Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
J Biol Inorg Chem ; 2024 Jul 16.
Artigo em Inglês | MEDLINE | ID: mdl-39009790

RESUMO

Lapachol (2-hydroxy-3-(3-methylbut-2-en-1-yl)naphthalene-1,4-dione) is a 1,4-naphthoquinone-derived natural product that presents numerous bioactivities and was shown to have cytotoxic effects against several human tumor cells. Indium(III) complexes with a variety of ligands also exhibit antineoplastic activity. Indium(III) complexes [In(lap)Cl2].4H2O (1), [In(lap)2Cl(Et3N)] (2), [In(lap)3]·2H2O (3) [In(lap)(bipy)Cl2] bipy = 2,2'-bipyridine (4) and [In(lap)(phen)Cl2] phen = 1,10-phenanthroline (5) were obtained with 2-hydroxy-3-(3-methylbut-2-en-1-yl)naphthalene-1,4-dione (lapachol). Crystal structure determinations for (4) and (5) revealed that the indium(III) center is coordinated to two O atoms from lapachol, two N atoms from 1,10-phenanthroline or 2,2'-bipyridine, and two chloride anions, in a distorted octahedral geometry. Although both complexes (4) and (5) interacted with CT-DNA in vitro by an intercalative mode, only 5 exhibited cytotoxicity against MCF-7 and MDA-MB breast tumor cells. 1,10-phenanthroline and complex (5) presented cytotoxic effects against MCF-7 and MDA-MB cells, with complex (5) being threefold more active than 1,10-phenanthroline on MCF-7 cells. In addition, complex (5) significantly reduced the formation of MDA-MB-231 colonies in a clonogenicity assay. The foregoing results suggest that further studies on the cytotoxic effects and cellular targets of complex (5) are of utmost relevance.

2.
Eur J Nucl Med Mol Imaging ; 48(4): 984-994, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33078260

RESUMO

PURPOSE: It was previously demonstrated that radiation effects can enhance the therapy outcome of immune checkpoint inhibitors. In this study, a syngeneic breast tumor mouse model was used to investigate the effect of [177Lu]Lu-DOTA-folate as an immune stimulus to enhance anti-CTLA-4 immunotherapy. METHODS: In vitro and in vivo studies were performed to characterize NF9006 breast tumor cells with regard to folate receptor (FR) expression and the possibility of tumor targeting using [177Lu]Lu-DOTA-folate. A preclinical therapy study was performed over 70 days with NF9006 tumor-bearing mice that received vehicle only (group A); [177Lu]Lu-DOTA-folate (5 MBq; 3.5 Gy absorbed tumor dose; group B); anti-CTLA-4 antibody (3 × 200 µg; group C), or both agents (group D). The mice were monitored regarding tumor growth over time and signs indicating adverse events of the treatment. RESULTS: [177Lu]Lu-DOTA-folate bound specifically to NF9006 tumor cells and tissue in vitro and accumulated in NF9006 tumors in vivo. The treatment with [177Lu]Lu-DOTA-folate or an anti-CTLA-4 antibody had only a minor effect on NF9006 tumor growth and did not substantially increase the median survival time of mice (23 day and 19 days, respectively) as compared with untreated controls (12 days). [177Lu]Lu-DOTA-folate sensitized, however, the tumors to anti-CTLA-4 immunotherapy, which became obvious by reduced tumor growth and, hence, a significantly improved median survival time of mice (> 70 days). No obvious signs of adverse effects were observed in treated mice as compared with untreated controls. CONCLUSION: Application of [177Lu]Lu-DOTA-folate had a positive effect on the therapy outcome of anti-CTLA-4 immunotherapy. The results of this study may open new perspectives for future clinical translation of folate radioconjugates.


Assuntos
Lutécio , Neoplasias , Animais , Linhagem Celular Tumoral , Ácido Fólico , Imunoterapia , Lutécio/uso terapêutico , Camundongos , Radioisótopos , Compostos Radiofarmacêuticos
3.
Adv Biol (Weinh) ; : e2400184, 2024 Jul 06.
Artigo em Inglês | MEDLINE | ID: mdl-38971965

RESUMO

Triple-negative breast cancer (TNBC) is the most invasive type of breast cancer with high risk of brain metastasis. To better understand interactions between breast tumors with the brain extracellular matrix (ECM), a 3D cell culture model is implemented using a thiolated hyaluronic acid (HA-SH) based hydrogel. The latter is used as HA represents a major component of brain ECM. Melt-electrowritten (MEW) scaffolds of box- and triangular-shaped polycaprolactone (PCL) micro-fibers for hydrogel reinforcement are utilized. Two different molecular weight HA-SH materials (230 and 420 kDa) are used with elastic moduli of 148 ± 34 Pa (soft) and 1274 ± 440 Pa (stiff). Both hydrogels demonstrate similar porosities. The different molecular weight of HA-SH, however, significantly changes mechanical properties, e.g., stiffness, nonlinearity, and hysteresis. The breast tumor cell line MDA-MB-231 forms mainly multicellular aggregates in both HA-SH hydrogels but sustains high viability (75%). Supplementation of HA-SH hydrogels with ECM components does not affect gene expression but improves cell viability and impacts cellular distribution and morphology. The presence of other brain cell types further support numerous cell-cell interactions with tumor cells. In summary, the present 3D cell culture model represents a novel tool establishing a disease cell culture model in a systematic way.

4.
Small ; 7(17): 2507-16, 2011 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-21739601

RESUMO

Luminescent silicon nanocrystals (ncSi) are showing great promise as photoluminescent tags for biological fluorescence imaging, with size-dependent emission that can be tuned into the near-infrared biological window and reported lack of toxicity. Here, colloidally stable ncSi with NIR photoluminescence are synthesized from (HSiO1.5)n sol-gel glasses and are used in biological fluorescence imaging. Modifications to the thermal processing conditions of (HSiO1.5)n sol-gel glasses, the development of new ncSi oxide liberation chemistry, and an appropriate alkyl surface passivation scheme lead to the formation of colloidally stable ncSi with photoluminescence centered at 955 nm. Water solubility and biocompatibility are achieved through encapsulation of the hydrophobic alkyl-capped ncSi within PEG-terminated solid lipid nanoparticles. Their applicability to biological imaging is demonstrated with the in-vitro fluorescence labelling of human breast tumor cells.


Assuntos
Nanopartículas , Imagem Óptica/métodos , Silício , Neoplasias da Mama/diagnóstico , Linhagem Celular Tumoral , Coloides , Feminino , Humanos , Luminescência , Microscopia Eletrônica de Transmissão , Nanopartículas/química , Nanopartículas/ultraestrutura , Pontos Quânticos/química , Pontos Quânticos/ultraestrutura , Silício/química , Espectroscopia de Luz Próxima ao Infravermelho
5.
Biomed Mater ; 16(4)2021 06 17.
Artigo em Inglês | MEDLINE | ID: mdl-34020433

RESUMO

Elimination of tumor cells is still a therapeutic challenge for breast cancer (BC) in men and women. Mammospheres serve as valuablein vitrotools for evaluating tumor behavior and sensitivity to anticancer treatments. Graphene nanosheets with unique physicochemical properties have been considered as potential biomedical approaches for drug delivery, bioimaging, and therapy. Graphene oxide (GO) and graphene quantum dots (GQDs) are suitable nanocarriers for hydrophobic and low bioaccessible anti-tumor materials like curcumin. Despite extensive studies on the potential application of graphene nanosheets in medicine, our knowledge of how different cells function and respond to these nanoparticles remains limited. Here, we evaluated cell death in mammospheres from MCF-7 and primary tumor cells in response to curcumin loaded on graphene nanosheets. Mammospheres were exposed to graphene oxide-curcumin (GO-Cur) and graphene quantum dots-curcumin (GQDs-Cur), and the incidence of cell death was evaluated by Hoechst 33342/propidium iodide double staining and flow cytometry. Besides, the expression of miR-21, miR-29a, Bax, and Bcl-2 genes were assessed using RT-qPCR. We observed, GO, and GQDs had no cytotoxic effect on Kerman male breast cancer/71 (KMBC/71) and MCF-7 tumor cells, while curcumin induced death in more than 50% of tumor cells. GO-Cur and GQDs-Cur synergistically enhanced anti-tumor activity of curcumin. Moreover, GQDs-Cur induced cell death in almost all cells of KMBC/71 mammospheres (99%;p< 0.0001). In contrast, GO-Cur induced cell death in only 21% of MCF-7 mammosphere cells (p< 0.0001). Also, the expression pattern of miR-21, miR-29a, and Bax/Bcl-2 ratio in KMBC/71 and MCF-7 mammospheres was different in response to GO-Cur and GQDs-Cur. Although KMBC/71 and MCF-7 tumor cells had similar clinical features and displayed similar responses to curcumin, more investigations are needed to clarify the detailed molecular mechanisms underlying observed differences in response to GO-Cur and GQDs-Cur.


Assuntos
Apoptose/efeitos dos fármacos , Neoplasias da Mama/metabolismo , Curcumina , Grafite/química , Pontos Quânticos , Sobrevivência Celular/efeitos dos fármacos , Curcumina/química , Curcumina/farmacologia , Feminino , Humanos , Células MCF-7 , Masculino , Esferoides Celulares/efeitos dos fármacos , Células Tumorais Cultivadas
6.
J Biomed Mater Res B Appl Biomater ; 108(3): 948-957, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31389180

RESUMO

Sonodynamic therapy (SDT) which employs ultrasound-triggered sonosensitizers to generate reactive oxygen species (ROS) has been proved to be effective for treatment of cancers. However, it is still desirable for sonosensitizers to be delivered to tumors as effectively as possible. In this study, we prepared the hematoporphyrin monomethyl ether (HMME)-loaded liposome as the sonosensitizers for SDT and evaluated their effects on human MCF-7 breast cancer cells in vitro and in vivo. Liposomes prepared by thin film hydration technique were about 100 nm in size with positive zeta potential and exhibited spherical in shape. Following irradiation of ultrasound which generates intracellular ROS, the liposome facilitated the delivery of HMME to tumor cells. HMME-loaded liposomes showed low cytotoxicity under basal condition but significant sonodynamic effects under ultrasonic irradiation. Notably, HMME-loaded liposomes exhibited spatial distribution of HMME in tumor tissues of mice. The promoted delivery of HMME into the tumors by liposomes was shown by the greater tumor growth inhibition than free HMME after 20-day treatment. Taken together, these results show that HMME-loaded liposome functions as a promising sonosensitizer for SDT, implying the efficient antitumor effects of HMME-based SDT on breast tumor.

7.
Int Immunopharmacol ; 84: 106514, 2020 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-32311671

RESUMO

The non-neuronal cholinergic system refers to the presence of acetylcholine, choline acetyltransferase, acetylcholinesterase and cholinergic receptors, nicotinic and muscarinic (mAChRs) expressed in non-neuronal cells. The presence of mAChRs has been detected in different type of tumor cells and they are linked with tumorigenesis. We had previously documented the expression of mAChRs in murine and human mammary adenocarcinomas and the absence of these receptors in normal mammary cells of the same origins. We also demonstrated that mAChRs are involved in breast cancer progression, pointing to a main role for mAChRs as oncogenic proteins. Since the long term treatment of breast cancer cells with the muscarinic agonist carbachol promoted cell death, here we investigated the ability of low doses of this agonist combined with paclitaxel (PX), a taxane usually administered to treat breast cancer, to inhibit the progression of human MCF-7 tumor cells. We demonstrated that PX plus carbachol reduced cell viability and tumor growth in vitro probably due to a down-regulation in cancer stem cells population and in the expression of ATP "binding cassette" G2 drug extrusion pump; also a reduction in malignant-induced angiogenesis was produced by the in vivo administration of the mentioned combination in a metronomic schedule to MCF-7 tumor-bearing NUDE mice. Our results confirm that mAChRs could be considered as therapeutic targets for metronomic therapy in breast cancer as well as the usefulness of a muscarinic agonist as repositioning drug in the treatment of this type of tumors.


Assuntos
Antineoplásicos Fitogênicos/administração & dosagem , Carbacol/administração & dosagem , Agonistas Colinérgicos/administração & dosagem , Neoplasias Mamárias Experimentais/tratamento farmacológico , Neovascularização Patológica/tratamento farmacológico , Paclitaxel/administração & dosagem , Receptores Muscarínicos/metabolismo , Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP/metabolismo , Administração Metronômica , Animais , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Interações Medicamentosas , Feminino , Humanos , Glândulas Mamárias Animais/irrigação sanguínea , Glândulas Mamárias Animais/efeitos dos fármacos , Glândulas Mamárias Animais/metabolismo , Glândulas Mamárias Animais/patologia , Neoplasias Mamárias Experimentais/metabolismo , Neoplasias Mamárias Experimentais/patologia , Camundongos Nus , Neovascularização Patológica/metabolismo , Neovascularização Patológica/patologia
8.
Biochim Biophys Acta Biomembr ; 1860(11): 2155-2165, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30409511

RESUMO

Antimicrobial peptides have been extensively described as bioactive agents, mainly considering their selective toxicity towards bacteria but not to healthy mammalian cells. In past years, this class of compounds has been classified as an attractive and novel family of anticancer agents. Pantinin peptides isolated from scorpion Pandinus imperator presented antimicrobial activity. In this study, we have explored the in vitro antitumor activity of antimicrobial pantinin peptides against the tumor cell lines MDA-MB-231 (breast adenocarcinoma) and DU - 145 (prostate adenocarcinoma) and healthy fibroblasts HGF - 1. To further improve our mechanistic understanding for this class of compounds, we have also performed a biophysical characterization of these peptides in lipid model membranes. Cell viability assays revealed that all peptides were more effective on tumor cells when compared to fibroblasts, indicating selectivity towards cancer cells. Furthermore, flow cytometry analysis revealed that all peptides induced apoptosis in cancer cells in a different way from fibroblasts. Circular dichroism spectroscopy showed that all peptides adopted an α-helical structure and an evaluation of the binding constant indicates a higher affinity of the peptides to negatively charged phospholipids. Additionally, permeabilization assays showed that POPG and POPS anionic vesicles were more susceptible to peptide-induced lysis than POPC:Chol and POPC:POPE vesicles. Moreover, we have observed that increasing concentrations of cholesterol inhibits peptide binding process. Therefore, our findings suggest that Pantinin peptides may have chemotherapeutic potential for cancer treatment.


Assuntos
Peptídeos Catiônicos Antimicrobianos/farmacologia , Antineoplásicos/farmacologia , Venenos de Escorpião/química , Peptídeos Catiônicos Antimicrobianos/química , Peptídeos Catiônicos Antimicrobianos/isolamento & purificação , Antineoplásicos/química , Antineoplásicos/isolamento & purificação , Apoptose/efeitos dos fármacos , Fenômenos Biofísicos , Varredura Diferencial de Calorimetria , Linhagem Celular Tumoral , Permeabilidade da Membrana Celular/efeitos dos fármacos , Dicroísmo Circular , Citometria de Fluxo , Humanos , Estrutura Secundária de Proteína
9.
J Steroid Biochem Mol Biol ; 138: 54-62, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23562642

RESUMO

About 70% of breast tumors express androgen receptors. In addition, there is clinical evidence suggesting that androgens can inhibit mammary epithelial proliferation. Vice versa, there is also significant evidence indicating that androgens can increase the risk of breast cancer via multiple mechanisms, e.g. direct conversion to estrogens that can bind to the estrogen receptor and thereby stimulate cell proliferation. We examined the effect of testosterone (T) and dihydroxytestosterone (DHT) on cell proliferation, pS2 and Ki-67 expression in three different breast cancer cell lines alone or in co-culture with primary human breast adipose fibroblasts (BAFs) obtained from breast cancer patients. In the co-cultures, T induced cell proliferation, pS2 and Ki-67 expression in the estrogen receptor positive (ER(+)) MCF-7 and T47D cells. This was not observed in the (ER(-)) MDA-MB-231 cells. The differences might be explained by the high expression of aromatase, which converts androgens to estrogens in BAFs followed by ER-mediated cell proliferation. In line with this absence of increased cell proliferation, pS2 and Ki-67 expression was observed in the presence of DHT, which is not a substrate for aromatase. In contrast, DHT caused a significant suppression of cell proliferation (68% and 38%), pS2 and Ki-67 expression in the (ER(+)) MCF-7 and T47D cells. More importantly, DHT decreased cell proliferation in (ER(-)) MDA-MB-231 cells by 38%. The results suggest that androgens that cannot be aromatized, like DHT, may provide a perspective for treatment of breast cancer patients, especially those with triple negative breast cancer.


Assuntos
Androgênios/farmacologia , Técnicas de Cocultura/métodos , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Glândulas Mamárias Humanas/citologia , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Estradiol/metabolismo , Feminino , Fibroblastos/citologia , Humanos , Hidroxitestosteronas/metabolismo , Hidroxitestosteronas/farmacologia , Receptores de Estrogênio/metabolismo , Testosterona/metabolismo , Testosterona/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA