Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
ACS Nano ; 18(13): 9413-9430, 2024 Apr 02.
Artigo em Inglês | MEDLINE | ID: mdl-38522084

RESUMO

Personalized in situ tumor vaccination is a promising immunotherapeutic modality. Currently, seeking immunogenic cell death (ICD) to generate in situ tumor vaccines is still mired by insufficient immunogenicity and an entrenched immunosuppressive tumor microenvironment (TME). Herein, a series of tetrazine-functionalized ruthenium(II) sonosensitizers have been designed and screened for establishing a bioorthogonal-activated in situ tumor vaccine via oncolytic pyroptosis induction. Based on nanodelivery-augmented bioorthogonal metabolic glycoengineering, the original tumor is selectively remolded to introduce artificial target bicycle [6.1.0] nonyne (BCN) into cell membrane. Through specific bioorthogonal ligation with intratumoral BCN receptors, sonosensitizers can realize precise membrane-anchoring and synchronous click-activation in desired tumor sites. Upon ultrasound (US) irradiation, the activated sonosensitizers can intensively disrupt the cell membrane with dual type I/II reactive oxygen species (ROS) generation for a high-efficiency sonodynamic therapy (SDT). More importantly, the severe membrane damage can eminently evoke oncolytic pyroptosis to maximize tumor immunogenicity and reverse immunosuppressive TME, ultimately eliciting powerful and durable systemic antitumor immunity. The US-triggered pyroptosis is certified to effectively inhibit the growths of primary and distant tumors, and suppress tumor metastasis and recurrence in "cold" tumor models. This bioorthogonal-driven tumor-specific pyroptosis induction strategy has great potential for the development of robust in situ tumor vaccines.


Assuntos
Vacinas Anticâncer , Neoplasias , Humanos , Piroptose , Neoplasias/terapia , Vacinação , Ultrassonografia , Imunossupressores , Microambiente Tumoral , Linhagem Celular Tumoral
2.
J Inorg Biochem ; 246: 112299, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37354603

RESUMO

Metal copper complexes have attracted extensive attention as potential alternatives to platinum-based anticancer drugs due to their possible different modes of action. Herein, a new copper(II) gluconate complex, namely [Cu(DPQ)(Gluc)]·2H2O (CuGluc, DPQ = pyrazino[2,3-f][1,10]phenanthroline), with good water-solubility and high anticancer activity was synthesized by using D-gluconic acid (Gluc-2H) as an auxiliary ligand. The complex was well characterized by single-crystal X-ray diffraction analysis, elemental analysis, molar conductivity, and Fourier transform infrared spectroscopy (FTIR). The DNA-binding experiments revealed that CuGluc was bound to DNA by intercalation with end-stacking binding. CuGluc could oxidatively cleave DNA, in which 1O2 and H2O2 were involved. In addition, CuGluc was bound to the IIA subdomain of human serum albumin (HSA) through hydrophobic interaction and hydrogen bonding, showing a good affinity for HSA. The complex showed superior anticancer activity toward several cancer cells than cisplatin in vitro. Further studies indicated that CuGluc caused apoptotic cell death in human liver cancer (HepG2) cells through elevated intracellular reactive oxygen species (ROS) levels, mitochondrial dysfunction, cell cycle arrest, and caspase activation. Interestingly, CuGluc also triggered the ferroptosis mechanism through lipid peroxide accumulation and inhibition of glutathione peroxidase 4 (GPX4) activity. More importantly, CuGluc significantly inhibited tumor growth in vivo, which may benefit from the combined effects of apoptosis and ferroptosis. This work provides a promising strategy to develop highly effective antitumor copper complexes by coordinating with the glucose metabolite D-gluconic acid and exploiting the synergistic effects of apoptosis and ferroptosis mechanisms.


Assuntos
Antineoplásicos , Complexos de Coordenação , Ferroptose , Neoplasias , Humanos , Cobre/química , Peróxido de Hidrogênio/farmacologia , Complexos de Coordenação/farmacologia , Complexos de Coordenação/química , Apoptose , Gluconatos/farmacologia , Antineoplásicos/farmacologia , Antineoplásicos/química , Albumina Sérica Humana , DNA/química , Linhagem Celular Tumoral
3.
Dalton Trans ; 52(23): 8040-8050, 2023 Jun 13.
Artigo em Inglês | MEDLINE | ID: mdl-37227152

RESUMO

Copper complexes have long been considered as a promising class of anticancer or antibacterial therapeutics. In this paper, two novel copper(II) complexes containing a ß-carboline derivative and amino acids, namely [Cu(1-Im-ßc)(L-Val)]ClO4·0.5H2O (Cu1) and [Cu(1-Im-ßc)(L-Phe)]ClO4·0.5H2O (Cu2), where 1-Im-ßc = 1-(2-imidazolyl)-ß-carboline, L-Val = L-valine, and L-Phe = L-phenylalanine, were designed and synthesized. The complexes were characterized by elemental analysis, infrared spectroscopy, molar conductivity measurements, and mass spectrometry to determine their spatial structures and compositions. Both complexes bind to DNA by insertion. The complexes also show a good affinity for human serum albumin (HSA). In addition, the antitumor activity of the two complexes against lung cancer cells (A549), cervical cancer cells (HeLa), and breast cancer cells (MBA-MD-231) is significantly superior to that of the traditional antitumor drug, cisplatin. Finally, the anticancer mechanism results show that the complexes can induce apoptosis in HeLa cells, which is associated with mitochondrial damage, oxidative stress caused by reactive oxygen species (ROS) production, and activation of the caspase protein family. This study demonstrates that the introduction of aromatic heterocyclic alkaloid ligands with a broad spectrum of biological activities and water-soluble amino acid ligands into copper complexes can regulate their amphiphilic properties and biological activity, so as to obtain highly efficient copper-based therapeutics.


Assuntos
Cobre , Humanos , Aminoácidos/química , Linhagem Celular Tumoral , Cobre/química , DNA/química , Lipídeos/química , Modelos Moleculares , Albumina Sérica Humana/química , Estrutura Terciária de Proteína , Apoptose/efeitos dos fármacos , Antineoplásicos/farmacologia
4.
Dalton Trans ; 52(5): 1291-1300, 2023 Jan 31.
Artigo em Inglês | MEDLINE | ID: mdl-36625001

RESUMO

Mitochondria-targeted photodynamic therapy (PDT) has recently been recognized as a promising strategy for effective cancer treatment. In this work, a mitochondria-targeted near-infrared (NIR) aggregation-induced emission (AIE)-active phosphorescent Ir(III) complex (Ir1) is reported with highly favourable mitochondria-targeted bioimaging and cancer PDT properties. Complex Ir1 has strong absorption in the visible light region (∼500 nm) and can effectively produce singlet oxygen (1O2) under green light (525 nm) irradiation. It preferentially accumulates in the mitochondria of human breast cancer MDA-MB-231 cells as revealed by colocalization analysis. Complex Ir1 displays high phototoxicity toward human breast cancer MDA-MB-231 cells and mouse breast cancer 4T1 cells. Complex Ir1 induces reactive oxygen species (ROS) production, mitochondrial dysfunction, and endoplasmic reticulum (ER) stress in MDA-MB-231 cells upon photoirradiation, leading to apoptotic cell death. The favorable PDT performance of Ir1in vivo has been further demonstrated in tumour-bearing mice. Together, the results suggest that Ir1 is a promising photosensitizer for mitochondria-targeted imaging and cancer phototherapy.


Assuntos
Neoplasias da Mama , Fotoquimioterapia , Camundongos , Humanos , Animais , Feminino , Irídio/farmacologia , Fármacos Fotossensibilizantes/farmacologia , Fármacos Fotossensibilizantes/uso terapêutico , Neoplasias da Mama/tratamento farmacológico , Mitocôndrias , Linhagem Celular Tumoral
5.
Chem Sci ; 13(38): 11360-11367, 2022 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-36320579

RESUMO

Regulation of tumor hypoxia and redox homeostasis is a promising strategy for cancer therapy. Nanocatalytic medicine has played more and more important roles in this field because it can cleverly convert the efficiency and selectivity of catalysis into high therapeutic efficiency. Herein, we developed a platinum(iv)-ruthenium hybrid prodrug, named as Pt-Ru, for efficient chemo-catalytic synergistic therapy of hypoxic tumors. The ruthenium hybridization endowed the Pt(iv) prodrug with multi-enzyme catalytic activity, that is, mimicking catalase (CAT) to generate O2 in situ, mimicking peroxidase (POD) to produce reactive oxygen species, and mimicking glutathione peroxidase (GPx) to deplete GSH, thus effectively overcoming tumor hypoxia and cisplatin resistance. As a result, Pt-Ru treatment led to a superior anticancer efficacy to cisplatin both in vitro and in vivo. This work suggested redox homeostasis regulation as a tantalizing angle for developing the next generation of platinum drugs.

6.
J Med Chem ; 65(21): 14692-14700, 2022 11 10.
Artigo em Inglês | MEDLINE | ID: mdl-36353870

RESUMO

DNA damage repair is considered to be an important mechanism of cisplatin resistance, and the roles of iron homeostasis in action mechanisms of cisplatin have not been studied yet. Herein, a Pt(IV) prodrug (DFX-Pt) integrating cisplatin and the clinical oral iron-chelating agent deferasirox (DFX) is found to be highly active toward cisplatin-insensitive triple-negative breast cancer cells both in vitro and in vivo. RNA-sequencing shows that DFX-Pt can downregulate genes related to the double-strand break (DSB) damage pathway significantly. DFX-Pt can reduce cellular free iron, regulate the expression of the RNA demethylase, and elevate the levels of RNA N6-methyladenosine (m6A), which degrades the DSB-related genes in an m6A-dependent manner. In all, we first reveal the roles of RNA modification in mechanisms of combating DNA damage repair and show that the combination of iron homeostasis intervention may bring new treatment regimens for cisplatin resistance.


Assuntos
Pró-Fármacos , Humanos , Pró-Fármacos/farmacologia , Deferasirox/uso terapêutico , Cisplatino/farmacologia , Metilação , RNA , Quelantes de Ferro/uso terapêutico , Ferro , Dano ao DNA
7.
Natl Sci Rev ; 9(7): nwab194, 2022 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-35958681

RESUMO

Endoplasmic reticulum (ER) degradation by autophagy (ER-phagy) is a recently revealed selective autophagy pathway that plays important roles in organelle turnover and protein degradation, but the biological functions of ER-phagy are largely unknown. Here, we present an ER-targeting Re(I) tricarbonyl complex (Re-ERLAD) that can accumulate in the ER, induce ER-to-lysosome-associated degradation (ERLAD) upon visible light irradiation, and label ER buds and track their morphological alterations during ER-phagy. The emission of Re-ERLAD is sensitive to viscosity, which is a key parameter reflecting the amount of unfolded protein in the ER. Quantitative detection using two-photon fluorescence lifetime imaging microscopy shows that ER viscosity initially increases and then decreases during ERLAD, which reveals that ERLAD is a pathway for alleviating ER stress caused by unfolded proteins. In conclusion, our work presents the first specific photoinducer and tracker of ERLAD, which can be used in studying the regulatory mechanism and function of this process.

8.
Chem Commun (Camb) ; 58(58): 8069-8072, 2022 Jul 19.
Artigo em Inglês | MEDLINE | ID: mdl-35762870

RESUMO

Carbonic anhydrase IX (CAIX) is overexpressed in many cancer types. Herein, a CAIX targeting and binding-enhanced ruthenium-based photodegrader, Ru-dppz-CAi, is constructed by conjugating the photosensitizer with the inhibiting group via a rotatable moiety. The binding of Ru-dppz-CAi and CAIX leads to significant enhancement in the emission and photosensitizing properties. Ru-dppz-CAi can photodegrade CAIX both in vitro and in living cells, which significantly inhibits its catalytic activity. The protein photodegradation method may provide new strategies for the development of tools for protein functional studies.


Assuntos
Anidrases Carbônicas , Rutênio , Antígenos de Neoplasias/metabolismo , Anidrase Carbônica IX , Anidrases Carbônicas/metabolismo , Fotólise , Fármacos Fotossensibilizantes/farmacologia
9.
ACS Appl Mater Interfaces ; 12(39): 43444-43455, 2020 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-32883070

RESUMO

Cisplatin resistance in tumor cells is known mainly due to the reduced accumulation of platinum ions by efflux, detoxification by intracellular GSH, and nucleotide excision repair machinery-mediated nuclear DNA repair. In this work, theranostic Pt(IV)-NPs, which are precisely self-assembled by biotin-labeled Pt(IV) prodrug derivative and cyclodextrin-functionalized IR780 in a 1:1 molecular ratio, have been developed for addressing all these hurdles via mitochondria-targeted chemotherapy solely or chemophotothermal therapy. In these nanoparticles, IR780 as a small-molecule dye acts as a mitochondria-targeting ligand to make Pt(IV)-NPs relocate finally in the mitochondria and release cisplatin. As demonstrated by in vitro and in vivo experiments, Pt(IV)-NPs can markedly facilitate cancer-specific mitochondrial targeting, inducing mitochondrial dysfunction and mitochondrial DNA (mtDNA) damage, thus greatly increasing the Pt accumulation, reducing the GSH levels, and avoiding DNA repair machinery in cisplatin-resistant cancer cells (A549R), finally resulting in significant inhibition of A549R tumor growth on animal models by chemotherapy solely. Upon near-infrared irradiation, mitochondria-targeted chemophotothermal synergistic therapy can be realized, further overcoming cisplatin resistance and even eliminating A549R tumors completely. Moreover, such novel Pt(IV)-NPs integrate multimodal targeting (cancer and mitochondria targeting), imaging (near-infrared imaging and photoacoustic imaging), and therapeutic (chemo- and photothermal therapy) moieties in a constant ratio (1:1:1) into a single, reproducible, and structurally homogeneous entity, avoiding nonuniform drug loading and premature leakage as well as the discrete steps of imaging and therapy, which thus is more beneficial for precise therapeutics and future clinical translation.


Assuntos
Antineoplásicos/farmacologia , Cisplatino/farmacologia , Mitocôndrias/efeitos dos fármacos , Nanopartículas/química , Terapia Fototérmica , Pró-Fármacos/farmacologia , Células A549 , Animais , Antineoplásicos/síntese química , Antineoplásicos/química , Apoptose/efeitos dos fármacos , Cisplatino/química , DNA Mitocondrial/efeitos dos fármacos , DNA Mitocondrial/metabolismo , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Mitocôndrias/metabolismo , Estrutura Molecular , Neoplasias Experimentais/diagnóstico por imagem , Neoplasias Experimentais/tratamento farmacológico , Neoplasias Experimentais/metabolismo , Tamanho da Partícula , Pró-Fármacos/síntese química , Pró-Fármacos/química , Propriedades de Superfície
10.
Dalton Trans ; 49(33): 11583-11590, 2020 Aug 25.
Artigo em Inglês | MEDLINE | ID: mdl-32766642

RESUMO

Chemotherapeutic agents that affect lysosomal functions represent a promising strategy for selective tumor therapy and overcoming drug resistance. In this work, two dinuclear phosphorescent rhenium(i) tricarbonyl complexes (DRe1 and DRe2) containing carboline derivatives have been synthesized, characterized and explored as potential chemotherapeutic and photodynamic therapy agents. The two dinuclear rhenium(i) complexes have good intrinsic phosphorescence properties and can label the lysosomes in cancer cells. Both dinuclear rhenium(i) complexes show potent anticancer activities toward several tested cancer cells. Moreover, they also have marked inhibitory activities against cisplatin-resistant human lung carcinoma cells (A549R), with complex DRe2 displaying 16-fold higher activity than cisplatin. Mechanism studies reveal that complex DRe2 can induce cancer cells to overproduce reactive oxygen species (ROS), including superoxide anion radicals, which leads to lysosomal membrane permeabilization (LMP) and subsequent cell apoptosis. Additionally, both DRe1 and DRe2 display significant phototoxicity under light (425 nm) irradiation in A549 cells, with phototoxicity index values of 60.8 and 41.8, respectively. Therefore, these two dinuclear organometallic rhenium(i) tricarbonyl complexes are potential anticancer agents for combined chemo-photodynamic therapy.


Assuntos
Antineoplásicos/síntese química , Complexos de Coordenação/síntese química , Substâncias Luminescentes/síntese química , Fármacos Fotossensibilizantes/síntese química , Rênio/química , Células A549 , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Carbolinas/química , Permeabilidade da Membrana Celular , Proliferação de Células/efeitos dos fármacos , Cisplatino/farmacologia , Complexos de Coordenação/farmacologia , Descoberta de Drogas , Células HeLa , Humanos , Ligantes , Substâncias Luminescentes/farmacologia , Lisossomos/metabolismo , Fotoquimioterapia , Fármacos Fotossensibilizantes/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Relação Estrutura-Atividade
11.
Angew Chem Int Ed Engl ; 59(42): 18755-18762, 2020 10 12.
Artigo em Inglês | MEDLINE | ID: mdl-32634290

RESUMO

The development and malignancy of cancer cells are closely related to the changes of the epigenome. In this work, a mitochondria-targeted rhenium(I) complex (DFX-Re3), integrating the clinical iron chelating agent deferasirox (DFX), has been designed. By relocating iron to the mitochondria and changing the key metabolic species related to epigenetic modifications, DFX-Re3 can elevate the methylation levels of histone, DNA, and RNA. As a consequence, DFX-Re3 affects the events related to apoptosis, RNA polymerases, and T-cell receptor signaling pathways. Finally, it is shown that DFX-Re3 induces immunogenic apoptotic cell death and exhibits potent antitumor activity in vivo. This study provides a new approach for the design of novel epigenetic drugs that can recode the cancer epigenome by intervening in mitochondrial metabolism and iron homeostasis.


Assuntos
Complexos de Coordenação/química , Ferro/metabolismo , Mitocôndrias/metabolismo , Rênio/química , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Complexos de Coordenação/farmacologia , Complexos de Coordenação/uso terapêutico , Deferasirox/química , Avaliação Pré-Clínica de Medicamentos , Epigenômica , Histonas/metabolismo , Humanos , Quelantes de Ferro/química , Metilação/efeitos dos fármacos , Camundongos , Mitocôndrias/efeitos dos fármacos , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Neoplasias/patologia , RNA Polimerase II/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Receptores de Antígenos de Linfócitos T/metabolismo , Transdução de Sinais/efeitos dos fármacos
12.
Angew Chem Int Ed Engl ; 59(42): 18556-18562, 2020 10 12.
Artigo em Inglês | MEDLINE | ID: mdl-32557982

RESUMO

Hypoxia and the acidic microenvironment play a vital role in tumor metastasis and angiogenesis, generally compromising the chemotherapeutic efficacy. This provides a tantalizing angle for the design of platinum(IV) prodrugs for the effective and selective killing of solid tumors. Herein, two carbonic anhydrase IX (CAIX)-targeting platinum(IV) prodrugs have been developed, named as CAIXplatins. Based on their strong affinity for and inhibition of CAIX, CAIXplatins can not only overcome hypoxia and the acidic microenvironment, but also inhibit metabolic pathways of hypoxic cancer cells, resulting in a significantly enhanced therapeutic effect on hypoxic MDA-MB-231 tumors both in vitro and in vivo compared with cisplatin/oxaliplatin, accompanied with excellent anti-metastasis and anti-angiogenesis activities. Furthermore, the cancer selectivity indexes of CAIXplatins are 70-90 times higher than those of cisplatin/oxaliplatin with effectively alleviated side-effects.


Assuntos
Anidrase Carbônica IX/antagonistas & inibidores , Hipóxia Celular , Complexos de Coordenação/química , Platina/química , Pró-Fármacos/química , Animais , Anidrase Carbônica IX/metabolismo , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Cisplatino/farmacologia , Análise por Conglomerados , Complexos de Coordenação/metabolismo , Complexos de Coordenação/farmacologia , Complexos de Coordenação/uso terapêutico , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Larva/efeitos dos fármacos , Larva/metabolismo , Camundongos , Camundongos Nus , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Pró-Fármacos/metabolismo , Pró-Fármacos/farmacologia , Pró-Fármacos/uso terapêutico , Proteoma/análise , Proteoma/efeitos dos fármacos , Proteômica , Peixe-Zebra/crescimento & desenvolvimento
13.
Dalton Trans ; 49(11): 3562-3569, 2020 Mar 17.
Artigo em Inglês | MEDLINE | ID: mdl-32123890

RESUMO

The combination of chemotherapeutic and photodynamic activities in an iridium-based molecular compound is less reported. Herein, two iridium complexes (IrC1 and IrC2) with ß-carboline alkaloid ligands were designed and synthesized. Both complexes exhibited high anticancer activities with IC50 values of around 1 µM in the dark against several cell lines tested. Notably, the cytotoxicity of these two complexes against lung cancer (A549) cells increased significantly under light (425 nm) irradiation, with phototoxicity index (PI) values of 120 and 93, respectively. They were specifically enriched in the mitochondria. Cell-based assays demonstrated that IrC1 induced an increase in intracellular reactive oxygen species (ROS) levels, reduction in ATP production, mitochondrial DNA damage, an increase in lipid peroxidation levels, and proteasomal activity inhibition. Under light conditions (in some cases a two-photon laser was also applied), these effects were greatly enhanced. Overall, we have demonstrated that these iridium complexes have dual activities of chemotherapy and photodynamic therapy, which may help to design new metal-based anticancer agents for combined chemo-photodynamic therapy.


Assuntos
Antineoplásicos/farmacologia , Complexos de Coordenação/farmacologia , Irídio/farmacologia , Fotoquimioterapia , Fármacos Fotossensibilizantes/farmacologia , Células A549 , Antineoplásicos/síntese química , Antineoplásicos/química , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Complexos de Coordenação/síntese química , Complexos de Coordenação/química , Dano ao DNA , DNA Mitocondrial/efeitos dos fármacos , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Irídio/química , Fármacos Fotossensibilizantes/síntese química , Fármacos Fotossensibilizantes/química
14.
Chem Commun (Camb) ; 55(70): 10472-10475, 2019 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-31411208

RESUMO

A mitochondria-targeted photodynamic therapy (PDT) agent was designed and synthesized. Upon light irradiation, it can produce photoacid and its photolysis products can further sensitize 1O2 generation, causing dual-mode (oxygen-independent and oxygen-dependent) photodynamic damage in mitochondria and killing cancer cells effectively even under hypoxic conditions.


Assuntos
Irídio/farmacologia , Mitocôndrias/metabolismo , Fotoquimioterapia , Linhagem Celular Tumoral , Cromatografia Líquida de Alta Pressão , Humanos , Espectroscopia de Prótons por Ressonância Magnética , Oxigênio Singlete/metabolismo
15.
Biomaterials ; 216: 119280, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31228705

RESUMO

Harsh photothermal temperatures, long-term body retention of nanoagents, elevated ROS and inflammation induction all threaten the normal tissues, thus hindering the translation of photothermal therapy (PTT) from bench to clinical practice. To resolve these problems, we have developed a disassembled theranostic nanodrug Qu-FeIIP based on the quercetin coordination. Herein, quercetin is not only the heat shock protein (Hsp 70) inhibitor but also the skeleton of Qu-FeIIP, realizing near-infrared light induced low-temperature PTT (45 °C) to ablate tumor completely without heat stress to normal tissues. Owing to the ROS scavenging ability of quercetin, Qu-FeIIP effectively reduces intracellular ROS and in vivo inflammatory factors (TNF-α, IL-6, IFN-γ) levels. Simultaneously, quercetin-Fe coordination is weakened when scavenging ROS, which triggers the Qu-FeIIP disassembling, resulting in effective clearance of nanoparticles from main organs 168 h post intravenous injection. Additionally, the photoacoustic and magnetic resonance dual-imaging capability of Qu-FeIIP offers excellent spatial resolution and imaging depth not only for precise tumor diagnosis but also for monitoring the nanodrug disassembling in vivo. Thus, Qu-FeIIP intrinsically integrates precise diagnosis, excellent low-temperature PTT efficacy, ROS elimination and anti-inflammatory action, dynamic disassembly and renal clearance ability into a single nanodrug, which is very promising for future clinical cancer treatment.


Assuntos
Anti-Inflamatórios/uso terapêutico , Antioxidantes/uso terapêutico , Compostos Ferrosos/uso terapêutico , Nanopartículas/uso terapêutico , Neoplasias/terapia , Quercetina/uso terapêutico , Animais , Temperatura Baixa , Crioterapia , Feminino , Humanos , Células MCF-7 , Imageamento por Ressonância Magnética , Camundongos Endogâmicos BALB C , Neoplasias/diagnóstico por imagem , Neoplasias/metabolismo , Técnicas Fotoacústicas , Fototerapia , Espécies Reativas de Oxigênio/metabolismo , Nanomedicina Teranóstica
16.
Dalton Trans ; 48(13): 4398-4404, 2019 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-30864598

RESUMO

Lysosomes play a critical role in the autophagy process. The impairment of lysosomes can affect the degradation of autophagic cargo, leading to the blockage of autophagy at the lysosomal stage and subsequent cell death. Herein, two phosphorescent Re(i) tricarbonyl complexes (Re1 and Re2) bearing ß-carboline derivatives have been synthesized and characterized. Both complexes show pH-dependent phosphorescence, which can be used to specifically image the lysosomes. Cytotoxicity assay shows that they exhibit high anticancer activity and are able to overcome cross-resistance to cisplatin. Re2 can induce autophagy, which is blocked at the lysosomal stage due to lysosomal dysfunction, such as the decrease of cathepsin B activity, subsequently leading to both autophagy and apoptosis dependent cell death. In vivo studies revealed that it could significantly inhibit tumor growth.


Assuntos
Antineoplásicos/farmacologia , Autofagia/efeitos dos fármacos , Carbolinas/farmacologia , Complexos de Coordenação/farmacologia , Lisossomos/efeitos dos fármacos , Rênio/química , Células A549 , Animais , Antineoplásicos/síntese química , Apoptose/efeitos dos fármacos , Carbolinas/síntese química , Sobrevivência Celular/efeitos dos fármacos , Cisplatino/uso terapêutico , Complexos de Coordenação/síntese química , Resistencia a Medicamentos Antineoplásicos , Xenoenxertos , Humanos , Concentração de Íons de Hidrogênio , Lisossomos/metabolismo , Camundongos Nus , Relação Estrutura-Atividade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...