Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Bioact Mater ; 37: 393-406, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38689659

RESUMO

Radiation-induced heart disease (RIHD), characterized by severe oxidative stress and immune dysregulation, is a serious condition affecting cancer patients undergoing thoracic radiation. Unfortunately, clinical interventions for RIHD are lacking. Selenium (Se) is a trace element with excellent antioxidant and immune-modulatory properties. However, its application in heart radioprotection remains challenging. Herein, we developed a novel bioactive Cordyceps militaris-based Se oral delivery system (Se@CM), which demonstrated superior radioprotection effects in vitro against X-ray-induced damage in H9C2 cells through suppressing excessive ROS generation, compared to the radioprotectant Amifostine. Moreover, Se@CM exhibited exceptional cardioprotective effects in vivo against X-ray irradiation, reducing cardiac dysfunction and myocardial fibrosis by balancing the redox equilibrium and modulating the expression of Mn-SOD and MDA. Additionally, Se@CM maintained immuno-homeostasis, as evidenced by the upregulated population of T cells and M2 macrophages through modulation of selenoprotein expression after irradiation. Together, these results highlight the remarkable antioxidant and immunity modulation properties of Se@CM and shed light on its promising application for cardiac protection against IR-induced disease. This research provides valuable insights into developing effective strategies for preventing and managing RIHD.

2.
Biomaterials ; 305: 122452, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38154440

RESUMO

Radiotherapy is still the recommended treatment for cervical cancer. However, radioresistance and radiation-induced side effects remain one of the biggest clinical problems. Selenium (Se) has been confirmed to exhibit radiation-enhancing effects for cancer treatment. However, Se species dominate the biological activities and which form of Se possesses better radiosensitizing properties and radiation safety remains elusive. Here, different Se species (the valence state of Se ranged from - 2, 0, +4 to + 6) synergy screen was carried out to identify the potential radiosensitizing effects and radiation safety of Se against cervical cancer. We found that the therapeutic effects varied with the changes in the Se valence state. Sodium selenite (+4) displayed strong cancer-killing effects but also possessed severe cytotoxicity. Sodium selenate (+6) neither enhanced the killing effects of X-ray nor possessed anticancer activity by its alone treatment. Although nano-selenium (0), especially Let-SeNPs, has better radiosensitizing activity, the - 2 organic Se, such as selenadiazole derivative SeD (-2) exhibited more potent anticancer effects and possessed a higher safe index. Overall, the selected Se drugs were able to synergize with X-ray to inhibit cell growth, clone formation, and cell migration by triggering G2/M phase arrest and apoptosis, and SeD (-2) was found to exhibit more potent enhancing capacity. Further mechanism studies showed that SeD mediated p53 pathway activation by inducing DNA damage through promoting ROS production. Additionally, SeD combined with X-ray therapy can induce an anti-tumor immune response in vivo. More importantly, SeD combined with X-ray significantly inhibited the liver metastasis of tumor cells and alleviated the side effects caused by radiation therapy in tumor-bearing mice. Taken together, this study demonstrates the radiosensitization and radiation safety effects of different Se species, which may shed light on the application of such Se-containing drugs serving as side effects-reducing agents for cervical cancer radiation treatment.


Assuntos
Neoplasias Hepáticas , Radiossensibilizantes , Selênio , Neoplasias do Colo do Útero , Humanos , Feminino , Camundongos , Animais , Selênio/farmacologia , Selênio/uso terapêutico , Neoplasias do Colo do Útero/tratamento farmacológico , Neoplasias do Colo do Útero/radioterapia , Proteína Supressora de Tumor p53 , Radiossensibilizantes/farmacologia , Radiossensibilizantes/uso terapêutico , Neoplasias Hepáticas/tratamento farmacológico
3.
J Nanobiotechnology ; 21(1): 47, 2023 Feb 09.
Artigo em Inglês | MEDLINE | ID: mdl-36759859

RESUMO

In clinical practice, cisplatin is the most commonly used chemotherapy drug to treat a range of malignancies. Severe ROS-regulated nephrotoxicity, however, restricts its applicability. Currently, the main mechanisms leading to cisplatin-induced nephrotoxicity in clinical settings involve hydration or diuresis. However, not all patients can be treated with massive hydration or diuretics. Therefore, it is crucial to develop a treatment modality that can effectively reduce nephrotoxicity through a foodborne route. Selenium has been reported to have strong antioxidant as well as anticancer effects when administered as spore oil. Herein, we established cellular and animal models of cisplatin-induced nephrotoxicity and synthesized spore oil-functionalized nano-selenium (GLSO@SeNPs). We found that GLSO@SeNPs inhibit the mitochondrial apoptotic pathway by maintaining oxidative homeostasis and regulating related signaling pathways (the MAPK, caspase, and AKT signaling pathways). In vivo, GLSO@SeNPs could effectively improve cisplatin-induced renal impairment, effectively maintaining oxidative homeostasis in renal tissues and thus inhibiting the process of renal injury. In addition, GLSO@SeNPs were converted into selenocysteine (SeCys2), which may exert protective effects. Furthermore, GLSO@SeNPs could effectively modulate the ratio of immune cells in kidneys and spleen, reducing the proportions of CD3+CD4+ T cells, CD3+CD8+ T cells, and M1 phenotype macrophages and increasing the proportion of anti-inflammatory regulatory T cells. In summary, in this study, we synthesized food-derived spore oil-functionalized nanomaterials, and we explored the mechanisms by which GLSO@SeNPs inhibit cisplatin-induced nephrotoxicity. Our study provides a basis and rationale for the inhibition of cisplatin-induced nephrotoxicity by food-derived nutrients.


Assuntos
Cisplatino , Selênio , Animais , Cisplatino/farmacologia , Selênio/farmacologia , Linfócitos T CD8-Positivos , Rim , Estresse Oxidativo , Imunidade , Esporos , Apoptose
4.
Chem Asian J ; 17(10): e202200181, 2022 May 16.
Artigo em Inglês | MEDLINE | ID: mdl-35343080

RESUMO

Molecular phototheranostics as an emerging field of modern precision medicine has recently attracted increasing research attention owing to non-invasiveness, high precision, and controllable nature of light. In this work, we reported promising gadolinium (Gd3+ ) porphyrinoids as phototheranostic agents for magnetic resonance imaging (MRI) and photodynamic therapy (PDT). The synthesized Gd-1-4-Glu featured with meso-glycosylation and ß-lactonization to endow good biocompatibility and improved photophysical properties. In particular, ß-lactonization of glycosylated Gd3+ porphyrinoids substantially red-shifted Q band absorption to near-infrared (NIR) region and boosted generation of reactive oxygen species including 1 O2 , and some radical species that engaged in both type II and type I PDT pathways. In addition, the number and regioisomerism of ß-oxazolone moieties was observed to play an essential role in improving longitude relaxivity (r1 ) of Gd-1-4-Glu of up to 4.3±0.2 mM-1 s-1 by affecting environmental water exchange. Taking Gd-4-Glu as a promising complex, we further achieved real-time T1 -weighted MRI and PDT on HeLa tumour mice in vivo, revealing the appealing potential of Gd3+ porphyrinoids in phototheranostics.


Assuntos
Gadolínio , Fotoquimioterapia , Animais , Gadolínio/farmacologia , Células HeLa , Humanos , Imageamento por Ressonância Magnética/métodos , Camundongos , Medicina de Precisão
5.
ACS Nano ; 16(3): 4379-4396, 2022 03 22.
Artigo em Inglês | MEDLINE | ID: mdl-35175721

RESUMO

Bismuth telluride (Bi2Te3) is an available thermoelectric material with the lowest band gap among bismuth chalcogenides, revealing a broad application in photocatalysis. Unfortunately, its size and morphology related to a radio-catalysis property have rarely been explored. Herein, an ethylenediaminetetraacetic acid (EDTA)-assisted hydrothermal strategy was introduced to synthesize polytypic Bi2Te3 nanoplates (BT NPs) that exhibit size-dependent radio-sensitization and metabolism characteristics in vivo. By simply varying the molar ratio of EDTA/Bi3+ during the reaction, BT NPs with different sizes and morphologies were obtained. EDTA acting as chelating agent and "capping" agent contributed to the homogeneous growth of BT NPs by eliminating dangling bonds and reducing the surface energy of different facets. Further analyzing the size-dependent radio-sensitization mechanism, larger-sized BT NPs generated holes that preferentially catalyzed the conversion of OH- to ·OH when irradiated with X-rays, while the smaller-sized BT NPs exhibited faster decay kinetics producing higher 1O2 levels to enhance radiotherapy effects. A metabolomic analysis revealed that larger-sized BT NPs were oxidized into Bi(Ox) in the liver via a citrate cycle pathway, whereas smaller-sized BT NPs accumulated in the kidney and were excreted in urine in the form of ions by regulating the metabolism of glutamate. In a cervical cancer model, BT NPs combined with X-ray irradiation significantly antagonized tumor suppression through the promotion of apoptosis in tumor cells. Consequently, in addition to providing a prospect of BT NPs as an efficient radio-sensitizer to boost the tumor radiosensitivity, we put forth a strategy that can be universally applied in synthesizing metal chalcogenides for catalysis-promoted radiotherapy.


Assuntos
Bismuto , Neoplasias , Bismuto/química , Ácido Edético , Humanos , Neoplasias/tratamento farmacológico , Neoplasias/radioterapia , Tolerância a Radiação , Telúrio/química
6.
Chem Commun (Camb) ; 56(92): 14495-14498, 2020 Nov 19.
Artigo em Inglês | MEDLINE | ID: mdl-33150881

RESUMO

Here, a triphenylphosphine (TPP)-labile prodrug of seleno-combretastatin-4 (CSeD) was designed and synthesized. A detailed investigation revealed that CSeD, which was shown to be very safe in circulating blood, could react with TPP to release CA-4 and a selenodiazole derivative, with accompanying powerful anticancer and antiangiogenesis effects, as well as radiosensitization properties.


Assuntos
Antineoplásicos/química , Bibenzilas/química , Compostos Organofosforados/química , Compostos Organosselênicos/química , Pró-Fármacos/química , Antineoplásicos/farmacologia , Permeabilidade da Membrana Celular , Relação Dose-Resposta a Droga , Liberação Controlada de Fármacos , Ensaios de Seleção de Medicamentos Antitumorais , Células Endoteliais da Veia Umbilical Humana , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Pró-Fármacos/farmacologia , Fator A de Crescimento do Endotélio Vascular/metabolismo
7.
Front Chem ; 8: 838, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33062637

RESUMO

Numerous pathological conditions, including cancer, inflammatory diseases, and neurodegenerative diseases, are accompanied by overproduction of reactive oxygen species (ROS). This makes ROS vital flagging molecules in disease pathology. ROS-responsive drug delivery platforms have been developed. Nanotechnology has been broadly applied in the field of biomedicine leading to the progress of ROS-responsive nanoparticles. In this review, we focused on the production and physiological/pathophysiological impact of ROS. Particular emphasis is put on the mechanisms and effects of abnormal ROS levels on oxidative stress diseases, including cancer, inflammatory disease, and neurodegenerative diseases. Finally, we summarized the potential biomedical applications of ROS-responsive nanocarriers in these oxidative stress diseases. We provide insights that will help in the designing of new ROS-responsive nanocarriers for various applications.

8.
Theranostics ; 10(22): 10154-10170, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32929340

RESUMO

Background: Lung cancer has a high mortality rate and is resistant to multiple chemotherapeutics. Natural Borneol (NB) is a monoterpenoid compound that facilitates the bioavailability of drugs. In this study, we investigated the effects of NB on chemosensitivity in the A549 human lung adenocarcinoma cell line and to elucidate therapeutic molecular target of NB. Methods: The chemosensitivity effects of NB in A549 cells were examined by MTT assay. The mechanism of NB action was evaluated using flow cytometry and Western blotting assays. Surface plasmon resonance (SPR) and LC-MS combined analysis (MS-SPRi) was performed to elucidate the candidate molecular target of NB. The chemosensitizing capacity of NB in vivo was assessed in nude mice bearing A549 tumors. Results: NB pretreatment sensitized A549 cells to low doxorubicin (DOX) dosage, leading to a 15.7% to 41.5% increase in apoptosis. This increase was correlated with ERK and AKT inactivation and activation of phospho-p38 MAPK, phospho-JNK, and phosphor-p53. Furthermore, this synergism depends on reactive oxygen species (ROS) generation. MS-SPRi analysis revealed that transient receptor potential melastatin-8 (TRPM8) is the candidate target of NB in potentiating DOX killing potency. Genetically, TRPM8 knock-down significantly suppresses the chemosensitizing effects of NB and inhibits ROS generation through restraining calcium mobilization. Moreover, pretreatment with NB synergistically enhances the anticancer effects of DOX to delay tumor progression in vivo. Conclusions: These results suggest that TRPM8 may be a valid therapeutic target in the potential application of NB, and show that NB is a chemosensitizer for lung cancer treatment.


Assuntos
Cálcio/metabolismo , Canfanos/farmacologia , Doxorrubicina/farmacocinética , Canais de Cátion TRPM/metabolismo , Células A549 , Animais , Linhagem Celular Tumoral , Sinergismo Farmacológico , Humanos , Camundongos , Camundongos Nus , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/efeitos dos fármacos , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
9.
ACS Nano ; 14(9): 11067-11082, 2020 09 22.
Artigo em Inglês | MEDLINE | ID: mdl-32806028

RESUMO

Natural killer (NK) cell-based immunotherapy represents a promising strategy to overcome the bottlenecks of cancer treatment. However, the therapeutic efficacy is greatly limited by downregulation of recognition ligands on the tumor cell surface, and the immunosuppressive effects can be thwarted by the tumor microenvironment such as secretion of transforming growth factor-beta (TGF-ß), which could stunt the NK cell-mediated immune response. To overcome these limitations, herein we developed a nanoemulsion system (SSB NMs) to co-deliver TGF-ß inhibitor and selenocysteine (SeC) to achieve amplified anticancer efficacy. SSB NMs significantly enhanced the lytic potency of NK92 cells by 2.1-fold. Moreover, a subtoxic dose of SSB NMs effectively sensitized MDA-MB-231 triple-negative breast cancer (TNBC) cells to NK cells derived from seven clinical patients, resulting in an up to 13.8-fold increase in cancer lysis. Mechanistic studies reveal that the sensitizing effects relied on natural killer group 2, member D (NKG2D)/NKG2D ligands (NKG2DLs) signaling with the involvement of DNA damage response. SSB NMs also effectively restrained TGF-ß/TGF-ß RI/Smad2/3 signaling, which thus enhanced NKG2DL expression on tumor cells and stimulated NKG2D surface expression on NK92 cells, ultimately contributing to the enhanced immune response. Furthermore, SSB NMs sustained release of SeC and TGF-ß inhibitor and synergized with NK92 cells to induce significant anticancer effects in vivo. Together, this study not only demonstrates a simple strategy for the design of a nanoemulsion to co-deliver synergistic drugs but also sheds light on the application and action mechanisms in NK cell adaptive therapy against breast cancer, especially TNBCs.


Assuntos
Neoplasias , Fator de Crescimento Transformador beta , Linhagem Celular Tumoral , Cistina/análogos & derivados , Humanos , Imunoterapia , Células Matadoras Naturais , Neoplasias/tratamento farmacológico , Compostos Organosselênicos , Fatores de Crescimento Transformadores
10.
Chem Commun (Camb) ; 56(2): 179-196, 2019 Dec 19.
Artigo em Inglês | MEDLINE | ID: mdl-31782422

RESUMO

Organo-seleno compounds (org-Se) have been widely used in antitumor, antiviral, and antiinflammatory therapy; antioxidation and other biological fields. As such, they have made an important contribution to overcoming various kinds of diseases, and researchers are increasingly attracted to org-Se's synthesis and functional design. This review is mainly focused on the design and synthesis of various kinds of org-Se, followed by their anticancer mechanisms such as the mitochondria mediated pathway induced by ROS, death receptor mediated pathways involving p53 phosphorylation, and the activation of the AMPK pathway to promote apoptosis. Org-Se also serves as a sensitizer in chemotherapy and radiotherapy, and an antagonist against the cytotoxic effects induced by chemotherapeutic agents. Finally, we will summarize the development of cancer-targeted org-Se containing complexes, and nanotechnology-based org-Se for anticancer application. This review could provide information for the future design of chemically innovative org-Se with anticancer potential, and shed light on the discovery of nanomaterial-based pharmaceuticals to improve drug development and formation.


Assuntos
Antineoplásicos/uso terapêutico , Neoplasias/tratamento farmacológico , Compostos Organosselênicos/uso terapêutico , Animais , Antineoplásicos/química , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Complexos de Coordenação/química , Complexos de Coordenação/farmacologia , Complexos de Coordenação/uso terapêutico , Desenho de Fármacos , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Nanopartículas/química , Neoplasias/radioterapia , Compostos Organosselênicos/química , Compostos Organosselênicos/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Receptores de Morte Celular/metabolismo , Proteína Supressora de Tumor p53/metabolismo
11.
Biomaterials ; 219: 119377, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31374478

RESUMO

Natural killer (NK) cells-based therapy has been used widely for cancer treatment in clinic trails. However, the immunotherapeutic efficacy of this method has been greatly hindered by tumor evasion and diminished activities of NK cells. In the present study, a selenium (Se)-bearing ruthenium (Ru) complex (RuSe) was designed that could synergistically potentiate NK cell-mediated killing against prostate cancer cells. As expected, pretreatment of cancer cells with subtoxic doses of RuSe effectively augmented the lysis potency of NK cells, with up to 2.46-fold enhancement than NK cells alone, against PC3 cells. More importantly, low concentrations of RuSe could augment the tumor destroying potency of NK cells derived from 10 clinical patients, with the enhancement range from 0.78- to 11.9-fold against PC3 cells and 0.67- to 3.8-fold against LNCAP cells. Mechanistic studies revealed that the sensitizing effect of RuSe primarily depended on TRAIL/TRAIL-R and Fas/FasL-mediated signaling. Furthermore, the increased expression level of these ligands highly relied on ROS overproduction-triggered DNA damage and the downstream ATM and ATR pathways. Furthermore, RuSe potently activated and synergized with NK cells to restrain tumor growth in vivo without causing toxic side effects on major organs. Taken together, the current study not only provides a strategy for application of metal complexes in chemo-immunotherapy but also sheds light on the potential roles and mechanisms of action on such Se-containing drugs as efficient immune-sensitizing agents for NK cell-based immunotherapy.


Assuntos
Proteína Ligante Fas/metabolismo , Imunoterapia , Células Matadoras Naturais/imunologia , Neoplasias da Próstata/imunologia , Neoplasias da Próstata/terapia , Rutênio/farmacologia , Selênio/farmacologia , Transdução de Sinais , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Degranulação Celular/efeitos dos fármacos , Citotoxicidade Imunológica/efeitos dos fármacos , Dano ao DNA , Humanos , Células Matadoras Naturais/efeitos dos fármacos , Masculino , Neoplasias da Próstata/patologia , Espécies Reativas de Oxigênio/metabolismo , Receptores de Morte Celular/metabolismo , Transdução de Sinais/efeitos dos fármacos , Regulação para Cima/efeitos dos fármacos , Receptor fas/metabolismo
12.
Chem Commun (Camb) ; 55(67): 9904-9914, 2019 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-31360938

RESUMO

The successful clinical application of the three generation platinum anticancer drugs, cisplatin, carboplatin and oxaliplatin, has promoted research interest in metallodrugs; however, the problems of drug resistance and adverse effects have hindered their further application and effects. Thus, scientists are searching for new anticancer metallodrugs with lower toxicity and higher efficacy. The ruthenium complexes have emerged as the most promising alternatives to platinum-based anticancer agents because of their unique multifunctional biochemical properties. In this review, we first focus on the anticancer applications of various ruthenium complexes in different signaling pathways, including the mitochondria-mediated pathway, the DNA damage-mediated pathway, and the death receptor-mediated pathway. We then discuss the functionalization and cancer-targeting designs of different ruthenium complexes in conjunction with other therapies such as photodynamic therapy, photothermal therapy, radiosensitization, targeted therapy and nanotechnology for precise cancer therapy. This review will help in designing and accelerating the research progress regarding new anticancer ruthenium complexes.


Assuntos
Antineoplásicos/química , Complexos de Coordenação/química , Rutênio/química , Animais , Antineoplásicos/uso terapêutico , Complexos de Coordenação/uso terapêutico , Descoberta de Drogas , Humanos , Estrutura Molecular , Terapia de Alvo Molecular , Nanopartículas/química , Fotoquimioterapia/métodos , Medicina de Precisão , Relação Estrutura-Atividade
13.
Chem Asian J ; 13(18): 2730-2738, 2018 Sep 17.
Artigo em Inglês | MEDLINE | ID: mdl-29963768

RESUMO

A promising cancer-targeting agent for the induction of apoptosis in tumor necrosis factor (TNF) proteins, the TNF-related apoptosis-inducing ligand (TRAIL) ligand, has found limited applications in the treatment of cancer cells, owing to its resistance by cancer cell lines. Therefore, the rational design of anticancer agents that could sensitize cancer cells towards TRAIL is of great significance. Herein, we report that synthetic iron(II)-polypyridyl complexes are capable of inhibiting the proliferation of glioblastoma cancer cells and efficiently enhancing TRAIL-induced cell apoptosis. Mechanistic studies demonstrated that the synthesized complexes induced cancer-cell apoptosis through triggering the activation of p38 and p53 and inhibiting the activation of ERK. Moreover, uPA and MMP-2/MMP-9, among the most important metastatic regulatory proteins, were also found to be significantly alerted after the treatment. Furthermore, we also found that tumor growth in nude mice was significantly inhibited by iron complex Fe2 through the induction of apoptosis without clear systematic toxicity, as indicated by histological analysis. Taken together, this study provides evidence for the further development of metal-based anticancer agents and chemosensitizers of TRAIL for the treatment of human glioblastoma cancer cells.


Assuntos
Antineoplásicos/uso terapêutico , Apoptose/efeitos dos fármacos , Complexos de Coordenação/uso terapêutico , Compostos Ferrosos/uso terapêutico , Glioblastoma/tratamento farmacológico , Fenantrolinas/uso terapêutico , Animais , Antineoplásicos/síntese química , Antineoplásicos/farmacologia , Antineoplásicos/toxicidade , Transporte Biológico , Linhagem Celular Tumoral , Complexos de Coordenação/síntese química , Complexos de Coordenação/farmacologia , Complexos de Coordenação/toxicidade , Regulação para Baixo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Compostos Ferrosos/síntese química , Compostos Ferrosos/farmacologia , Compostos Ferrosos/toxicidade , Humanos , Ligantes , Masculino , Metaloproteinase 9 da Matriz/genética , Camundongos Nus , Fenantrolinas/síntese química , Fenantrolinas/farmacologia , Fenantrolinas/toxicidade , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/efeitos dos fármacos , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Ativador de Plasminogênio Tipo Uroquinase/genética , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
14.
Chem Asian J ; 13(11): 1447-1457, 2018 Jun 04.
Artigo em Inglês | MEDLINE | ID: mdl-29575811

RESUMO

Selenadiazole derivatives (SeDs) have been found to show promise in chemo-/radiotherapy applications by activating various downstream signaling pathways. However, the functional role of SeDs on angiogenesis, which is pivotal for tumor progression and metastasis, has not yet been elucidated. In the present study, we have examined the antiangiogenic activities of SeDs and elucidated their underlying mechanisms. The results showed that the as-synthesized SeDs not only enhanced their anticancer activities against several human cancer cells but also showed more potent inhibition on human umbilical vein endothelial cells (HUVECs). The in vitro results suggested that SeDs, especially 1 a, dose-dependently inhibited the vascular endothelial growth factor (VEGF)-induced cell migration, invasion, and capillary-like structure formation of HUVECs. Compound 1 a also significantly suppressed VEGF-induced angiogenesis in a Matrigel plug assay as part of a C57/BL6 mice assay by means of down regulation of VEGF. Furthermore, we found that 1 a significantly inhibited MCF-7 human breast tumor growth in nude mice without severe systematic cytotoxicity. Compound 1 a was more effective in inhibiting cell proliferation and induced a much more pronounced apoptosis effect in endothelial cells than MCF-7 cells, which implies that endothelial cells might be the primary target of 1 a. Further mechanistic studies on tumor growth inhibition effects and neovessel formation suppression demonstrated that 1 a inhibited cell viability of MCF-7 and HUVECs by induction of cell apoptosis, accompanied by poly(adenosine diphosphate ribose)polymerase (PARP) cleavage and caspase activation. Additionally, the 1 a-induced antiangiogenesis effect was achieved by abolishing the VEGF-VEGFR2-ERK/AKT (ERK=extracellular signal-regulated kinases; AKT=protein kinease B) signal axis and enhanced the apoptosis effect by triggering reactive oxygen species (ROS)-mediated DNA damage. Taken together, these results clearly demonstrate the antiangiogenic potency of SeDs and the underlying molecular mechanisms.


Assuntos
Inibidores da Angiogênese/uso terapêutico , Antineoplásicos/uso terapêutico , Benzimidazóis/uso terapêutico , Compostos Organosselênicos/uso terapêutico , Inibidores da Angiogênese/síntese química , Inibidores da Angiogênese/toxicidade , Animais , Antineoplásicos/síntese química , Antineoplásicos/toxicidade , Apoptose/efeitos dos fármacos , Benzimidazóis/síntese química , Benzimidazóis/toxicidade , Neoplasias da Mama/tratamento farmacológico , Movimento Celular/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Células Endoteliais da Veia Umbilical Humana , Humanos , Células MCF-7 , Camundongos Endogâmicos C57BL , Invasividade Neoplásica/prevenção & controle , Compostos Organosselênicos/síntese química , Compostos Organosselênicos/toxicidade , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
15.
Chem Asian J ; 12(9): 982-987, 2017 May 04.
Artigo em Inglês | MEDLINE | ID: mdl-28233940

RESUMO

Antiangiogenesis therapy is a proven strategy for the treatment of cancers. Herein, we demonstrate that iron(II) complexes containing 1,10-phenanthroline(phen) derivatives were capable of suppressing angiogenesis in vitro in a dose-dependent manner. Interestingly, the introduction of selenium into an iron(II) complex ((Fe(phenSe)3 (ClO4 )2 (phenSe=2-selenoimidazole[4,5-f]1,10-phenanthroline)) could enhance its antiangiogenic efficacy. Mechanistic studies demonstrated that the complex potently induced endothelial cell apoptosis as evidenced by activation of caspases and PARP cleavage. The iron(II) complex activated p53-mediated mitochondrial dysfunction as can be seen by the upregulation in the expression of p53 and proapoptotic Bcl-2 family proteins, and downregulation in the expression of Bcl-2 family proteins. Additionally, the complex inhibited VEGF expression and gave rise to dephosphorylated AKT, which suppressed the transmission of the mitogenic signaling pathway. Taken together, this study could provide a strategy for the rational design of high-efficacy antivascular agents.


Assuntos
Inibidores da Angiogênese/farmacologia , Compostos Ferrosos/farmacologia , Neovascularização Patológica/tratamento farmacológico , Selênio/farmacologia , Inibidores da Angiogênese/síntese química , Inibidores da Angiogênese/química , Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Compostos Ferrosos/química , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Humanos , Estrutura Molecular , Neovascularização Patológica/patologia , Selênio/química , Relação Estrutura-Atividade
16.
Nanomedicine ; 12(6): 1627-39, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-26961468

RESUMO

Angiogenesis is essential for tumorigenesis, progression and metastasis. Herein we described the synthesis of RGD peptide-decorated and doxorubicin-loaded selenium nanoparticles (RGD-NPs) targeting tumor vasculature to enhance the cellular uptake and antiangiogenic activities in vitro and in vivo. After internalization by receptor-mediated endocytosis, this nanosystem disassembled under acidic condition with the presence of lysozymes and cell lysate, leading to bioresponsive triggered drug release. Mechanistic investigation revealed that RGD-NPs inhibited angiogenesis through induction of apoptosis and cell cycle arrest in human umbilical vein endothelial cells (HUVECs) via suppression of VEGF-VEGFR2-ERK/AKT signaling axis by triggering ROS-mediated DNA damage. Additionally, RGD-NPs can inhibit MCF-7 tumor growth and angiogenesis in nude mice via down-regulation of VEGF-VEGFR2, effectively reduce the toxicity and prolong the blood circulation in vivo. Our results suggest that the strategy to use RGD-peptide functionalized SeNPs as carriers of anticancer drugs is an efficient way to achieve cancer-targeted antiangiogenesis synergism.


Assuntos
Nanopartículas , Selênio , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Inibidores da Angiogênese , Animais , Apoptose , Humanos , Camundongos , Camundongos Nus , Neoplasias/terapia , Proteínas Proto-Oncogênicas c-akt , Fator A de Crescimento do Endotélio Vascular
17.
Biomaterials ; 51: 30-42, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25770995

RESUMO

Radioresistance and limitation of irradiative dosage usually lead to failure in depletion of hypoxic tumors. Herein we developed multifunctional mesoporous silica nanoparticles (MSNs) as a carrier of a novel anticancer selenoamino acid (selenocystine, SeC), to achieve synergistic chemo-/radiotherapy. This multifunctional nanosystem effectively sensitizes cancer cells to X-ray radiotherapy. Conjugation of TAT cell penetrating peptide and transferrin to the surface of MSNs significantly enhances its internalization in cancer cells through receptor-mediated endocytosis. SeC@MSNs-Tf/TAT significantly enhanced X-ray-induced growth inhibition in cervical cancer cells by induction of apoptosis, mainly through death receptor-mediated extrinsic apoptotic pathway. Upon radiation, SeC@MSNs-Tf/TAT promoted intracellular ROS overproduction, which induced apoptotic cell death by affecting p53, AKT and MAPKs pathways. Furthermore, SeC@MSNs-Tf/TAT also significantly inhibited HeLa tumor growth in nude mice model through suppression of cell proliferation and induction of apoptosis. In vivo toxicity of the SeC@MSNs-Tf/TAT nanoparticles was investigated using the mouse model. The results of histological analysis revealed that, the nanoparticles did not show any obvious damage to these major organs under the experimental conditions, including heart, liver, spleen, lung and kidney. Taken together, this study demonstrates an effective and safe strategy for cancer-targeted chemo-/radiotherapy of human cancers.


Assuntos
Sistemas de Liberação de Medicamentos , Nanopartículas/química , Neoplasias/tratamento farmacológico , Neoplasias/radioterapia , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais , Animais , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Cistina/análogos & derivados , Cistina/química , Dano ao DNA , Humanos , Espaço Intracelular/metabolismo , Masculino , Camundongos Endogâmicos ICR , Camundongos Nus , Nanopartículas/ultraestrutura , Neoplasias/diagnóstico por imagem , Nitrogênio/química , Compostos Organosselênicos/química , Espectroscopia Fotoeletrônica , Porosidade , Radiografia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/efeitos da radiação , Dióxido de Silício/química , Transferrina/metabolismo , Raios X , Ensaios Antitumorais Modelo de Xenoenxerto , Produtos do Gene tat do Vírus da Imunodeficiência Humana/farmacologia
18.
Metallomics ; 7(3): 439-47, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25630534

RESUMO

Antiangiogenic therapy is considered to be a promising strategy for the treatment of cancers. VEGF and its receptors are important angiogenic factors involved in tumor growth. In the present study, the new ruthenium(ii) complexes containing 2,6-bis(benzimidazolyl)pyridine have been identified as potent antiangiogenic agents in vitro and in vivo, through activation of distinct antiangiogenic signaling pathways. Specifically, [Ru(bbp)(p-mpip)Cl]ClO4 (complex , bbp = 2,6-bis(benzimidazolyl)pyridine; p-mpip = 2-(4-methylphenyl)imidazo[4,5-f]-1,10-phenanthroline) exhibited the highest antiangiogenic activity, as evidenced by significant suppression of neovessel formation in chick chorioallantoic membranes and blockage of the angiogenesis in a matrigel plugs assay, which are significantly higher than those of the most accepted anti-metastasis ruthenium-based drug NAMI-A. Generally, these kinds of complexes induced the G0/G1cell cycle by inhibiting the formation of a Cyclin D1/CDK4 complex and CDK2 activation, through up regulation of the expression levels of p15(INK4B), p21(Cip1) and p27(Kip1). Moreover, the complexes also triggered intracellular DNA damage, and thus activated the phosphorylation of ATM, ATR, CHK1, Histone and p53. The suppression of Akt and ERK1/2 pathways reinforced the cell cycle perturbation effects of the complexes. Interestingly, complex displayed strong inhibition on the activation of VEGF and VEGFR-2 phosphorylation, which blocked the transmission of the mitogenic signal through Akt and ERK1/2 pathways, and thus enhanced cell cycle arrest. In contrast, we found that the most accepted anti-metastasis ruthenium based drug NAMI-A exerted lower antiangiogenic activity via activation of the DNA damage-mediated pathway, but showed no effects on VEGF and VEGFR-2 phosphorylation. Taken together, this study clearly demonstrates the distinct antiangiogenic mechanisms of metal complexes, and these kinds of complexes can be further developed as anti-vascularized drugs and as alternative agents of NAMI-A for the treatment of cancers.


Assuntos
Inibidores da Angiogênese/farmacologia , Benzimidazóis/farmacologia , Complexos de Coordenação/química , Complexos de Coordenação/farmacologia , Rutênio/farmacologia , Transdução de Sinais/efeitos dos fármacos , Animais , Benzimidazóis/síntese química , Benzimidazóis/química , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Galinhas , Complexos de Coordenação/síntese química , Dano ao DNA , Células Endoteliais da Veia Umbilical Humana/citologia , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Masculino , Camundongos Endogâmicos C57BL , Modelos Biológicos , Neovascularização Fisiológica/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular/farmacologia , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
19.
J Mater Chem B ; 3(42): 8383-8393, 2015 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-32262891

RESUMO

The lack of early and timely diagnosis of tumors and the monitoring of their response to therapeutics have limited the successful cancer treatments. Theranostic agents are expected to realize the dual-purpose of simultaneous diagnosis and therapy for treatments of cancers. In the present study, we have examined the effects of the chemical structure of selenadiazole derivatives (SeDs) on their anticancer efficacy and radio-sensitization against clinically used X-rays. The results showed that the introduction of a nitro group (-NO2) into SeD-3 significantly enhanced the anticancer activity of SeDs. The higher lipophilicity endowed SeD-3 with higher cellular internalization ability, resulting in higher cellular uptake and anticancer efficacy. Specifically, the capacity of autofluorescence allowed the use of SeD-3 as a promising theranostic agent to directly monitor the cellular uptake, localization and biodistribution in vitro and in vivo. Interestingly, SeD-3 also significantly enhanced the sensitivity of HeLa cervical cells to X-ray-induced apoptosis by targeting the inhibition of TrxR and promoting intracellular ROS overproduction, which activated the downstream ROS-mediated signaling pathways to regulate cell apoptosis. Furthermore, SeD-3 exhibited satisfactory in vivo antitumor efficacy through the inhibition of tumor proliferation and induction of tumor cell apoptosis, and showed no toxicity to the main organs. Moreover, from the results of hematological analysis, we found that not only inhibiting the tumor growth, treatment of SeD-3 also alleviated the damage of liver, kidney and heart function of nude mice induced by HeLa xenografts. Taken together, this study demonstrates that SeDs could be further developed as an effective and safe theranostic agent for simultaneous cancer chemo-/radiotherapy.

20.
Metallomics ; 6(8): 1480-90, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24823440

RESUMO

TrxR is an NADPH-dependent selenoenzyme upregulated in a number of cancers. It plays a pivotal role in cancer progression and represents an increasingly attractive target for anticancer drugs. The limitations of cisplatin in cancer treatment have motivated the extensive investigation to other metal complexes, especially ruthenium (Ru) complexes. In this study, we present the in vitro biological evaluation of four Ru(II) polypridyl complexes with diimine ligands, namely, [Ru(bpy)3](2+) (1), [Ru(phen)3](2+) (2), [Ru(ip)3](2+) (3), [Ru(pip)3](2+) (4) (bpy = 2,2'-bipyridine, phen = 1,10-phenanthroline, ip = imidazole[4,5-f][1,10]phenanthroline, pip = 2-phenylimidazo[4,5-f][1,10]phenanthroline), and demonstrate that they exhibit antiproliferative activities against A375 human melanoma cells through inhibition of TrxR. As the planarity of the structure increases, their TrxR-inhibitory effects and in vitro anticancer activities were enhanced. Among them, complex 4 exhibited higher antiproliferative activity than cisplatin, and the TrxR-inhibitory potency of 4 was more effective than auranofin, a positive TrxR inhibitor. Complex 4 suppressed the cancer cell growth through induction of apoptosis as evidenced by accumulation of sub-G1 cell population, DNA fragmentation and nuclear condensation. Moreover, complex 4 was able to localize in mitochondria and therein induced ROS-dependent apoptosis by inhibition of TrxR activity. Activation of MAPKs, AKT, DNA damage-mediated p53 phosphorylation and inhibition of VEGFR signaling were also triggered in cells exposed to complex 4. On the basis of this evidence, we suggest that Ru polypyridyl complexes could be developed as TrxR-targeted agents that demonstrate application potentials for treatment of cancers.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Rutênio/farmacologia , Tiorredoxina Dissulfeto Redutase/metabolismo , Humanos , Espécies Reativas de Oxigênio/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA