Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 4.088
Filtrar
Mais filtros

Tipo de documento
Intervalo de ano de publicação
1.
Nature ; 591(7850): 471-476, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33627869

RESUMO

The behaviour of Dictyostelium discoideum depends on nutrients1. When sufficient food is present these amoebae exist in a unicellular state, but upon starvation they aggregate into a multicellular organism2,3. This biology makes D. discoideum an ideal model for investigating how fundamental metabolism commands cell differentiation and function. Here we show that reactive oxygen species-generated as a consequence of nutrient limitation-lead to the sequestration of cysteine in the antioxidant glutathione. This sequestration limits the use of the sulfur atom of cysteine in processes that contribute to mitochondrial metabolism and cellular proliferation, such as protein translation and the activity of enzymes that contain an iron-sulfur cluster. The regulated sequestration of sulfur maintains D. discoideum in a nonproliferating state that paves the way for multicellular development. This mechanism of signalling through reactive oxygen species highlights oxygen and sulfur as simple signalling molecules that dictate cell fate in an early eukaryote, with implications for responses to nutrient fluctuations in multicellular eukaryotes.


Assuntos
Dictyostelium/citologia , Dictyostelium/metabolismo , Privação de Alimentos/fisiologia , Nutrientes/metabolismo , Enxofre/metabolismo , Aminoácidos Essenciais/metabolismo , Aminoácidos Essenciais/farmacologia , Antioxidantes/metabolismo , Agregação Celular/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Respiração Celular/efeitos dos fármacos , Cisteína/química , Cisteína/metabolismo , Cisteína/farmacologia , Dictyostelium/efeitos dos fármacos , Glutationa/química , Glutationa/metabolismo , Glutationa/farmacologia , Proteínas Ferro-Enxofre/metabolismo , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Oxigênio/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/efeitos dos fármacos
2.
Mol Cell ; 70(4): 614-627.e7, 2018 05 17.
Artigo em Inglês | MEDLINE | ID: mdl-29754824

RESUMO

Bleach (HOCl) is a powerful oxidant that kills bacteria in part by causing protein aggregation. It inactivates ATP-dependent chaperones, rendering cellular proteins mostly dependent on holdases. Here we identified Escherichia coli CnoX (YbbN) as a folding factor that, when activated by bleach via chlorination, functions as an efficient holdase, protecting the substrates of the major folding systems GroEL/ES and DnaK/J/GrpE. Remarkably, CnoX uniquely combines this function with the ability to prevent the irreversible oxidation of its substrates. This dual activity makes CnoX the founding member of a family of proteins, the "chaperedoxins." Because CnoX displays a thioredoxin fold and a tetratricopeptide (TPR) domain, two structural motifs conserved in all organisms, this investigation sets the stage for the discovery of additional chaperedoxins in bacteria and eukaryotes that could cooperate with proteins from both the Hsp60 and Hsp70 families.


Assuntos
Proteínas de Escherichia coli/metabolismo , Escherichia coli/metabolismo , Glutationa/metabolismo , Proteínas de Choque Térmico/metabolismo , Chaperonas Moleculares/metabolismo , Oxirredutases atuantes sobre Doadores de Grupo Enxofre/metabolismo , Repetições de Tetratricopeptídeos , Tiorredoxinas/metabolismo , Sequência de Aminoácidos , Clareadores/farmacologia , Chaperonina 10/metabolismo , Chaperonina 60/metabolismo , Escherichia coli/efeitos dos fármacos , Escherichia coli/crescimento & desenvolvimento , Proteínas de Escherichia coli/química , Glutationa/química , Proteínas de Choque Térmico HSP40/metabolismo , Proteínas de Choque Térmico HSP70/metabolismo , Halogenação , Chaperonas Moleculares/química , Oxirredução , Oxirredutases atuantes sobre Doadores de Grupo Enxofre/química , Conformação Proteica , Desnaturação Proteica , Dobramento de Proteína , Homologia de Sequência , Tiorredoxinas/química
3.
J Biol Chem ; 300(5): 107293, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38636660

RESUMO

Unsaturated fatty acid ketones with αß,γδ conjugation are susceptible to Michael addition of thiols, with unresolved issues on the site of adduction and precise structures of the conjugates. Herein we reacted 13-keto-octadecadienoic acid (13-oxo-ODE or 13-KODE) with glutathione (GSH), N-acetyl-cysteine, and ß-mercaptoethanol and identified the adducts. HPLC-UV analyses indicated none of the products exhibit a conjugated enone UV chromophore, a result that conflicts with the literature and is relevant to the mass spectral interpretation of 1,4 versus 1,6 thiol adduction. Aided by the development of an HPLC solvent system that separates the GSH diastereomers and thus avoids overlap of signals in proton NMR experiments, we established the two major conjugates are formed by 1,6 addition of GSH at the 9-carbon of 13-oxo-ODE with the remaining double bond α to the thiol in the 10,11 position. N-acetyl cysteine reacts similarly, while ß-mercaptoethanol gives equal amounts of 1,4 and 1,6 addition products. Equine glutathione transferase catalyzed 1,6 addition of GSH to the two major diastereomers in 44:56 proportions. LC-MS in positive ion mode gives a product ion interpreted before as evidence of 1,4-thiol adduction, whereas here we find this ion using the authentic 1,6 adduct. LC-MS with negative ion APCI gave a fragment selective for 1,4 adduction. These results clarify the structures of thiol conjugates of a prototypical unsaturated keto-fatty acid and have relevance to the application of LC-MS for the structural analysis of keto-fatty acid glutathione conjugation.


Assuntos
Glutationa , Compostos de Sulfidrila , Glutationa/química , Glutationa/metabolismo , Compostos de Sulfidrila/química , Espectrometria de Massas/métodos , Cromatografia Líquida de Alta Pressão/métodos , Cromatografia Líquida/métodos , Mercaptoetanol/química , Espectrometria de Massa com Cromatografia Líquida
4.
J Biol Chem ; 300(5): 107289, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38636663

RESUMO

Vitamin B12 (cobalamin or Cbl) functions as a cofactor in two important enzymatic processes in human cells, and life is not sustainable without it. B12 is obtained from food and travels from the stomach, through the intestine, and into the bloodstream by three B12-transporting proteins: salivary haptocorrin (HC), gastric intrinsic factor, and transcobalamin (TC), which all bind B12 with high affinity and require proteolytic degradation to liberate Cbl. After intracellular delivery of dietary B12, Cbl in the aquo/hydroxocobalamin form can coordinate various nucleophiles, for example, GSH, giving rise to glutathionylcobalamin (GSCbl), a naturally occurring form of vitamin B12. Currently, there is no data showing whether GSCbl is recognized and transported in the human body. Our crystallographic data shows for the first time the complex between a vitamin B12 transporter and GSCbl, which compared to aquo/hydroxocobalamin, binds TC equally well. Furthermore, sequence analysis and structural comparisons show that TC recognizes and transports GSCbl and that the residues involved are conserved among TCs from different organisms. Interestingly, haptocorrin and intrinsic factor are not structurally tailored to bind GSCbl. This study provides new insights into the interactions between TC and Cbl.


Assuntos
Glutationa , Ratos , Transcobalaminas , Vitamina B 12 , Animais , Cristalografia por Raios X , Glutationa/metabolismo , Glutationa/análogos & derivados , Glutationa/química , Ligação Proteica , Transcobalaminas/metabolismo , Transcobalaminas/química , Vitamina B 12/metabolismo , Vitamina B 12/análogos & derivados , Vitamina B 12/química
5.
Nano Lett ; 24(25): 7800-7808, 2024 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-38870391

RESUMO

Metal nanoclusters feature a hierarchical structure, facilitating their ability to mimic enzyme-catalyzed reactions. However, the lack of true catalytic centers, compounded by tightly bound surface ligands hindering electron transfers to substrates, underscores the need for universal rational design methodologies to emulate the structure and mechanisms of natural enzymes. Motivated by the electron transfer in active centers with specific chemical structures, by integrating the peroxidase cofactor Fe-TCPP onto the surface of glutathione-stabilized gold nanoclusters (AuSG), we engineered AuSG-Fe-TCPP clusterzymes with a remarkable 39.6-fold enhancement in peroxidase-like activity compared to AuSG. Fe-TCPP not only mimics the active center structure, enhancing affinity to H2O2, but also facilitates the electron transfer process, enabling efficient H2O2 activation. By exemplifying the establishment of a detecting platform for trace H2O2 produced by ultrasonic cleaners, we substantiate that the bioinspired surface-ligand-engineered electron transfer can improve sensing performance with a wider linear range and lower detection limit.


Assuntos
Ouro , Peróxido de Hidrogênio , Nanopartículas Metálicas , Ouro/química , Peróxido de Hidrogênio/química , Transporte de Elétrons , Ligantes , Catálise , Nanopartículas Metálicas/química , Técnicas Biossensoriais/métodos , Glutationa/química
6.
Nano Lett ; 24(5): 1717-1728, 2024 Feb 07.
Artigo em Inglês | MEDLINE | ID: mdl-38270376

RESUMO

Surgery is the primary method to treat malignant melanoma; however, the residual microtumors that cannot be resected completely often trigger tumor recurrence, causing tumor-related mortality following melanoma resection. Herein, we developed a feasible strategy based on the combinational chemoimmunotherapy by cross-linking carboxymethyl chitosan (CMCS)-originated polymetformin (PolyMetCMCS) with cystamine to prepare stimuli-responsive nanogel (PMNG) owing to the disulfide bond in cystamine that can be cleaved by the massive glutathione (GSH) in tumor sites. Then, chemotherapeutic agent doxorubicin (DOX) was loaded in PMNG, which was followed by a hyaluronic acid coating to improve the overall biocompatibility and targeting ability of the prepared nanogel (D@HPMNG). Notably, PMNG effectively reshaped the tumor immune microenvironment by reprogramming tumor-associated macrophage phenotypes and recruiting intratumoral CD8+ T cells owing to the inherited immunomodulatory capability of metformin. Consequently, D@HPMNG treatment remarkably suppressed melanoma growth and inhibited its recurrence after surgical resection, proposing a promising solution for overcoming lethal melanoma recurrence.


Assuntos
Melanoma , Polietilenoglicóis , Polietilenoimina , Humanos , Nanogéis , Macrófagos Associados a Tumor , Cistamina , Linfócitos T CD8-Positivos , Doxorrubicina , Glutationa/química , Microambiente Tumoral , Linhagem Celular Tumoral
7.
Nano Lett ; 24(26): 8008-8016, 2024 Jul 03.
Artigo em Inglês | MEDLINE | ID: mdl-38912749

RESUMO

Piezoelectric dynamic therapy (PzDT) is an effective method of tumor treatment by using piezoelectric polarization to generate reactive oxygen species. In this paper, two-dimensional Cu-doped BiOCl nanosheets with surface vacancies are produced by the photoetching strategy. Under ultrasound, a built-in electric field is generated to promote the electron and hole separation. The separated carriers achieve O2 reduction and GSH oxidation, inducing oxidative stress. The bandgap of BiOCl is narrowed by introducing surface oxygen vacancies, which act as charge traps and facilitate the electron and hole separation. Meanwhile, Cu doping induces chemodynamic therapy and depletes GSH via the transformation from Cu(II) to Cu(I). Both in vivo and in vitro results confirmed that oxidative stress can be enhanced by exogenous ultrasound stimulation, which can cause severe damage to tumor cells. This work emphasizes the efficient strategy of doping engineering and defect engineering for US-activated PzDT under exogenous stimulation.


Assuntos
Cobre , Nanoestruturas , Oxigênio , Oxigênio/química , Cobre/química , Humanos , Nanoestruturas/química , Nanoestruturas/uso terapêutico , Animais , Camundongos , Neoplasias/terapia , Estresse Oxidativo/efeitos dos fármacos , Linhagem Celular Tumoral , Bismuto/química , Espécies Reativas de Oxigênio/metabolismo , Glutationa/química
8.
Nano Lett ; 24(25): 7764-7773, 2024 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-38864366

RESUMO

Inducing immunogenic cell death (ICD) during photothermal therapy (PTT) has the potential to effectively trigger photothermal immunotherapy (PTI). However, ICD induced by PTT alone is often limited by inefficient PTT, low immunogenicity of tumor cells, and a dysregulated redox microenvironment. Herein, we develop MoSe2 nanosheets with high-percentage metallic 1T phase and rich exposed active Mo centers through phase and defect engineering of MoSe2 as an effective nanoagent for PTI. The metallic 1T phase in MoSe2 nanosheets endows them with strong PTT performance, and the abundant exposed active Mo centers endow them with high activity for glutathione (GSH) depletion. The MoSe2-mediated high-performance PTT synergizing with efficient GSH depletion facilitates the release of tumor-associated antigens to induce robust ICD, thus significantly enhancing checkpoint blockade immunotherapy and activating systemic immune response in mouse models of colorectal cancer and triple-negative metastatic breast cancer.


Assuntos
Imunoterapia , Molibdênio , Terapia Fototérmica , Animais , Camundongos , Imunoterapia/métodos , Humanos , Molibdênio/química , Feminino , Linhagem Celular Tumoral , Nanoestruturas/química , Nanoestruturas/uso terapêutico , Glutationa/química , Glutationa/metabolismo , Neoplasias Colorretais/terapia , Neoplasias Colorretais/patologia , Neoplasias Colorretais/imunologia , Morte Celular Imunogênica/efeitos dos fármacos , Neoplasias de Mama Triplo Negativas/terapia , Neoplasias de Mama Triplo Negativas/imunologia , Neoplasias de Mama Triplo Negativas/patologia , Raios Infravermelhos , Selênio/química , Selênio/uso terapêutico , Fototerapia/métodos
9.
Med Res Rev ; 44(3): 1013-1054, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38140851

RESUMO

The burgeoning prodrug strategy offers a promising avenue toward improving the efficacy and specificity of cytotoxic drugs. Elevated intracellular levels of glutathione (GSH) have been regarded as a hallmark of tumor cells and characteristic feature of the tumor microenvironment. Considering the pivotal involvement of elevated GSH in the tumorigenic process, a diverse repertoire of GSH-triggered prodrugs has been developed for cancer therapy, facilitating the attenuation of deleterious side effects associated with conventional chemotherapeutic agents and/or the attainment of more efficacious therapeutic outcomes. These prodrug formulations encompass a spectrum of architectures, spanning from small molecules to polymer-based and organic-inorganic nanomaterial constructs. Although the GSH-triggered prodrugs have been gaining increasing interests, a comprehensive review of the advancements made in the field is still lacking. To fill the existing lacuna, this review undertakes a retrospective analysis of noteworthy research endeavors, based on a categorization of these molecules by their diverse recognition units (i.e., disulfides, diselenides, Michael acceptors, and sulfonamides/sulfonates). This review also focuses on explaining the distinct benefits of employing various chemical architecture strategies in the design of these prodrug agents. Furthermore, we highlight the potential for synergistic functionality by incorporating multiple-targeting conjugates, theranostic entities, and combinational treatment modalities, all of which rely on the GSH-triggering. Overall, an extensive overview of the emerging field is presented in this review, highlighting the obstacles and opportunities that lie ahead. Our overarching goal is to furnish methodological guidance for the development of more efficacious GSH-triggered prodrugs in the future. By assessing the pros and cons of current GSH-triggered prodrugs, we expect that this review will be a handful reference for prodrug design, and would provide a guidance for improving the properties of prodrugs and discovering novel trigger scaffolds for constructing GSH-triggered prodrugs.


Assuntos
Antineoplásicos , Pró-Fármacos , Humanos , Pró-Fármacos/farmacologia , Pró-Fármacos/química , Estudos Retrospectivos , Antineoplásicos/farmacologia , Antineoplásicos/química , Glutationa/química , Linhagem Celular Tumoral
10.
J Am Chem Soc ; 146(21): 14875-14888, 2024 May 29.
Artigo em Inglês | MEDLINE | ID: mdl-38750611

RESUMO

Most of the nanozymes have been obtained based on trial and error, for which the application is usually compromised by enzymatic activity regulation due to a vague catalytic mechanism. Herein, a hollow axial Mo-Pt single-atom nanozyme (H-MoN5@PtN4/C) is constructed by a two-tier template capture strategy. The axial ligand can induce Mo 4d orbital splitting, leading to a rearrangement of spin electrons (↑ ↑ → ↑↓) to regulate enzymatic activity. This creates catalase-like activity and enhances oxidase-like activity to catalyze cascade enzymatic reactions (H2O2 → O2 → O2•-), which can overcome tumor hypoxia and accumulate cytotoxic superoxide radicals (O2•-). Significantly, H-MoN5@PtN4/C displays destructive d-π conjugation between the metal and substrate to attenuate the restriction of orbitals and electrons. This markedly improves enzymatic performance (catalase-like and oxidase-like activity) of a Mo single atom and peroxidase-like properties of a Pt single atom. Furthermore, the H-MoN5@PtN4/C can deplete overexpressed glutathione (GSH) through a redox reaction, which can avoid consumption of ROS (O2•- and •OH). As a result, H-MoN5@PtN4/C can overcome limitations of a complex tumor microenvironment (TME) for tumor-specific therapy based on TME-activated catalytic activity.


Assuntos
Elétrons , Ligantes , Humanos , Platina/química , Catalase/química , Catalase/metabolismo , Catálise , Peróxido de Hidrogênio/química , Peróxido de Hidrogênio/metabolismo , Glutationa/química , Glutationa/metabolismo , Nanoestruturas/química
11.
Magn Reson Med ; 91(4): 1314-1322, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38044723

RESUMO

PURPOSE: To demonstrate J-difference coediting of glutamate using Hadamard encoding and reconstruction of Mescher-Garwood-edited spectroscopy (HERMES). METHODS: Density-matrix simulations of HERMES (TE 80 ms) and 1D J-resolved (TE 31-229 ms) of glutamate (Glu), glutamine (Gln), γ-aminobutyric acid (GABA), and glutathione (GSH) were performed. HERMES comprised four sub-experiments with editing pulses applied as follows: (A) 1.9/4.56 ppm simultaneously (ONGABA /ONGSH ); (B) 1.9 ppm only (ONGABA /OFFGSH ); (C) 4.56 ppm only (OFFGABA /ONGSH ); and (D) 7.5 ppm (OFFGABA /OFFGSH ). Phantom HERMES and 1D J-resolved experiments of Glu were performed. Finally, in vivo HERMES (20-ms editing pulses) and 1D J-resolved (TE 31-229 ms) experiments were performed on 137 participants using 3 T MRI scanners. LCModel was used for quantification. RESULTS: HERMES simulation and phantom experiments show a Glu-edited signal at 2.34 ppm in the Hadamard sum combination A+B+C+D with no overlapping Gln signal. The J-resolved simulations and phantom experiments show substantial TE modulation of the Glu and Gln signals across the TEs, whose average yields a well-resolved Glu signal closely matching the Glu-edited signal from the HERMES sum spectrum. In vivo quantification of Glu show that the two methods are highly correlated (p < 0.001) with a bias of ∼10%, along with similar between-subject coefficients of variation (HERMES/TE-averaged: ∼7.3%/∼6.9%). Other Hadamard combinations produce the expected GABA-edited (A+B-C-D) or GSH-edited (A-B+C-D) signal. CONCLUSION: HERMES simulation and phantom experiments show the separation of Glu from Gln. In vivo HERMES experiments yield Glu (without Gln), GABA, and GSH in a single MRS scan.


Assuntos
Ácido Glutâmico , Imageamento por Ressonância Magnética , Humanos , Espectroscopia de Ressonância Magnética/métodos , Glutamina , Glutationa/química , Ácido gama-Aminobutírico/química
12.
J Biol Inorg Chem ; 29(2): 251-264, 2024 03.
Artigo em Inglês | MEDLINE | ID: mdl-38494554

RESUMO

Organometallic η6-arene ruthenium(II) complexes with 3-chloro-6-(1H-pyrazol-1-yl)pyridazine (Ru1, Ru2, and Ru5) and 3-chloro-6-(3,5-dimethyl-1H-pyrazol-1-yl)pyridazine (Ru3-4) N,N' heterocyclic and η6-arene (cymene (Ru1-4) or toluene (Ru 5)) have been synthesized. The ruthenium(II) complexes have common "three-legged piano-stool" pseudo-octahedral structures known for half-sandwich complexes. Evolution of their UV-Visible absorption spectra in PBS buffer or DMSO over 24 h confirmed their good solvolysis stability. Titrations of the complexes with the calf thymus DNA (CT-DNA) were monitored using UV-Visible absorption and fluorescence spectroscopies. The complexes interact moderately with CT-DNA and their binding constants are in the order of 104 M-1. Competitive binding of the complexes to a DNA-Hoechst 33,258 depicted competitive displacement of Hoechst from DNA's minor grooves. These complexes bind to glutathione forming GSH-adducts through S coordination by replacement of a halide, with the iodo-analogues having higher binding constants than the chloro-complexes. Cyclic voltammograms of the complexes exhibited one electron-transfer quasi-reversible process. Trends in the molecular docking data of Ru1-5/DNA were similar to those for DNA binding constants. Of the five, only Ru1, Ru3 and Ru5 showed some activity (moderate) against the MCF-7 breast cancer cells with IC50 values in the range of 59.2-39.9 for which Ru5 was the most active. However, the more difficult-to-treat cell line, MDA-MB 231 cell was recalcitrant to the treatment by these complexes.


Assuntos
Antineoplásicos , Complexos de Coordenação , DNA , Glutationa , Rutênio , DNA/química , DNA/metabolismo , Humanos , Rutênio/química , Ligantes , Complexos de Coordenação/química , Complexos de Coordenação/farmacologia , Complexos de Coordenação/síntese química , Antineoplásicos/farmacologia , Antineoplásicos/química , Antineoplásicos/síntese química , Glutationa/química , Glutationa/metabolismo , Bovinos , Pirazóis/química , Pirazóis/farmacologia , Pirazóis/síntese química , Animais , Estrutura Molecular , Ensaios de Seleção de Medicamentos Antitumorais , Proliferação de Células/efeitos dos fármacos , Células MCF-7 , Linhagem Celular Tumoral
13.
Chemistry ; 30(28): e202400021, 2024 May 17.
Artigo em Inglês | MEDLINE | ID: mdl-38477386

RESUMO

The development of novel and effective drug delivery systems aimed at enhancing therapeutic profile and efficacy of therapeutic agents is a critical challenge in modern medicine. This study presents an intelligent drug delivery system based on self-assembled two-dimensional peptide nanosheets (2D PNSs). Leveraging the tunable properties of amino acid structures and sequences, we design a peptide with the sequence of Fmoc-FKKGSHC, which self-assembles into 2D PNSs with uniform structure, high biocompatibility, and excellent degradability. Covalent attachment of thiol-modified doxorubicin (DOX) drugs to 2D PNSs via disulfide bond results in the peptide-drug conjugates (PDCs), which is denoted as PNS-SS-DOX. Subsequently, the PDCs are encapsulated within the injectable, thermosensitive chitosan (CS) hydrogels for drug delivery. The designed drug delivery system demonstrates outstanding pH-responsiveness and sustained drug release capabilities, which are facilitated by the characteristics of the CS hydrogels. Meanwhile, the covalently linked disulfide bond within the PNS-SS-DOX is responsive to intracellular glutathione (GSH) within tumor cells, enabling controlled drug release and significantly inhibiting the cancer cell growth. This responsive peptide-drug conjugate based on a 2D peptide nanoplatform paves the way for the development of smart drug delivery systems and has bright prospects in the future biomedicine field.


Assuntos
Quitosana , Doxorrubicina , Liberação Controlada de Fármacos , Glutationa , Hidrogéis , Nanoestruturas , Peptídeos , Hidrogéis/química , Doxorrubicina/química , Doxorrubicina/farmacologia , Doxorrubicina/administração & dosagem , Quitosana/química , Glutationa/química , Peptídeos/química , Humanos , Nanoestruturas/química , Sistemas de Liberação de Medicamentos , Portadores de Fármacos/química , Concentração de Íons de Hidrogênio
14.
Mol Pharm ; 21(7): 3434-3446, 2024 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-38781419

RESUMO

Chemodynamic therapy (CDT) is a novel antitumor strategy that employs Fenton or Fenton-like reactions to generate highly toxic hydroxyl radical (OH•) from hydrogen peroxide (H2O2) for inducing tumor cell death. However, the antitumor efficacy of the CDT strategy is harshly limited by the redox homeostasis of tumor cells; especially the OH • is easily scavenged by glutathione (GSH) and the intracellular H2O2 level is insufficient in the tumor cells. Herein, we propose the Mn2+-menadione (also known as vitamin K3, MK3) cascade biocatalysis strategy to disrupt the redox homeostasis of tumor cells and induce a OH• storm, resulting in enhanced CDT effect. A nanoliposome encapsulating Mn-MK3 (Mn-MK3@LP) was prepared for the treatment of hepatic tumors in this study. After Mn-MK3@LPs were taken up by tumor cells, menadione could facilitate the production of intracellular H2O2 via redox cycling, and further the cytotoxic OH • burst was induced by Mn2+-mediated Fenton-like reaction. Moreover, high-valent manganese ions were reduced by GSH and the depletion of GSH further disrupted the redox homeostasis of tumor cells, thus achieving synergistically enhanced CDT. Overall, both cellular and animal experiments confirmed that the Mn-MK3@LP cascade biocatalysis nanoliposome exhibited excellent biosafety and tumor suppression efficacy. This study may provide deep insights for developing novel CDT-based strategies for tumor therapy.


Assuntos
Glutationa , Peróxido de Hidrogênio , Radical Hidroxila , Vitamina K 3 , Animais , Radical Hidroxila/química , Radical Hidroxila/metabolismo , Humanos , Camundongos , Glutationa/metabolismo , Glutationa/química , Vitamina K 3/química , Vitamina K 3/farmacologia , Biocatálise , Linhagem Celular Tumoral , Manganês/química , Oxirredução/efeitos dos fármacos , Camundongos Endogâmicos BALB C , Lipossomos/química , Ensaios Antitumorais Modelo de Xenoenxerto , Neoplasias Hepáticas/tratamento farmacológico , Neoplasias Hepáticas/metabolismo , Camundongos Nus , Células Hep G2 , Antineoplásicos/farmacologia , Antineoplásicos/química , Nanopartículas/química , Ferro
15.
Mol Pharm ; 21(5): 2394-2405, 2024 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-38647653

RESUMO

Doxorubicin (DOX) is one of the most commonly used anticancer drugs; however, its clinical application is greatly limited due to its toxicity and chemotherapy resistance. The delivery of DOX by liposomes (Lipos) can improve the blood circulation time in vivo and reduce toxic side effects, but the drug's accumulation in the tumor is often insufficient for effective treatment. In this study, we present a calcium cross-linked liposome gel for the encapsulation of DOX, demonstrating its superior long-term release capabilities compared to conventional Lipos. By leveraging this enhanced long-term release, we can enhance drug accumulation within tumors, ultimately leading to improved antitumor efficacy. Lipos were prepared using the thin-film dispersion method in this study. We utilized the ion-responsiveness of glutathione-gelatin (GSH-GG) to form the gel outside the Lipos and named the nanoparticles coated with GSH-GG on the outside of Lipos as Lipos@GSH-GG. The average size of Lipos@GSH-GG was around 342.9 nm, with a negative charge of -25.6 mV. The in vitro experiments revealed that Lipos@GSH-GG exhibited excellent biocompatibility and slower drug release compared to conventional Lipos. Further analysis of cellular uptake and cytotoxicity demonstrated that Lipos@GSH-GG loading DOX (DOX&Lipos@GSH-GG) exhibited superior long-term release effects and lower toxic side effects compared to Lipos loading DOX (DOX&Lipos). Additionally, the findings regarding the long-term release effect in vivo and the tumor accumulation within tumor-bearing mice of Lipos@GSH-GG suggested that, compared to Lipos, it demonstrated superior long-term release capabilities and achieved greater drug accumulation within tumors. In vivo antitumor efficacy experiments showed that DOX&Lipos@GSH-GG demonstrated superior antitumor efficacy to DOX&Lipos. Our study highlights Lipos@GSH-GG as a promising nanocarrier with the potential to enhance efficacy and safety by means of long-term release effects and may offer an alternative approach for effective antitumor therapy in the future.


Assuntos
Cálcio , Doxorrubicina , Liberação Controlada de Fármacos , Glutationa , Lipossomos , Doxorrubicina/farmacologia , Doxorrubicina/química , Doxorrubicina/administração & dosagem , Doxorrubicina/farmacocinética , Animais , Camundongos , Lipossomos/química , Humanos , Cálcio/química , Cálcio/metabolismo , Glutationa/química , Feminino , Géis/química , Gelatina/química , Camundongos Nus , Nanopartículas/química , Camundongos Endogâmicos BALB C , Linhagem Celular Tumoral , Ensaios Antitumorais Modelo de Xenoenxerto , Antibióticos Antineoplásicos/administração & dosagem , Antibióticos Antineoplásicos/farmacologia , Antibióticos Antineoplásicos/química , Antibióticos Antineoplásicos/farmacocinética , Preparações de Ação Retardada/química , Preparações de Ação Retardada/farmacocinética , Antineoplásicos/farmacologia , Antineoplásicos/química , Antineoplásicos/administração & dosagem , Reagentes de Ligações Cruzadas/química , Sistemas de Liberação de Medicamentos/métodos
16.
Langmuir ; 40(23): 12167-12178, 2024 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-38808371

RESUMO

Gold nanoparticles (AuNPs) hold promise in biomedicine, but challenges like aggregation, protein corona formation, and insufficient biocompatibility must be thoroughly addressed before advancing their clinical applications. Designing AuNPs with specific protein corona compositions is challenging, and strategies for corona elimination, such as coating with polyethylene glycol (PEG), have limitations. In this study, we introduce a commercially available zwitterionic derivative of glutathione, glutathione monoethyl ester (GSHzwt), for the surface coating of colloidal AuNPs. Particles coated with GSHzwt were investigated alongside four other AuNPs coated with various ligands, including citrate ions, tiopronin, glutathione, cysteine, and PEG. We then undertook a head-to-head comparison of these AuNPs to assess their behavior in biological fluid. GSHzwt-coated AuNPs exhibited exceptional resistance to aggregation and protein adsorption. The particles could also be readily functionalized with biotin and interact with streptavidin receptors in human plasma. Additionally, they exhibited significant blood compatibility and noncytotoxicity. In conclusion, GSHzwt provides a practical and easy method for the surface passivation of AuNPs, creating "stealth" particles for potential clinical applications.


Assuntos
Glutationa , Ouro , Nanopartículas Metálicas , Propriedades de Superfície , Ouro/química , Nanopartículas Metálicas/química , Glutationa/química , Humanos , Tamanho da Partícula , Adsorção , Materiais Revestidos Biocompatíveis/química , Materiais Revestidos Biocompatíveis/farmacologia
17.
Langmuir ; 40(21): 11098-11105, 2024 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-38739904

RESUMO

Disulfide bonding has attracted intense interest in the tumor intracellular microenvironment-activated drug delivery systems (DDSs) in the last decades. Although various molecular structures of redox-responsive disulfide-containing DDSs have been developed, no investigation was reported on the effect of aggregation structures. Here, the effect of aggregation structures on pH/GSH dual-triggered drug release was investigated with the simplest pH/GSH dual-triggered doxorubicin-based drug self-delivery system (DSDS), the disulfide/α-amide-bridged doxorubicin dimeric prodrug (DDOX), as a model. By fast precipitation or slow self-assembly, DDOX nanoparticles were obtained. With similar diameters, they exhibited different pH/GSH dual-triggered drug releases, demonstrating the effect of aggregation structures. The π-π stacking in different degrees was revealed by the UV-vis, fluorescence, and BET analysis of the DDOX nanoparticles. The effect of the π-π stacking between the dimeric prodrug and its activated products on drug release was also explored with the molecular simulation approach. The finding opens new ideas in the design of high-performance DDSs for future precise tumor treatment.


Assuntos
Dissulfetos , Doxorrubicina , Liberação Controlada de Fármacos , Glutationa , Pró-Fármacos , Pró-Fármacos/química , Pró-Fármacos/farmacologia , Doxorrubicina/química , Doxorrubicina/farmacologia , Concentração de Íons de Hidrogênio , Dissulfetos/química , Glutationa/química , Amidas/química , Nanopartículas/química , Dimerização , Portadores de Fármacos/química
18.
Analyst ; 149(13): 3522-3529, 2024 Jun 24.
Artigo em Inglês | MEDLINE | ID: mdl-38787653

RESUMO

Bioinspired nanochannel-based sensors have elicited significant interest because of their excellent sensing performance, and robust mechanical and tunable chemical properties. However, the existing designs face limitations due to material constraints, which hamper broader application possibilities. Herein, a heteromembrane system composed of a periodic mesoporous organosilica (PMO) layer with three-dimensional (3D) network nanochannels is constructed for glutathione (GSH) detection. The unique hierarchical pore architecture provides a large surface area, abundant reaction sites and plentiful interconnected pathways for rapid ionic transport, contributing to efficient and sensitive detection. Moreover, the thioether groups in nanochannels can be selectively cleaved by GSH to generate hydrophilic thiol groups. Benefiting from the increased hydrophilic surface, the proposed sensor achieves efficient GSH detection with a detection limit of 1.2 µM by monitoring the transmembrane ionic current and shows good recovery ranges in fetal bovine serum sample detection. This work paves an avenue for designing and fabricating nanofluidic sensing systems for practical and biosensing applications.


Assuntos
Glutationa , Limite de Detecção , Compostos de Organossilício , Glutationa/química , Glutationa/análise , Glutationa/sangue , Porosidade , Compostos de Organossilício/química , Animais , Bovinos , Técnicas Biossensoriais/métodos , Membranas Artificiais , Técnicas Eletroquímicas/métodos
19.
Analyst ; 149(12): 3372-3379, 2024 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-38712551

RESUMO

A series of novel near-infrared (NIR) xanthene-chalcone fluorophores were constructed through a modular synthesis with the electron-donating xanthene moiety and the electron-withdrawing chalcone moiety. These fluorophores are convenient for fluorescence imaging in living cells, benefiting from their NIR emissions (650-710 nm), large Stokes shifts (>100 nm), moderate quantum yields and low cytotoxicity. The substituted hydroxyl group of the xanthene-chalcone fluorophore HCA-E facilitates the development of multifunctional fluorescent probes. As an example, a highly sensitive and selective probe N-HCA-E for glutathione (GSH) detection was developed based on the fluorophore HCA-E. A 4-nitrobenzenesulfonyl (4-Ns) group was introduced to cage the hydroxyl group of HCA-E, which was used as a selective recognition site for the thiol of GSH and an effective fluorescence quencher. Probe N-HCA-E revealed NIR "turn-on" fluorescence (709 nm) for endogenous and exogenous GSH detection in lysosomes with a large Stokes shift (129 nm) and high anti-interference ability.


Assuntos
Corantes Fluorescentes , Glutationa , Imagem Óptica , Xantenos , Corantes Fluorescentes/química , Corantes Fluorescentes/toxicidade , Corantes Fluorescentes/síntese química , Xantenos/química , Humanos , Glutationa/química , Imagem Óptica/métodos , Chalconas/química , Células HeLa , Lisossomos/química , Lisossomos/metabolismo , Raios Infravermelhos , Chalcona/química
20.
Inorg Chem ; 63(23): 10455-10465, 2024 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-38743433

RESUMO

Organomercurials (RHg+), especially methylmercury (MeHg+) and ethylmercury (EtHg+), are considered to be more neurotoxic than the inorganic counterpart (Hg2+). They cause massive DNA damage in cells, especially in neurons, where cellular glutathione (GSH) levels are significantly low. However, the mechanism by which RHg+ exerts massive DNA damage at cytotoxic concentrations in brain cells remains obscure. In this study, we investigated the effect of RHg+ on the structural and electronic properties of nucleosides and its effects on DNA damage. The direct interaction of RHg+ with the nucleoside significantly weakens N-glycosidic bonds, decreases the C-H bond energy of sugar moieties, and increases the electrophilicity of the C8-center of purine bases. As a consequence, RHg+-conjugated DNA molecules are extremely labile and highly sensitive to any nucleophiles/radicals present in GSH-depleted cells and, thus, undergo enhanced oxidative and unusual alkylative DNA damage. We also report a functional model of organomercurial lyase, which showed excellent cytoprotective effect against RHg+-induced cytotoxicity; this reverses the activity of glutathione reductase inhibited by MeHgCl and ceases oxidative and alkylating DNA damage. This intriguing finding provides new mechanistic insight into the mode of action of organomercurials in GSH-depleted cells and their adverse effects on individuals with neurodegenerative disorders associated with oxidative stress.


Assuntos
Dano ao DNA , Glutationa , Compostos de Metilmercúrio , Compostos de Metilmercúrio/farmacologia , Compostos de Metilmercúrio/química , Glutationa/metabolismo , Glutationa/química , Dano ao DNA/efeitos dos fármacos , Humanos , DNA/química , DNA/efeitos dos fármacos , Estrutura Molecular , Animais , Sobrevivência Celular/efeitos dos fármacos , Teoria da Densidade Funcional
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA