Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 833
Filtrar
1.
Toxicol Appl Pharmacol ; 484: 116866, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38367674

RESUMO

BACKGROUND: ABC transporter-mediated multidrug resistance (MDR) remains a major obstacle for cancer pharmacological treatment. Some tyrosine kinase inhibitors (TKIs) have been shown to reverse MDR. The present study was designed to evaluate for the first time whether foretinib, a multitargeted TKI, can circumvent ABCB1 and ABCG2-mediated MDR in treatment-resistant cancer models. METHODS: Accumulation of fluorescent substrates of ABCB1 and ABCG2 in ABCB1-overexpressing MES-SA/DX5 and ABCG2-overexpressing MCF-7/MX and their parenteral cells was evaluated by flow cytometry. The growth inhibitory activity of single and combination therapy of foretinib and chemotherapeutic drugs on MDR cells was examined by MTT assay. Analysis of combined interaction effects was performed using CalcuSyn software. RESULTS: It was firstly proved that foretinib increased the intracellular accumulation of rhodamine 123 and mitoxantrone in MES-SA/DX5 and MCF-7/MX cancer cells, with accumulation ratios of 12 and 2.2 at 25 µM concentration, respectively. However, it did not affect the accumulation of fluorescent substrates in the parental cells. Moreover, foretinib synergistically improved the cytotoxic effects of doxorubicin and mitoxantrone. The means of combination index (CI) values at fraction affected (Fa) values of 0.5, 0.75, and 0.9 were 0.64 ± 0.08 and 0.47 ± 0.09, in MES-SA/DX5 and MCF-7/MX cancer cells, respectively. In silico analysis also suggested that the drug-binding domain of ABCB1 and ABCG2 transporters could be considered as potential target for foretinib. CONCLUSION: Overall, our results suggest that foretinib can target MDR-linked ABCB1 and ABCG2 transporters in clinical cancer therapy.


Assuntos
Anilidas , Antineoplásicos , Neoplasias , Quinolinas , Humanos , Proteínas Proto-Oncogênicas c-met/farmacologia , Mitoxantrona/farmacologia , Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP , Resistencia a Medicamentos Antineoplásicos , Resistência a Múltiplos Medicamentos , Antineoplásicos/uso terapêutico , Neoplasias/tratamento farmacológico , Linhagem Celular Tumoral , Proteínas de Neoplasias , Subfamília B de Transportador de Cassetes de Ligação de ATP
2.
Mol Biol Rep ; 51(1): 90, 2024 Jan 09.
Artigo em Inglês | MEDLINE | ID: mdl-38194158

RESUMO

BACKGROUND: CDC25B, as a member of the cell cycle regulating protein family, is located in the cytoplasm and is involved in the transition of the cell cycle and mitosis. CDC25B is highly expressed in various tumors and is a newly discovered oncogene. This study aimed to investigate the impact of CDC25B on mitoxantrone resistance in stomach adenocarcinoma (STAD) and its possible mechanisms. METHODS: This study analyzed the expression of CDC25B and its potential transcription factor E2F3 in STAD, as well as the IC50 values of tumor tissues by bioinformatics analysis. Expression levels of CDC25B and E2F3 in STAD cells were measured by qRT-PCR. MTT was utilized to evaluate cell viability and IC50 values of STAD cells, and comet assay was utilized to analyze the level of DNA damage in STAD cells. Western blot was used to analyze the expression of DNA damage-related proteins. The targeting relationship between E2F3 and CDC25B was validated by dual-luciferase and ChIP assays. RESULTS: Bioinformatics analysis and molecular experiments showed that CDC25B and E2F3 were highly expressed in STAD, and CDC25B was enriched in the mismatch repair and nucleotide excision repair pathways. The IC50 values of tumor tissues with high expression of CDC25B were relatively high. Dual-luciferase and ChIP assays confirmed that CDC25B could be transcriptionally activated by E2F3. Cell experiments revealed that CDC25B promoted mitoxantrone resistance in STAD cells by regulating DNA damage. Further research found that low expression of E2F3 inhibited mitoxantrone resistance in STAD cells by DNA damage, but overexpression of CDC25B reversed the impact of E2F3 knockdown on mitoxantrone resistance in STAD cells. CONCLUSION: This study confirmed a novel mechanism by which E2F3/CDC25B mediated DNA damage to promote mitoxantrone resistance in STAD cells, providing a new therapeutic target for STAD treatment.


Assuntos
Adenocarcinoma , Neoplasias Gástricas , Humanos , Mitoxantrona/farmacologia , Neoplasias Gástricas/tratamento farmacológico , Neoplasias Gástricas/genética , Adenocarcinoma/tratamento farmacológico , Adenocarcinoma/genética , Dano ao DNA , Mitose , Luciferases , Fator de Transcrição E2F3 , Fosfatases cdc25/genética
3.
Mol Oncol ; 18(2): 280-290, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-37727134

RESUMO

Success of chemotherapy is often hampered by multidrug resistance. One mechanism for drug resistance is the elimination of anticancer drugs through drug transporters, such as breast cancer resistance protein (BCRP; also known as ABCG2), and causes a poor 5-year survival rate of human patients. Co-treatment of chemotherapeutics and natural compounds, such as baicalein, is used to prevent chemotherapeutic resistance but is limited by rapid metabolism. Boron-based clusters as meta-carborane are very promising phenyl mimetics to increase target affinity; we therefore investigated the replacement of a phenyl ring in baicalein by a meta-carborane to improve its affinity towards the human ABCG2 efflux transporter. Baicalein strongly inhibited the ABCG2-mediated efflux and caused a fivefold increase in mitoxantrone cytotoxicity. Whereas the baicalein derivative 5,6,7-trimethoxyflavone inhibited ABCG2 efflux activity in a concentration of 5 µm without reversing mitoxantrone resistance, its carborane analogue 5,6,7-trimethoxyborcalein significantly enhanced the inhibitory effects in nanomolar ranges (0.1 µm) and caused a stronger increase in mitoxantrone toxicity reaching similar values as Ko143, a potent ABCG2 inhibitor. Overall, in silico docking and in vitro studies demonstrated that the modification of baicalein with meta-carborane and three methoxy substituents leads to an enhanced reversal of ABCG2-mediated drug resistance. Thus, this seems to be a promising basis for the development of efficient ABCG2 inhibitors.


Assuntos
Antineoplásicos , Flavanonas , Mitoxantrona , Humanos , Mitoxantrona/farmacologia , Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP/genética , Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP/metabolismo , Resistencia a Medicamentos Antineoplásicos , Proteínas de Neoplasias/metabolismo , Antineoplásicos/farmacologia
4.
DNA Repair (Amst) ; 133: 103606, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38039951

RESUMO

Mitoxantrone (1,4-dihydroxy-5,8-bis[2-(2-hydroxyethylamino)ethylamino]-anthracene-9,10-dione) is a clinically-relevant synthetic anthracenedione that functions as a topoisomerase II poison by trapping DNA double-strand break intermediates. Mitoxantrone binds to DNA via both stacking interactions with DNA bases and hydrogen bonding with the sugar-phosphate backbone. It has been shown that mitoxantrone inhibits apurinic/apyrimidinic (AP) endonuclease 1 (APE1)-catalyzed incision of DNA containing a tetrahydrofuran (THF) moiety and more recently, that mitoxantrone forms Schiff base conjugates at AP sites in DNA. In this study, mitoxantrone-mediated inhibition of APE1 at THF sites was shown to be consistent with preferential binding to, and thermal stabilization of DNA containing a THF site as compared to non-damaged DNA. Investigations into the properties of mitoxantrone at AP and 3' α,ß-unsaturated aldehyde sites demonstrated that in addition to being a potent inhibitor of APE1 at these biologically-relevant substrates (∼ 0.5 µM IC50 on AP site-containing DNA), mitoxantrone also incised AP site-containing DNA by catalyzing ß- and ß/δ-elimination reactions. The efficiency of these reactions to generate the 3' α,ß-unsaturated aldehyde and 3' phosphate products was modulated by DNA structure. Although these cell-free reactions revealed that mitoxantrone can generate 3' phosphates, cells lacking polynucleotide kinase phosphatase did not show increased sensitivity to mitoxantrone treatment. Consistent with its ability to inhibit APE1 activity on DNAs containing either an AP site or a 3' α,ß-unsaturated aldehyde, combined exposures to clinically-relevant concentrations of mitoxantrone and a small molecule APE1 inhibitor revealed additive cytotoxicity. These data suggest that in a cellular context, mitoxantrone may interfere with APE1 DNA repair functions.


Assuntos
DNA , Mitoxantrona , Mitoxantrona/farmacologia , DNA/metabolismo , Reparo do DNA , Aldeídos , Fosfatos , Endonucleases/metabolismo , DNA Liase (Sítios Apurínicos ou Apirimidínicos)/metabolismo
5.
Mol Inform ; 43(3): e202300284, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38123523

RESUMO

Tuberculosis (TB) is the second leading cause of mortality after COVID-19, with a global death toll of 1.6 million in 2021. The escalating situation of drug-resistant forms of TB has threatened the current TB management strategies. New therapeutics with novel mechanisms of action are urgently required to address the current global TB crisis. The essential mycobacterial primase DnaG with no structural homology to homo sapiens presents itself as a good candidate for drug targeting. In the present study, Mitoxantrone and Vapreotide, two FDA-approved drugs, were identified as potential anti-mycobacterial agents. Both Mitoxantrone and Vapreotide exhibit a strong Minimum Inhibitory Concentration (MIC) of ≤25µg/ml against both the virulent (M.tb-H37Rv) and avirulent (M.tb-H37Ra) strains of M.tb. Extending the validations further revealed the inhibitory potential drugs in ex vivo conditions. Leveraging the computational high-throughput multi-level docking procedures from the pool of ~2700 FDA-approved compounds, Mitoxantrone and Vapreotide were screened out as potential inhibitors of DnaG. Extensive 200 ns long all-atoms molecular dynamic simulation of DnaGDrugs complexes revealed that both drugs bind strongly and stabilize the DnaG during simulations. Reduced solvent exposure and confined motions of the active centre of DnaG upon complexation with drugs indicated that both drugs led to the closure of the active site of DnaG. From this study's findings, we propose Mitoxantrone and Vapreotide as potential anti-mycobacterial agents, with their novel mechanism of action against mycobacterial DnaG.


Assuntos
Mycobacterium tuberculosis , Somatostatina/análogos & derivados , Humanos , Antituberculosos/farmacologia , DNA Primase/química , DNA Primase/metabolismo , Mitoxantrona/farmacologia
6.
Biomed Pharmacother ; 170: 116038, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38141281

RESUMO

Cholangiocarcinomas (CCAs) are cancers originated in the biliary tree, which are characterized by their high mortality and marked chemoresistance, partly due to the activity of ATP-binding cassette (ABC) export pumps, whose inhibition has been proposed as a strategy for enhancing the response to chemotherapy. We have previously shown that ß-caryophyllene oxide (CRYO) acts as a chemosensitizer in hepatocellular carcinoma by inhibiting ABCB1, MRP1, and MRP2. Here, we have evaluated the usefulness of CRYO in inhibiting BCRP and improving the response of CCA to antitumor drugs. The TCGA-CHOL cohort (n = 36) was used for in silico analysis. BCRP expression (mRNA and protein) was assayed in samples from intrahepatic (iCCA) and extrahepatic (eCCA) tumors (n = 50) and CCA-derived cells (EGI-1 and TFK-1). In these cells, BCRP-dependent mitoxantrone transport was determined by flow cytometry. At non-toxic concentrations, CRYO inhibited BCRP function, which enhanced the cytostatic effect of drugs used in the treatment of CCA. The BCRP ability to confer resistance to a panel of antitumor drugs was determined in Chinese hamster ovary (CHO) cells with stable BCRP expression. At non-toxic concentrations, CRYO markedly reduced BCRP-induced resistance to known substrate drugs (mitoxantrone and SN-38) and cisplatin, gemcitabine, sorafenib, and 5-FU but not oxaliplatin. Neither CRYO nor cisplatin alone significantly affected the growth of BCRP-expressing tumors subcutaneously implanted in immunodeficient mice. In contrast, intratumor drug content was enhanced when administered together, and tumor growth was inhibited. In sum, the combined treatment of drugs exported by BCRP with CRYO can improve the response to chemotherapy in CCA patients.


Assuntos
Antineoplásicos , Carcinoma Hepatocelular , Colangiocarcinoma , Neoplasias Hepáticas , Cricetinae , Humanos , Camundongos , Animais , Cisplatino/farmacologia , Mitoxantrona/farmacologia , Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP/genética , Células CHO , Resistencia a Medicamentos Antineoplásicos , Transportadores de Cassetes de Ligação de ATP , Proteínas de Neoplasias/metabolismo , Cricetulus , Antineoplásicos/farmacologia , Colangiocarcinoma/tratamento farmacológico , Linhagem Celular Tumoral
7.
Med Oncol ; 40(11): 337, 2023 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-37864019

RESUMO

Cervical cancer remains a significant global health concern that starts in the cervix, the lower part of the uterus that connects to the vagina and is caused by the human papillomavirus (HPV), necessitating the development of effective multitargeted effective and resistance-proof therapies. In early-stage cervical cancer may not show any symptoms, however, as the cancer progresses, some people may experience- abnormal vaginal bleeding, watery or bloody vaginal discharge, pain in the pelvis or lower back, pain during sex, and frequent and painful urination. In this study, we screened the complete FDA-approved drug library using a multitargeted inhibitory approach against four cervical cancer proteins, namely mitotic arrest deficient -2, DNA polymerase epsilon B-subunit, benzimidazole-related -1, and threonine-protein kinase-1 which crucially plays its role for the in its development process. We employed the HTVS, SP and XP algorithms for efficient filtering and screening that helped to identify Mitoxantrone 2HCl against all of them with docking and MM\GBSA scores ranging from - 11.63 to - 7.802 kcal/mol and - 74.38 to - 47.73 kcal/mol, respectively. We also evaluated the interaction patterns of each complex and the pharmacokinetics properties that helped gain insight into interactions. Subsequently, we performed multiscale MD simulations for 100 ns to understand the dynamic behaviour and stability of the Mitoxantrone 2HCl -protein complexes that revealed the formation of stable drug-protein complexes and provided insights into the molecular interactions that contribute to Mitoxantrone's inhibitory effects on these proteins and can be a better drug for cervical cancer. However, experimental studies of these findings could pave the way for therapies to combat cervical cancer effectively.


Assuntos
Simulação de Dinâmica Molecular , Neoplasias do Colo do Útero , Humanos , Feminino , Simulação de Acoplamento Molecular , Mitoxantrona/farmacologia , Neoplasias do Colo do Útero/tratamento farmacológico , Proteínas de Ciclo Celular , Dor
8.
Bioorg Med Chem Lett ; 94: 129465, 2023 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-37669721

RESUMO

Mitoxantrone (MX) is a robust chemotherapeutic with well-characterized applications in treating certain leukemias and advanced breast and prostate cancers. The canonical mechanism of action associated with MX is its ability to intercalate DNA and inhibit topoisomerase II, giving it the designation of a topoisomerase II poison. Years after FDA approval, investigations have unveiled novel protein-binding partners, such as methyl-CpG-binding domain protein (MBD2), PIM1 serine/threonine kinase, RAD52, and others that may contribute to the therapeutic profile of MX. Moreover, recent proteomic studies have revealed MX's ability to modulate protein expression, illuminating the complex cellular interactions of MX. Although mechanistically relevant, the differential expression across the proteome does not address the direct interaction with potential binding partners. Identification and characterization of these MX-binding cellular partners will provide the molecular basis for the alternate mechanisms that influence MX's cytotoxicity. Here, we describe the design and synthesis of a MX-biotin probe (MXP) and negative control (MXP-NC) that can be used to define MX's cellular targets and expand our understanding of the proteome-wide profile for MX. In proof of concept studies, we used MXP to successfully isolate a recently identified protein-binding partner of MX, RAD52, in a cell lysate pulldown with streptavidin beads and western blotting.


Assuntos
Mitoxantrona , Humanos , Masculino , DNA Topoisomerases Tipo II , Proteínas de Ligação a DNA , Mitoxantrona/farmacologia , Neoplasias da Próstata/tratamento farmacológico , Proteoma , Proteômica , Sondas Moleculares/química , Sondas Moleculares/farmacologia , Neoplasias da Mama/tratamento farmacológico , Feminino
9.
Biomolecules ; 13(6)2023 05 31.
Artigo em Inglês | MEDLINE | ID: mdl-37371499

RESUMO

Doxorubicin (DOX) and mitoxantrone (MTX) are classical chemotherapeutic agents used in cancer that induce similar clinical cardiotoxic effects, although it is not clear if they share similar underlying molecular mechanisms. We aimed to assess the effects of DOX and MTX on the cardiac remodeling, focusing mainly on metabolism and autophagy. Adult male CD-1 mice received pharmacologically relevant cumulative doses of DOX (18 mg/kg) and MTX (6 mg/kg). Both DOX and MTX disturbed cardiac metabolism, decreasing glycolysis, and increasing the dependency on fatty acids (FA) oxidation, namely, through decreased AMP-activated protein kinase (AMPK) and glyceraldehyde-3-phosphate dehydrogenase (GAPDH) content and decreased free carnitine (C0) and increased acetylcarnitine (C2) concentration. Additionally, DOX heavily influenced glycolysis, oxidative metabolism, and amino acids turnover by exclusively decreasing phosphofructokinase (PFKM) and electron transfer flavoprotein-ubiquinone oxidoreductase (ETFDH) content, and the concentration of several amino acids. Conversely, both drugs downregulated autophagy given by the decreased content of autophagy protein 5 (ATG5) and microtubule-associated protein light chain 3 (LC3B), with MTX having also an impact on Beclin1. These results emphasize that DOX and MTX modulate cardiac remodeling differently, despite their clinical similarities, which is of paramount importance for future treatments.


Assuntos
Antineoplásicos , Mitoxantrona , Masculino , Camundongos , Animais , Mitoxantrona/farmacologia , Mitoxantrona/metabolismo , Remodelação Ventricular , Antineoplásicos/farmacologia , Doxorrubicina/farmacologia , Doxorrubicina/metabolismo , Autofagia , Aminoácidos/metabolismo , Miócitos Cardíacos/metabolismo , Apoptose , Estresse Oxidativo
10.
Naunyn Schmiedebergs Arch Pharmacol ; 396(12): 3723-3732, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-37310508

RESUMO

Cancer endocrine therapy can promote evolutionary dynamics and lead to changes in the gene expression profile of tumor cells. We aimed to assess the effect of tamoxifen (TAM)-resistance induction on ABCG2 pump mRNA, protein, and activity in ER + MCF-7 breast cancer cells. We also evaluated if the resistance to TAM leads to the cross-resistance toward mitoxantrone (MX), a well-known substrate of the ABCG2 pump. The ABCG2 mRNA and protein expression were compared in MCF-7 and its TAM-resistant derivative MCF-7/TAMR cells using RT-qPCR and western blot methods, respectively. Cross-resistance of MCF-7/TAMR cells toward MX was evaluated by the MTT method. Flow cytometry was applied to compare ABCG2 function between cell lines using MX accumulation assay. ABCG2 mRNA expression was also analyzed in tamoxifen-sensitive (TAM-S) and tamoxifen-resistant (TAM-R) breast tumor tissues. The levels of ABCG2 mRNA, protein, and activity were significantly higher in MCF-7/TAMR cells compared to TAM-sensitive MCF-7 cells. MX was also less toxic in MCF-7/TAMR compared to MCF-7 cells. ABCG2 was also upregulated in tissue samples obtained from TAM-R cancer patients compared to TAM-S patients. Prolonged exposure of ER + breast cancer cells to the active form of TAM and clonal evolution imposed by the selective pressure of the drug can lead to higher expression of the ABCG2 pump in the emerged TAM-resistant cells. Therefore, in choosing a sequential therapy for a patient who develops resistance to TAM, the possibility of the cross-resistance of the evolved tumor to chemotherapy drugs that are ABCG2 substrates should be considered. Prolonged exposure of MCF-7 breast cancer cells to tamoxifen can cause resistance to it and an increase in the expression of the ABCG2 mRNA and protein levels in the cells. Tamoxifen resistance can lead to cross-resistance to mitoxantrone.


Assuntos
Neoplasias da Mama , Tamoxifeno , Humanos , Feminino , Tamoxifeno/farmacologia , Tamoxifeno/uso terapêutico , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Mitoxantrona/farmacologia , Mitoxantrona/uso terapêutico , Células MCF-7 , Regulação para Cima , Antineoplásicos Hormonais/farmacologia , Antineoplásicos Hormonais/uso terapêutico , Resistencia a Medicamentos Antineoplásicos , RNA Mensageiro/metabolismo , Regulação Neoplásica da Expressão Gênica , Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP/genética , Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP/metabolismo , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Proteínas de Neoplasias/farmacologia
11.
Apoptosis ; 28(11-12): 1534-1545, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-37243774

RESUMO

Target-negative relapse after CD19 chimeric antigen receptor engineered (CAR) T cell therapy for patients with B lineage acute lymphoblastic leukemia (B-ALL) presents limited treatment options with dismal outcomes. Although CD22-CAR T cells mediate similarly potent antineoplastic effects in patients with CD19dim or even CD19-negative relapse following CD19-directed immunotherapy, a high rate of relapse associated with diminished CD22 cell surface expression has also been observed. Therefore, it is unclear whether any other therapeutic options are available. Mitoxantrone has shown significant antineoplastic activity in patients with relapsed or refractory leukemia over the past decades, and in some cases, the addition of bortezomib to conventional chemotherapeutic agents has demonstrated improved response rates. However, whether this mitoxantrone and bortezomib combination therapy is effective for those patients who have relapsed B-ALL after receiving CD19-CAR T cell therapy remains to be elucidated. In this study, we established a cellular model system using a CD19-positive B-ALL cell line Nalm-6 to investigate the treatment options for CD19-negative relapsed B-ALL after CD19-CAR T cell therapy. In addition to CD22-CAR T therapy, we observed that the combination of bortezomib and mitoxantrone exhibited effective anti-leukemia activity in the CD19-negative Nalm-6 cell line by downregulating p-AKT and p-mTOR. These results suggest that this combination therapy is a possible option for target-negative refractory leukemia cells after CAR-T cell treatment.


Assuntos
Antineoplásicos , Leucemia-Linfoma Linfoblástico de Células Precursoras , Receptores de Antígenos Quiméricos , Humanos , Imunoterapia Adotiva/efeitos adversos , Imunoterapia Adotiva/métodos , Mitoxantrona/farmacologia , Mitoxantrona/uso terapêutico , Bortezomib/farmacologia , Bortezomib/uso terapêutico , Apoptose , Recidiva , Linhagem Celular , Antígenos CD19 , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Lectina 2 Semelhante a Ig de Ligação ao Ácido Siálico
12.
Genes (Basel) ; 14(5)2023 04 26.
Artigo em Inglês | MEDLINE | ID: mdl-37239338

RESUMO

Occurrence of non-canonical G-quadruplex (G4) DNA structures in the genome have been recognized as key factors in gene regulation and several other cellular processes. The mosR and ndhA genes involved in pathways of oxidation sensing regulation and ATP generation, respectively, make Mycobacterium tuberculosis (Mtb) bacteria responsible for oxidative stress inside host macrophage cells. Circular Dichroism spectra demonstrate stable hybrid G4 DNA conformations of mosR/ndhA DNA sequences. Real-time binding of mitoxantrone to G4 DNA with an affinity constant ~105-107 M-1, leads to hypochromism with a red shift of ~18 nm, followed by hyperchromism in the absorption spectra. The corresponding fluorescence is quenched with a red shift ~15 nm followed by an increase in intensity. A change in conformation of the G4 DNA accompanies the formation of multiple stoichiometric complexes with a dual binding mode. The external binding of mitoxantrone with a partial stacking with G-quartets and/or groove binding induces significant thermal stabilization, ~20-29 °C in ndhA/mosR G4 DNA. The interaction leads to a two/four-fold downregulation of transcriptomes of mosR/ndhA genes apart from the suppression of DNA replication by Taq polymerase enzyme, establishing the role of mitoxantrone in targeting G4 DNA, as an alternate strategy for effective anti-tuberculosis action in view of deadly multi-drug resistant tuberculosis disease causing bacterial strains t that arise from existing therapeutic treatments.


Assuntos
Quadruplex G , Mycobacterium tuberculosis , Mitoxantrona/farmacologia , Mitoxantrona/química , Mycobacterium tuberculosis/genética , Mycobacterium tuberculosis/metabolismo , DNA/genética , Sequência de Bases
13.
J Mol Model ; 29(5): 153, 2023 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-37086344

RESUMO

The PI3K/Akt/mTOR pathway is one of the important pathways in many cancers. Akt is a serine-threonine kinase protein identified as a drug target for cancer treatment. Therefore, anticancer drugs are essential therapeutic targets for this pathway. In the current study, the inhibitory effect of two anticancer molecules, XFE and mitoxantrone, on AKT1 protein that can impact the activity of the AKT1 protein was investigated by using molecular docking and molecular dynamics (MD) simulations. The molecular docking results presented a relatively higher binding affinity of the mitoxantrone molecule in interaction with AKT1 than the XFE molecule. These results were validated by the MM/PBSA technique that was performed on obtained trajectories of 25 ns MD simulations. The mitoxantrone molecule has an intense binding energy of - 880.536 kcal/mol with AKT1 protein, while the XFE molecule shows a binding energy value of - 83.569 kcal/mol. Our findings from molecular dynamics simulations indicated that both molecules have favorite interactions with AKT1 protein. Other analyses, such as RMSF and hydrogen binding on trajectories obtained from MD simulations, indicated that the mitoxantrone molecule could be a relatively potent inhibitor for AKT1. Based on the results of this study and the structure of mitoxantrone, it is expected to be a good candidate for cancer treatment as a (PI3K)/Akt/mTOR inhibitor.


Assuntos
Mitoxantrona , Proteínas Proto-Oncogênicas c-akt , Mitoxantrona/farmacologia , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/química , Proteínas Proto-Oncogênicas c-akt/metabolismo , Serina-Treonina Quinases TOR/metabolismo
14.
IEEE Trans Nanobioscience ; 22(4): 872-880, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37022888

RESUMO

The development nano-carriers based therapeutic methods is a potent strategy for enhancing cellular delivery of drugs and therapeutic efficiency in cancer chemotherapy. In the study, silymarin(SLM) and metformin (Met) were co-loaded into mesoporous silica nanoparticles (MSNs) and evaluated the synergistic inhibitory effect of these natural herbal compound in improving chemotherapeutic efficiency against MCF7MX and MCF7 human breast cancer cells. Nanoparticles have been synthesized and characterized by FTIR, BET, TEM, SEM, and X-ray diffraction. Drug loading capacity and release determined. The both single and combined form of SLM and Met (free and loaded MSN) were used for MTT assay, colony formation and real time-PCR in cellular study. The synthesis MSN were uniformity in size and shape with particle size of approximately 100 nm and pore size of approximately 2 nm. The Met-MSNs IC30, SLM -MSNs IC50 and dual-drug loaded MSNs IC50 were much lower than of free-Met IC30, free-SLM IC50 and free Met-SLM IC50 MCF7MX and MCF7cells. The co-loaded MSNs treated cells were increased sensitivity to mitoxantrone with the inhibition of BCRP mRNA expressions and could induce apoptosis in MCF7MX and MCF7 cells in comparison with other groups. Colony numbers were significantly reduced in comparison to with other groups in the co-loaded MSNs -treated cells ( ). Our results indicate that Nano-SLM enhances the anti-cancer effects of SLM against human breast cancer cells. The findings of the present study suggest that the anti-cancer effects of both metformin and silymarin enhances against breast cancer cells when MSNs are used as a drug delivery system.


Assuntos
Neoplasias da Mama , Nanopartículas , Silimarina , Humanos , Feminino , Células MCF-7 , Mitoxantrona/farmacologia , Dióxido de Silício , Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP , Neoplasias da Mama/tratamento farmacológico , Proteínas de Neoplasias , Sistemas de Liberação de Medicamentos/métodos , Porosidade , Portadores de Fármacos
15.
ChemMedChem ; 18(11): e202300094, 2023 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-36988057

RESUMO

The role of ATP-binding cassette (ABC) transporter-mediated multidrug resistance (MDR) in anti-cancer therapy is often challenging, frequently leading to inefficiency of treatments. Cancer cells exploit efflux transporters, like the breast cancer resistance protein (BCRP, ABCG2), to secrete chemotherapeutic substances. In this study, an N-phenyl-2-carboranylquinazolin-4-amine (8) was designed as inorganic-organic hybrid BCRP inhibitor. In particular, the ABCG2-transporter inhibitor-prominent scaffold N-phenylquinazolin-4-amine was combined with a boron-carbon cluster (carborane) moiety. Introducing a carborane at 2-position of the quinazoline scaffold resulted in an increased inhibitory activity towards human ABCG2 (hABCG2) compared to its recently published regioisomer N-carboranyl-2-phenyl-quinazolin-4-amine. The carboranylquinazoline 8 further showed the ability to reverse hABCG2-mediated drug resistance in MDCKII-hABCG2 cells by lowering the IC50 value of the BCRP-substrate mitoxantrone, similar to the standard reference and strong inhibitor Ko143, without exhibiting intrinsic toxicity in the lower micromolar ranges. These results make compound 8 a promising scaffold for the design of further BCRP inhibitors.


Assuntos
Antineoplásicos , Proteínas de Neoplasias , Humanos , Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP , Transportadores de Cassetes de Ligação de ATP/metabolismo , Transportadores de Cassetes de Ligação de ATP/farmacologia , Mitoxantrona/farmacologia , Resistência a Múltiplos Medicamentos , Resistencia a Medicamentos Antineoplásicos , Antineoplásicos/farmacologia , Antineoplásicos/química
16.
Int J Mol Sci ; 24(6)2023 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-36982905

RESUMO

A collection of repurposing drugs (Prestwick Chemical Library) containing 1200 compounds was screened to investigate the drugs' antimicrobial effects against planktonic cultures of the respiratory pathogen Streptococcus pneumoniae. After four discrimination rounds, a set of seven compounds was finally selected, namely (i) clofilium tosylate; (ii) vanoxerine; (iii) mitoxantrone dihydrochloride; (iv) amiodarone hydrochloride; (v) tamoxifen citrate; (vi) terfenadine; and (vii) clomiphene citrate (Z, E). These molecules arrested pneumococcal growth in a liquid medium and induced a decrease in bacterial viability between 90.0% and 99.9% at 25 µM concentration, with minimal inhibitory concentrations (MICs) also in the micromolar range. Moreover, all compounds but mitoxantrone caused a remarkable increase in the permeability of the bacterial membrane and share a common, minimal chemical structure consisting of an aliphatic amine linked to a phenyl moiety via a short carbon/oxygen linker. These results open new possibilities to tackle pneumococcal disease through drug repositioning and provide clues for the design of novel membrane-targeted antimicrobials with a related chemical structure.


Assuntos
Anti-Infecciosos , Infecções Pneumocócicas , Humanos , Streptococcus pneumoniae , Antibacterianos/farmacologia , Reposicionamento de Medicamentos , Mitoxantrona/farmacologia , Infecções Pneumocócicas/tratamento farmacológico , Anti-Infecciosos/farmacologia , Testes de Sensibilidade Microbiana , Membrana Celular
17.
Bioorg Chem ; 135: 106481, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-36966672

RESUMO

Three series of phenylurea indole derivatives were synthesized with potent inhibitory activities on ABCG2 with simple and efficient synthetic routes. Among these compounds, four phenylurea indole derivatives 3c-3f with extended π system were discovered as the most potent ABCG2 inhibitors, while these compounds showed no inhibition on ABCB1. Compounds 3c and 3f were selected for further investigation to explore the mechanisms of action on reversing ABCG2-mediated multidrug resistance (MDR). The results revealed that compounds 3c and 3f increased the accumulation of mitoxantrone (MX) in ABCG2-overexpressing cells, but they did not alter the expression level or localization of ABCG2 in cells. In addition, both 3c and 3f significantly stimulated the ATP hydrolysis of ABCG2 transporter indicating that they can be competitive substrates of ABCG2 transporter, and thereby increase the accumulation of mitoxantrone in ABCG2-overexpressing H460/MX20 cells. Both 3c and 3f was docked into the drug-binding site of the human ABCG2 transporter protein (PDB 6FFC) with high affinities. This study showed that extending the π system of phenylurea indole derivatives enhanced their inhibitory activities on ABCG2, which may provide a clue for the further research to discover more potent ABCG2 inhibitors.


Assuntos
Antineoplásicos , Humanos , Antineoplásicos/química , Mitoxantrona/farmacologia , Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP/metabolismo , Resistencia a Medicamentos Antineoplásicos , Compostos de Fenilureia/farmacologia , Linhagem Celular Tumoral , Indóis/farmacologia , Proteínas de Neoplasias/metabolismo
18.
Sci Adv ; 9(8): eadd9280, 2023 02 22.
Artigo em Inglês | MEDLINE | ID: mdl-36812322

RESUMO

Antibiotic resistance critically limits treatment options for infection caused by opportunistic pathogens such as enterococci. Here, we investigate the antibiotic and immunological activity of the anticancer agent mitoxantrone (MTX) in vitro and in vivo against vancomycin-resistant Enterococcus faecalis (VRE). We show that, in vitro, MTX is a potent antibiotic against Gram-positive bacteria through induction of reactive oxygen species and DNA damage. MTX also synergizes with vancomycin against VRE, rendering the resistant strains more permeable to MTX. In a murine wound infection model, single-dose MTX treatment effectively reduces VRE numbers, with further reduction when combined with vancomycin. Multiple MTX treatments accelerate wound closure. MTX also promotes macrophage recruitment and proinflammatory cytokine induction at the wound site and augments intracellular bacterial killing in macrophages by up-regulating the expression of lysosomal enzymes. These results show that MTX represents a promising bacterium- and host-targeted therapeutic for overcoming vancomycin resistance.


Assuntos
Enterococcus faecalis , Enterococos Resistentes à Vancomicina , Animais , Camundongos , Enterococcus faecalis/genética , Resistência a Vancomicina/genética , Vancomicina/farmacologia , Mitoxantrona/farmacologia , Antibacterianos/farmacologia , Enterococos Resistentes à Vancomicina/genética
19.
Nanoscale ; 15(4): 1914-1924, 2023 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-36617999

RESUMO

The anti-apoptotic B-cell lymphoma-2 (Bcl-2) family of proteins are critical regulators of cell death that are overexpressed in many cancer cells, especially in multi-drug resistant cancer cells. Combinatorial gene- and chemotherapies using antisense oligonucleotides (ASOs) to suppress the expression of Bcl-2-family mRNA and restore the sensitivity of the cell to chemodrugs provide a promising pathway for anticancer treatment. However, intrinsic differences between macromolecular ASOs and small molecular chemodrugs make their co-delivery challenging. Moreover, extraneous carriers may induce immunogenicity and inflammation problems. Herein, we develop a targeted nanodrug delivery system using the cationic amphiphilic chemodrug mitoxantrone (Mito), which interacts with Bcl-2 ASO through electrostatic interaction and self-assembles into nanoparticles (NP[Bcl-2/Mito]), whose size can be controlled by regulating the ratio of ASO and Mito. NP[Bcl-2/Mito] can protect the ASO from degradation during delivery and combine gene- and chemotherapies to improve the anticancer effect. Furthermore, cancer cell membranes (CCMs) derived from homologous tumors were used to camouflage NP[Bcl-2/Mito] (NP[Bcl-2/Mito]@CCM) to achieve immune escape and tumor targeting. Both in vitro and in vivo assessments demonstrate the excellent performance of NP[Bcl-2/Mito]@CCM for drug-resistant breast tumor therapy. This CCM-camouflaged ASO/chemodrug nanoplatform provides a promising pathway for the targeted delivery of ASOs and chemodrugs for tumor combination therapy.


Assuntos
Neoplasias , Oligonucleotídeos Antissenso , Humanos , Oligonucleotídeos Antissenso/farmacologia , Oligonucleotídeos Antissenso/genética , Resistência a Múltiplos Medicamentos , Resistencia a Medicamentos Antineoplásicos , Oligonucleotídeos , Neoplasias/metabolismo , Mitoxantrona/farmacologia , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo
20.
Photodiagnosis Photodyn Ther ; 41: 103269, 2023 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-36596330

RESUMO

BACKGROUND: Conventional cancer treatments are associated with a number of limitations, including non-selectivity, toxicity and multidrug resistance, so new nanotechnologies are being developed forcancer diagnosis and therapy. Phototherapy approach based on nanotechnology is a hopeful strategy to overcome these problems. Photothermal (PTT) and photodynamic therapies (PDT), in addition to having non-invasive properties, are known as promising methods for treatment of tumors. In this study, CoFe2O4 theranostic magnetic nanoparticles coated with spiky gold nanoparticles were designed and synthesized and its photothermal effects were evaluated in combination with the photodynamic and chemotherapeutic effects of mitoxantrone (MTX) under in vitro conditions. METHODS AND MATERIALS: At first, CoFe2O4 @Spiky Au nanostructure was synthesized and after its characterization, cytotoxicity of MTX, CoFe2O4 @ Spiky Au (MGNS) and CoFe2O4 @ Au were determined on MDA-MB-231 cell line. Then, the concentrations required for inducing 50% cell death (IC50) and appropriate concentration for this study was obtained. Cells were irradiated by an 808 nm laser and a non-synchronous light source at 670 nm at the separate groups. The viability of treated cells was determined via MTT test 48 h after treatment. RESULTS: In the groups receiving energy density (5-40) J/cm2, at the lower laser dose an increase in cell survival was observed (P < 0.05) and then cell survival was decreased (P < 0.05). In the groups receiving non-coherent light (2-18 J/cm2) from the beginning, a decreasing trend in cell survival is observed. CONCLUSION: The overlap of the emission spectrum of the light source and the absorption spectrum of the nanostructure amplified the cell death. Similar to the Hormesis model reported for ionizing radiation effects, at low light doses with the bio-phasic response dose model, increased cell survival and proliferation can be expected.


Assuntos
Neoplasias da Mama , Hipertermia Induzida , Nanopartículas Metálicas , Fotoquimioterapia , Humanos , Feminino , Fotoquimioterapia/métodos , Mitoxantrona/farmacologia , Ouro/química , Neoplasias da Mama/tratamento farmacológico , Fármacos Fotossensibilizantes/uso terapêutico , Linhagem Celular Tumoral , Hipertermia Induzida/métodos , Nanopartículas Metálicas/química , Fototerapia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...