated AKT (p-AKT), ERK, p-ERK, cyclin D1, and brain-derived neurotrophic factor (BDNF), while enhancing p53 levels. Cell proliferation was also impaired by inhibitors of PI3K or ERK, alone or combined with AG1478. CONCLUSIONS: Our findings reveal novel aspects of EGFR regulation of ES cells and provide early evidence for antitumor activities of EGFR inhibitors in ES.


Subject(s)
Bone Neoplasms/pathology , Cell Proliferation/drug effects , Enzyme Inhibitors/pharmacology , ErbB Receptors/metabolism , Quinazolines/pharmacology , Sarcoma, Ewing/pathology , Tyrphostins/pharmacology , Brain-Derived Neurotrophic Factor/metabolism , Cell Line, Tumor , Cyclin D1/metabolism , Epidermal Growth Factor/pharmacology , ErbB Receptors/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , G1 Phase Cell Cycle Checkpoints/drug effects , Humans , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation/drug effects , Signal Transduction/drug effects , Tumor Suppressor Protein p53/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL