Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 101
Filtrar
1.
Int J Pharm ; 657: 124187, 2024 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-38697585

RESUMEN

Drug repositioning is a high-priority and feasible strategy in the field of oncology research, where the unmet medical needs are continuously unbalanced. Disulfiram is a potential non-chemotherapeutic, adjuvant anticancer agent. However, the clinical translation is limited by the drug's poor bioavailability. Therefore, the molecular encapsulation of disulfiram with cyclodextrins is evaluated to enhance the solubility and stability of the drug. The present work describes for the first time the complexation of disulfiram with randomly methylated-ß-cyclodextrin. A parallel analytical andin vitrobiological comparison of disulfiram inclusion complexes with hydroxypropyl-ß-cyclodextrin, randomly methylated-ß-cyclodextrin and sulfobutylether-ß-cyclodextrin is conducted. A significant drug solubility enhancement by about 1000-folds and fast dissolution in 1 min is demonstrated. Thein vitrodissolution-permeation studies and proliferation assays demonstrate the solubility-dependent efficacy of the drug. Throughout the different cancer cell lines' characteristics and disulfiram unspecific antitumoral activity, the inhibitory efficacy of the cyclodextrin encapsulated drug on melanoma (IC50 about 100 nM) and on glioblastoma (IC50 about 7000 nM) cell lines differ by a magnitude. This pre-formulation screening experiment serves as a proof of concept of using cyclodextrin encapsulation as a platform tool for further drug delivery development in repositioning areas.


Asunto(s)
Antineoplásicos , Disulfiram , Reposicionamiento de Medicamentos , Solubilidad , beta-Ciclodextrinas , Disulfiram/farmacología , Disulfiram/química , Disulfiram/administración & dosificación , Humanos , Antineoplásicos/farmacología , Antineoplásicos/química , Antineoplásicos/administración & dosificación , Línea Celular Tumoral , beta-Ciclodextrinas/química , 2-Hidroxipropil-beta-Ciclodextrina/química , Ciclodextrinas/química , Ciclodextrinas/farmacología , Proliferación Celular/efectos de los fármacos , Composición de Medicamentos/métodos , Glioblastoma/tratamiento farmacológico
2.
Biomacromolecules ; 25(5): 2770-2779, 2024 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-38687975

RESUMEN

Drug repurposing uses approved drugs as candidate anticancer therapeutics, harnesses previous research and development efforts, and benefits from available clinically suitable formulations and evidence of patient tolerability. In this work, the drug used clinically to treat chronic alcoholism, disulfiram (DSF), was studied for its antitumor efficacy in a copper-dependent manner. The combination of DSF and copper could achieve a tumor cell growth inhibition effect comparable to those of 5-fluorouracil and taxol on head and neck cancer cells. Both bulk dendrimer hydrogel and microsized dendrimer hydrogel particles were utilized for the localized sustained release of copper in the tumor site. The localized sustained release of copper facilitated the tumor inhibition effect following intratumoral injection in a mouse's head and neck cancer model.


Asunto(s)
Cobre , Preparaciones de Acción Retardada , Disulfiram , Neoplasias de Cabeza y Cuello , Disulfiram/farmacología , Disulfiram/química , Disulfiram/administración & dosificación , Animales , Cobre/química , Cobre/farmacología , Ratones , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Neoplasias de Cabeza y Cuello/patología , Humanos , Preparaciones de Acción Retardada/química , Preparaciones de Acción Retardada/farmacología , Línea Celular Tumoral , Antineoplásicos/farmacología , Antineoplásicos/química , Antineoplásicos/administración & dosificación , Ensayos Antitumor por Modelo de Xenoinjerto , Ratones Desnudos
3.
Adv Healthc Mater ; 13(11): e2303955, 2024 04.
Artículo en Inglés | MEDLINE | ID: mdl-38271271

RESUMEN

Traditional chemotherapy has faced tough challenges of systemic toxicity, hypoxia resistance, and inadequacy of monotherapy. Developing the tumor-specific O2-supply-enhanced chemotherapy without toxic drugs while combing other precise treatments can substantially improve therapeutic efficacy. Herein, a CeO2-enriched CuO nanozyme with O2 supply and oxidative stress amplification for tumor-specific disulfiram (DSF) chemotherapy and intensified chemodynamic therapy by synergistic in situ "nontoxicity-toxicity" activation is developed. Notably, CeO2 can not only act as a morphological "regulator," but also serve as a cascaded enzyme-mimetic catalyst via tumor-microenvironment-responsive cascaded-logical programmable valence conversion. Once internalized inside tumor cells, the nanozyme can be degraded by lysosomal acidity to release nontoxic DSF and Cu2+, which can trigger in situ "Cu2+-DSF" chelation, generating a highly toxic Cu(DTC)2 for in situ chemotherapy. Moreover, the enriched CeO2 with catalase-mimetic activity can decompose the endogenous H2O2 into O2, which can relieve the hypoxia to enhance the chemotherapeutic efficacy. Furthermore, the simultaneously generated Ce3+ can exert peroxidase-mimetic activity to catalyze H2O2 into hydroxyl radicals (•OH) for chemodynamic therapy. This Fenton-like chemistry is accompanied by the regeneration of Ce4+, which can deplete the intracellular overproduced GSH to amplify the oxidative stress. Therefore, this nanozyme can provide an alternative to precise cancer treatment.


Asunto(s)
Cerio , Cobre , Disulfiram , Estrés Oxidativo , Microambiente Tumoral , Disulfiram/farmacología , Disulfiram/química , Cerio/química , Cerio/farmacología , Cobre/química , Microambiente Tumoral/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Humanos , Animales , Ratones , Línea Celular Tumoral , Oxígeno/química , Oxígeno/metabolismo , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Antineoplásicos/química , Antineoplásicos/farmacología , Peróxido de Hidrógeno/química , Peróxido de Hidrógeno/metabolismo
4.
Redox Biol ; 69: 103007, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38150993

RESUMEN

Hepatocellular carcinoma (HCC) is one of the most prevalent malignant tumors and the fourth leading cause of cancer-related death globally, which is characterized by complicated pathophysiology, high recurrence rate, and poor prognosis. Our previous study has demonstrated that disulfiram (DSF)/Cu could be repurposed for the treatment of HCC by inducing ferroptosis. However, the effectiveness of DSF/Cu may be compromised by compensatory mechanisms that weaken its sensitivity. The mechanisms underlying these compensatory responses are currently unknown. Herein, we found DSF/Cu induces endoplasmic reticulum stress with disrupted ER structures, increased Ca2+ level and activated expression of ATF4. Further studies verified that DSF/Cu induces both ferroptosis and cuproptosis, accompanied by the depletion of GSH, elevation of lipid peroxides, and compensatory increase of xCT. Comparing ferroptosis and cuproptosis, it is interesting to note that GSH acts at the crossing point of the regulation network and therefore, we hypothesized that compensatory elevation of xCT may be a key aspect of the therapeutic target. Mechanically, knockdown of ATF4 facilitated the DSF/Cu-induced cell death and exacerbated the generation of lipid peroxides under the challenge of DSF/Cu. However, ATF4 knockdown was unable to block the compensatory elevation of xCT and the GSH reduction. Notably, we found that DSF/Cu induced the accumulation of ubiquitinated proteins, promoted the half-life of xCT protein, and dramatically dampened the ubiquitination-proteasome mediated degradation of xCT. Moreover, both pharmacologically and genetically suppressing xCT exacerbated DSF/Cu-induced cell death. In conclusion, the current work provides an in-depth study of the mechanism of DSF/Cu-induced cell death and describes a framework for the further understanding of the crosstalk between ferroptosis and cuproptosis. Inhibiting the compensatory increase of xCT renders HCC cells more susceptible to DSF/Cu, which may provide a promising synergistic strategy to sensitize tumor therapy and overcome drug resistance, as it activates different programmed cell death.


Asunto(s)
Carcinoma Hepatocelular , Ferroptosis , Neoplasias Hepáticas , Humanos , Disulfiram/farmacología , Disulfiram/química , Cobre/química , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/genética , Línea Celular Tumoral , Peróxidos Lipídicos , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/genética
5.
Int J Mol Sci ; 23(21)2022 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-36362068

RESUMEN

Differentiated thyroid carcinomas (DTCs), which have papillary and follicular types, are common endocrine malignancies worldwide. Cancer stem cells (CSCs) are a particular type of cancer cells within bulk tumors involved in cancer initiation, drug resistance, and metastasis. Cells with high intracellular aldehyde hydrogenase (ALDH) activity are a population of CSCs in DTCs. Disulfiram (DSF), an ALDH inhibitor used for the treatment of alcoholism, reportedly targets CSCs in various cancers when combined with copper. This study reported for the first time that DSF/copper can inhibit the proliferation of papillary and follicular DTC lines. DSF/copper suppressed thyrosphere formation, indicating the inhibition of CSC activity. Molecular mechanisms of DSF/copper involved downregulating the expression of B lymphoma Mo-MLV insertion region 1 homolog (BMI1) and cell cycle-related proteins, including cyclin B2, cyclin-dependent kinase (CDK) 2, and CDK4, in a dose-dependent manner. BMI1 overexpression diminished the inhibitory effect of DSF/copper in the thyrosphere formation of DTC cells. BMI1 knockdown by RNA interference in DTC cells also suppressed the self-renewal capability. DSF/copper could inhibit the nuclear localization and transcriptional activity of c-Myc and the binding of E2F1 to the BMI1 promoter. Overexpression of c-Myc or E2F1 further abolished the inhibitory effect of DSF/copper on BMI1 expression, suggesting that the suppression of c-Myc and E2F1 by DSF/copper was involved in the downregulation of BMI1 expression. In conclusion, DSF/copper targets CSCs in DTCs by inhibiting c-Myc- or E2F1-mediated BMI1 expression. Therefore, DSF is a potential therapeutic agent for future therapy in DTCs.


Asunto(s)
Cobre , Disulfiram , Células Madre Neoplásicas , Neoplasias de la Tiroides , Humanos , Aldehído Deshidrogenasa/metabolismo , Línea Celular Tumoral , Cobre/química , Cobre/farmacología , Disulfiram/farmacología , Disulfiram/química , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/metabolismo , Complejo Represivo Polycomb 1/antagonistas & inhibidores , Complejo Represivo Polycomb 1/metabolismo , Neoplasias de la Tiroides/tratamiento farmacológico , Neoplasias de la Tiroides/metabolismo
6.
Int J Pharm ; 621: 121788, 2022 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-35504431

RESUMEN

The old alcohol-aversion drug disulfiram (DSF) has aroused wide attention as a drug repurposing strategy in terms of cancer therapy because of the high antitumor efficacy in combination with copper ion. However, numerous defects of DSF (e.g., the short half-life and acid instability) have limited the application in cancer treatment. Cu (DDC)2, the complex of diethyldithiocarbamate (DDC, DSF metabolite) and Cu2+, have been proven as the vital active component on cancer, which have aroused the attention of researchers from DSF to Cu (DDC)2. However, the poor water solubility of Cu (DDC)2 increase more difficulties to the treatment and in-depth investigations of Cu (DDC)2. In this study, sphingomyelin (SM)-based PEGylated liposomes (SM/Chol/DSPE-mPEG2000 (55:40:5, mole%)) were produced as the carriers for Cu (DDC)2 delivery to enhance the water solubility. DDC was added to Cu-containing liposomes with a higher encapsulation efficiency of more than 90%, and it reacted with Cu2+ to synthesize Cu (DDC)2. Due to the high phase transition temperature of SM and strong intermolecular hydrogen bonds with cholesterol, SM-based liposomes would be conducive to enhancing the stability of Cu (DDC)2 and preventing drug leakage during delivery. As proven by pharmacokinetic studies, loading Cu (DDC)2 into liposomes improve bioavailability, and the area under the curve (AUC0-t) and the mean elimination half-life (t1/2) increased 1.9-time and 1.3-time to those of free Cu (DDC)2, respectively. Furthermore, the anticancer effect of Cu (DDC)2 was enhanced by the liposomal encapsulation, thus resulting in remarkable cell apoptosis in vitro and a tumor-inhibiting rate of 77.88% in vivo. Thus, it was concluded that Cu (DDC)2 liposomes could be promising in cancer treatment.


Asunto(s)
Liposomas , Neoplasias , Descarboxilasas de Aminoácido-L-Aromático/uso terapéutico , Línea Celular Tumoral , Cobre/química , Disulfiram/química , Ditiocarba/química , Ditiocarba/farmacocinética , Humanos , Liposomas/química , Neoplasias/tratamiento farmacológico , Esfingomielinas/uso terapéutico , Agua
7.
J Colloid Interface Sci ; 615: 517-526, 2022 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-35152072

RESUMEN

Off-target toxicity remains a major limitation of current cancer therapy, necessitating an alternative precision approach to treat cancers. Herein, a tumor microenvironment (TME)-triggered anticancer strategy was developed by constructing an anti-alcoholism drug disulfiram (DSF)-loaded, Cu-doped zeolite imidazolate frameworks-8 (DSF-Cu/ZIF-8) nanoparticle followed by PEGylation (PEG-DSF-Cu/ZIF-8) to realize in situ generation of cytotoxic compounds specifically in TME. The PEG-DSF-Cu/ZIF-8 demonstrated excellent hydrolytic stability in normal physiological conditions, guaranteeing the minimized off-target release of disulfiram and Cu ions. Under the TME condition, the PEG-DSF-Cu/ZIF-8 exhibited acidity-triggered biodegradation and the associated payload release, through which low-toxic compounds (disulfiram and Cu2+ ions) were converted to highly cytotoxic Cu-chelate product to kill cells specifically in TME. Tumor-sensitive anti-cancer performance was further enhanced by hydroxyl radical generation via TME-responsive Fenton-like reactions catalyzed by Cu+ presenting in the PEG-DSF-Cu/ZIF-8 structure and Cu+ produced during formation of the chelate product. Anti-cancer therapeutic evaluation was performed in 2D 4T1 tumor cell culture and 3D 4T1 tumor spheroids, and demonstrated highly TME-responsive, low-dose induced anti-cancer effect. This proof-of-concept work provides a nanoparticle-based drug repurposing strategy by developing a tumor-sensitive anti-cancer agent for low-toxic and efficacious cancer therapy.


Asunto(s)
Estructuras Metalorgánicas , Neoplasias , Línea Celular Tumoral , Cobre/química , Disulfiram/química , Disulfiram/farmacología , Estructuras Metalorgánicas/farmacología , Neoplasias/tratamiento farmacológico , Microambiente Tumoral
8.
Molecules ; 27(2)2022 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-35056791

RESUMEN

Aldehyde dehydrogenase-1a1 (ALDH1a1), the enzyme responsible for the oxidation of retinal into retinoic acid, represents a key therapeutic target for the treatment of debilitating disorders such as cancer, obesity, and inflammation. Drugs that can inhibit ALDH1a1 include disulfiram, an FDA-approved drug to treat chronic alcoholism. Disulfiram, by carbamylation of the catalytic cysteines, irreversibly inhibits ALDH1a1 and ALDH2. The latter is the isozyme responsible for important physiological processes such as the second stage of alcohol metabolism. Given the fact that ALDH1a1 has a larger substrate tunnel than that in ALDH2, replacing disulfiram ethyl groups with larger motifs will yield selective ALDH1a1 inhibitors. We report herein the synthesis of new inhibitors of ALDH1a1 where (hetero)aromatic rings were introduced into the structure of disulfiram. Most of the developed compounds retained the anti-ALDH1a1 activity of disulfiram; however, they were completely devoid of inhibitory activity against ALDH2.


Asunto(s)
Inhibidores del Acetaldehído Deshidrogenasa/química , Inhibidores del Acetaldehído Deshidrogenasa/farmacología , Familia de Aldehído Deshidrogenasa 1/antagonistas & inhibidores , Disulfiram/química , Disulfiram/farmacología , Retinal-Deshidrogenasa/antagonistas & inhibidores , Inhibidores del Acetaldehído Deshidrogenasa/síntesis química , Inhibidores del Acetaldehído Deshidrogenasa/metabolismo , Familia de Aldehído Deshidrogenasa 1/química , Familia de Aldehído Deshidrogenasa 1/metabolismo , Aldehído Deshidrogenasa Mitocondrial/antagonistas & inhibidores , Aldehído Deshidrogenasa Mitocondrial/química , Aldehído Deshidrogenasa Mitocondrial/metabolismo , Disulfiram/análogos & derivados , Disulfiram/síntesis química , Humanos , Simulación del Acoplamiento Molecular , Proteínas Recombinantes/metabolismo , Retinal-Deshidrogenasa/química , Retinal-Deshidrogenasa/metabolismo
9.
Mol Oncol ; 16(7): 1541-1554, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-34632694

RESUMEN

Disulfiram (DSF), an established alcohol-aversion drug, is a candidate for repurposing in cancer treatment. DSF's antitumor activity is supported by preclinical studies, case reports, and small clinical trials; however, ongoing clinical trials of advanced-stage cancer patients encounter variable results. Here, we show that one reason for the inconsistent clinical effects of DSF may reflect interference by other drugs. Using a high-throughput screening and automated microscopy, we identify cannabidiol, an abundant component of the marijuana plant used by cancer patients to mitigate side effects of chemotherapy, as a likely cause of resistance to DSF. Mechanistically, in cancer cells, cannabidiol triggers the expression of metallothioneins providing protective effects by binding heavy metal-based substances including the bis-diethyldithiocarbamate-copper complex (CuET). CuET is the documented anticancer metabolite of DSF, and we show here that the CuET's anticancer toxicity is effectively neutralized by metallothioneins. Overall, this work highlights an example of undesirable interference between cancer therapy and the concomitant usage of marijuana products. In contrast, we report that insufficiency of metallothioneins sensitizes cancer cells toward CuET, suggesting a potential predictive biomarker for DSF repurposing in oncology.


Asunto(s)
Cannabidiol , Disulfiram , Cannabidiol/farmacología , Cannabidiol/uso terapéutico , Línea Celular Tumoral , Cobre/química , Cobre/farmacología , Disulfiram/química , Disulfiram/farmacología , Disulfiram/uso terapéutico , Humanos , Metalotioneína
10.
Cells ; 10(12)2021 11 24.
Artículo en Inglés | MEDLINE | ID: mdl-34943802

RESUMEN

Nephropathic cystinosis is a rare disease caused by mutations of the CTNS gene that encodes for cystinosin, a lysosomal cystine/H+ symporter. The disease is characterized by early-onset chronic kidney failure and progressive development of extra-renal complications related to cystine accumulation in all tissues. At the cellular level, several alterations have been demonstrated, including enhanced apoptosis, altered autophagy, defective intracellular trafficking, and cell oxidation, among others. Current therapy with cysteamine only partially reverts some of these changes, highlighting the need to develop additional treatments. Among compounds that were identified in a previous drug-repositioning study, disulfiram (DSF) was selected for in vivo studies. The cystine depleting and anti-apoptotic properties of DSF were confirmed by secondary in vitro assays and after treating Ctns-/- mice with 200 mg/kg/day of DSF for 3 months. However, at this dosage, growth impairment was observed. Long-term treatment with a lower dose (100 mg/kg/day) did not inhibit growth, but failed to reduce cystine accumulation, caused premature death, and did not prevent the development of renal lesions. In addition, DSF also caused adverse effects in cystinotic zebrafish larvae. DSF toxicity was significantly more pronounced in Ctns-/- mice and zebrafish compared to wild-type animals, suggesting higher cell toxicity of DSF in cystinotic cells.


Asunto(s)
Cistinosis/patología , Disulfiram/toxicidad , Enfermedades Renales/patología , Pruebas de Toxicidad , Acetilcisteína/farmacología , Animales , Apoptosis , Cistina/metabolismo , Cistinosis/orina , Modelos Animales de Enfermedad , Disulfuros/metabolismo , Disulfiram/química , Embrión no Mamífero/metabolismo , Humanos , Enfermedades Renales/orina , Larva/metabolismo , Ratones Noqueados , Pez Cebra/embriología
11.
J Phys Chem Lett ; 12(44): 10880-10885, 2021 Nov 11.
Artículo en Inglés | MEDLINE | ID: mdl-34730355

RESUMEN

Despite the fact that chemotherapy has been widely used in the clinical treatment of breast cancer, the toxicity of chemotherapeutics to normal tissues cannot be ignored due to the low specificity. Therefore, due to the non-negligible toxicity of chemotherapeutic agents to normal tissues, tumor microenvironment (TME)-responsive cancer therapy has attracted a great deal of attention. Here, we report a TME-responsive theranostic nanoagent MnOx@PAA@HKUST-1-DSF@BSA fabricated via a layer-by-layer synthesis method. Once endocytosed by tumor cells, the nanoagent can be degraded into Mn2+ for magnetic resonance imaging and Cu2+ for Fenton-like reaction and chelating with released disulfiram in situ, achieving enhanced chemotherapy. Both in vitro and in vivo experiments demonstrate that the TME-targeted nanoagent can efficiently kill tumor cells. This work provides an alternative option for effective imaging and treatment of breast cancer without collateral damage to normal tissues.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Disulfiram/farmacología , Animales , Antineoplásicos/síntesis química , Antineoplásicos/química , Neoplasias de la Mama/patología , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Disulfiram/síntesis química , Disulfiram/química , Humanos , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Neoplasias Mamarias Experimentales/patología , Ratones , Tamaño de la Partícula , Nanomedicina Teranóstica , Microambiente Tumoral/efectos de los fármacos
12.
ACS Appl Mater Interfaces ; 13(43): 50760-50773, 2021 Nov 03.
Artículo en Inglés | MEDLINE | ID: mdl-34672620

RESUMEN

Therapeutic nanosystems triggered by a specific tumor microenvironment (TME) offer excellent safety and selectivity in the treatment of cancer by in situ conversion of a less toxic substance into effective anticarcinogens. However, the inherent antioxidant systems, hypoxic environment, and insufficient hydrogen peroxide (H2O2) in tumor cells severely limit their efficacy. Herein, a new strategy has been developed by loading the chemotherapy prodrug disulfiram (DSF) and coating glucose oxidase (GOD) on the surface of Cu/ZIF-8 nanospheres and finally encapsulating manganese dioxide (MnO2) nanoshells to achieve efficient DSF-based cancer chemotherapy and dual-enhanced chemodynamic therapy (CDT). In an acidic TME, the nanocatalyst can biodegrade rapidly and accelerate the release of internal active substances. The outer layer of MnO2 depletes glutathione (GSH) to destroy the reactive oxygen defensive mechanisms and achieves continuous oxygen generation, thus enhancing the catalytic efficiency of GOD to burst H2O2. Benefiting from the chelation reaction between the released Cu2+ and DSF, a large amount of cytotoxic CuET products is generated, and the Cu+ are concurrently released, thereby achieving efficient chemotherapy and satisfactory CDT efficacy. Furthermore, the release of Mn2+ can initiate magnetic resonance imaging signals for the tracking of the nanocatalyst.


Asunto(s)
Antineoplásicos/farmacología , Disulfiram/farmacología , Peróxido de Hidrógeno/farmacología , Antineoplásicos/química , Antineoplásicos/metabolismo , Catálisis , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Cobre/química , Cobre/metabolismo , Cobre/farmacología , Disulfiram/química , Disulfiram/metabolismo , Ensayos de Selección de Medicamentos Antitumorales , Glucosa Oxidasa/química , Glucosa Oxidasa/metabolismo , Células HeLa , Humanos , Peróxido de Hidrógeno/química , Peróxido de Hidrógeno/metabolismo , Iones/química , Iones/metabolismo , Iones/farmacología , Compuestos de Manganeso/química , Compuestos de Manganeso/metabolismo , Compuestos de Manganeso/farmacología , Estructura Molecular , Óxidos/química , Óxidos/metabolismo , Óxidos/farmacología , Tamaño de la Partícula , Zeolitas/química , Zeolitas/metabolismo , Zeolitas/farmacología
13.
Biol Pharm Bull ; 44(10): 1557-1564, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34602566

RESUMEN

Disulfiram (DSF) chelated with copper has been confirmed to have a strong anti-tumor ability. In this study, we determined that DSF-Cu induced mitochondria-dependent apoptosis in osteosarcoma (OS), reflecting in DSF-Cu induces mitochondrial membrane potential decline, the production of reactive oxygen species (ROS), and inhibiting cells migration and invasion along with decreasing the concentration of intracellular glutathione (GSH) and facilitating the opening of mitochondrial permeability transition pore (PT) in osteosarcoma cells. These anti-tumor activities can be reversed by Cyclosporine A (CsA, PT inhibitors) and N-acetyl-L-cysteine (NAC, antioxidants). Our results suggested that DSF-Cu exerts its anti-tumor effects in OS via regulation of the ROS/Mitochondria pathway. Our findings provide the basis for DSF-Cu to treat osteosarcoma, even might develop as a potential therapy for other tumors.


Asunto(s)
Neoplasias Óseas/tratamiento farmacológico , Disulfiram/farmacología , Osteosarcoma/tratamiento farmacológico , Animales , Apoptosis/efectos de los fármacos , Neoplasias Óseas/patología , Línea Celular Tumoral , Quelantes/química , Cobre/química , Disulfiram/química , Disulfiram/uso terapéutico , Humanos , Masculino , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones , Osteosarcoma/patología , Especies Reactivas de Oxígeno/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
14.
Biophys Chem ; 276: 106610, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34089978

RESUMEN

In the new millennium, the outbreak of new coronavirus has happened three times: SARS-CoV, MERS-CoV, and SARS-CoV-2. Unfortunately, we still have no pharmaceutical weapons against the diseases caused by these viruses. The pandemic of SARS-CoV-2 reminds us the urgency to search new drugs with totally different mechanism that may target the weaknesses specific to coronaviruses. Herein, we disclose a computational evaluation of targeted oxidation strategy (TOS) for potential inhibition of SARS-CoV-2 by disulfiram, a 70-year-old anti-alcoholism drug, and predict a multiple-target mechanism. A preliminary list of promising TOS drug candidates targeting the two thiol proteases of SARS-CoV-2 are proposed upon virtual screening of 32,143 disulfides.


Asunto(s)
Disuasivos de Alcohol/química , Antivirales/química , Proteasas 3C de Coronavirus/antagonistas & inhibidores , Proteasas Similares a la Papaína de Coronavirus/antagonistas & inhibidores , Disulfiram/química , Inhibidores de Proteasas/química , SARS-CoV-2/química , Disuasivos de Alcohol/farmacología , Antivirales/farmacología , Dominio Catalítico , Proteasas 3C de Coronavirus/química , Proteasas 3C de Coronavirus/genética , Proteasas 3C de Coronavirus/metabolismo , Proteasas Similares a la Papaína de Coronavirus/química , Proteasas Similares a la Papaína de Coronavirus/genética , Proteasas Similares a la Papaína de Coronavirus/metabolismo , Disulfiram/farmacología , Reposicionamiento de Medicamentos , Expresión Génica , Humanos , Cinética , Simulación del Acoplamiento Molecular , Oxidación-Reducción , Inhibidores de Proteasas/farmacología , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Estructura Secundaria de Proteína , Teoría Cuántica , SARS-CoV-2/enzimología , Especificidad por Sustrato , Termodinámica , Tratamiento Farmacológico de COVID-19
15.
Phys Chem Chem Phys ; 23(21): 12204-12215, 2021 Jun 02.
Artículo en Inglés | MEDLINE | ID: mdl-34008604

RESUMEN

Hepatitis C virus (HCV) is a notorious member of the Flaviviridae family of enveloped, positive-strand RNA viruses. Non-structural protein 5A (NS5A) plays a key role in HCV replication and assembly. NS5A is a multi-domain protein which includes an N-terminal amphipathic membrane anchoring alpha helix, a highly structured domain-1, and two intrinsically disordered domains 2-3. The highly structured domain-1 contains a zinc finger (Zf)-site, and binding of zinc stabilizes the overall structure, while ejection of this zinc from the Zf-site destabilizes the overall structure. Therefore, NS5A is an attractive target for anti-HCV therapy by disulfiram, through ejection of zinc from the Zf-site. However, the zinc ejection mechanism is poorly understood. To disclose this mechanism based on three different states, A-state (NS5A protein), B-state (NS5A + Zn), and C-state (NS5A + Zn + disulfiram), we have performed molecular dynamics (MD) simulation in tandem with DFT calculations in the current study. The MD results indicate that disulfiram triggers Zn ejection from the Zf-site predominantly through altering the overall conformation ensemble. On the other hand, the DFT assessment demonstrates that the Zn adopts a tetrahedral configuration at the Zf-site with four Cys residues, which indicates a stable protein structure morphology. Disulfiram binding induces major conformational changes at the Zf-site, introduces new interactions of Cys39 with disulfiram, and further weakens the interaction of this residue with Zn, causing ejection of zinc from the Zf-site. The proposed mechanism elucidates the therapeutic potential of disulfiram and offers theoretical guidance for the advancement of drug candidates.


Asunto(s)
Antivirales/farmacología , Disulfiram/farmacología , Hepacivirus/efectos de los fármacos , Proteínas no Estructurales Virales/antagonistas & inhibidores , Zinc/farmacología , Antivirales/síntesis química , Antivirales/química , Teoría Funcional de la Densidad , Disulfiram/química , Humanos , Simulación de Dinámica Molecular , Zinc/química
16.
Int J Mol Sci ; 22(7)2021 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-33918312

RESUMEN

Disulfiram (DSF), an irreversible aldehyde dehydrogenase inhibitor, is being used in anticancer therapy, as its effects in humans are known and less adverse than conventional chemotherapy. We explored the potential mechanism behind the cytotoxicity of DSF-Cu+/Cu2+ complexes in oral epidermoid carcinoma meng-1 (OECM-1) and human gingival epithelial Smulow-Glickman (SG) cells. Exposure to CuCl2 or CuCl slightly but concentration-dependently decreased cell viability, while DSF-Cu+/Cu2+ induced cell death in OECM-1 cells, but not SG cells. DSF-Cu+/Cu2+ also increased the subG1 population and decreased the G1, S, and G2/M populations in OECM-1 cells, but not SG cells, and suppressed cell proliferation in both OECM-1 and SG cells. ALDH enzyme activity was inhibited by CuCl and DSF-Cu+/Cu2+ in SG cells, but not OECM-1 cells. ROS levels and cellular senescence were increased in DSF-Cu+/Cu2+-treated OECM-1 cells, whereas they were suppressed in SG cells. DSF-Cu+/Cu2+ induced mitochondrial fission in OECM-1 cells and reduced mitochondrial membrane potential. CuCl2 increased but DSF- Cu2+ impaired oxygen consumption rates and extracellular acidification rates in OECM-1 cells. CuCl2 stabilized HIF-1α expression under normoxia in OECM-1 cells, and complex with DSF enhanced that effect. Levels of c-Myc protein and its phosphorylation at Tyr58 and Ser62 were increased, while levels of the N-terminal truncated form (Myc-nick) were decreased in DSF-Cu+/Cu2-treated OECM-1 cells. These effects were all suppressed by pretreatment with the ROS scavenger NAC. Overexpression of c-Myc failed to induce HIF-1α expression. These findings provide novel insight into the potential application of DSF-CuCl2 complex as a repurposed agent for OSCC cancer therapy.


Asunto(s)
Inhibidores del Acetaldehído Deshidrogenasa/uso terapéutico , Carcinoma de Células Escamosas/tratamiento farmacológico , Cobre/uso terapéutico , Disulfiram/uso terapéutico , Neoplasias de la Boca/tratamiento farmacológico , Inhibidores del Acetaldehído Deshidrogenasa/química , Inhibidores del Acetaldehído Deshidrogenasa/farmacología , Carcinoma de Células Escamosas/metabolismo , Cobre/química , Disulfiram/química , Disulfiram/farmacología , Reposicionamiento de Medicamentos , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Mitocondrias/efectos de los fármacos , Neoplasias de la Boca/metabolismo , Cultivo Primario de Células , Proteínas Proto-Oncogénicas c-myc/metabolismo
17.
Int J Biol Macromol ; 176: 490-497, 2021 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-33582217

RESUMEN

Disulfiram is a promising repurposed drug that, combining with radiation and chemotherapy, exhibits effective anticancer activities in several preclinical models. The cellular metabolites of disulfiram have been established, however, the intracellular targets of disulfiram remain largely unexplored. We have previously reported that disulfiram suppresses the coronaviral papain-like proteases through attacking their zinc-finger domains, suggesting an inhibitory function potentially on other proteases with similar catalytic structures. Ubiquitin-specific proteases (USPs) share a highly-conserved zinc-finger subdomain that structurally similar to the papain-like proteases and are attractive anticancer targets as upregulated USPs levels are found in a variety of tumors. Here, we report that disulfiram functions as a competitive inhibitor for both USP2 and USP21, two tumor-related deubiquitinases. In addition, we also observed a synergistic inhibition of USP2 and USP21 by disulfiram and 6-Thioguanine (6TG), a clinical drug for acute myeloid leukemia. Kinetic analyses revealed that both drugs exhibited a slow-binding mechanism, moderate inhibitory parameters, and a synergistically inhibitory effect on USP2 and USP21, suggesting the potential combinatory use of these two drugs for USPs-related tumors. Taken together, our study provides biochemical evidence for repurposing disulfiram and 6TG as a combinatory treatment in clinical applications.


Asunto(s)
Disulfiram/química , Inhibidores Enzimáticos/química , Tioguanina/química , Ubiquitina Tiolesterasa , Disulfiram/agonistas , Sinergismo Farmacológico , Humanos , Tioguanina/agonistas , Ubiquitina Tiolesterasa/antagonistas & inhibidores , Ubiquitina Tiolesterasa/química
18.
Nat Commun ; 12(1): 121, 2021 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-33402676

RESUMEN

p97, also known as valosin-containing protein (VCP) or Cdc48, plays a central role in cellular protein homeostasis. Human p97 mutations are associated with several neurodegenerative diseases. Targeting p97 and its cofactors is a strategy for cancer drug development. Despite significant structural insights into the fungal homolog Cdc48, little is known about how human p97 interacts with its cofactors. Recently, the anti-alcohol abuse drug disulfiram was found to target cancer through Npl4, a cofactor of p97, but the molecular mechanism remains elusive. Here, using single-particle cryo-electron microscopy (cryo-EM), we uncovered three Npl4 conformational states in complex with human p97 before ATP hydrolysis. The motion of Npl4 results from its zinc finger motifs interacting with the N domain of p97, which is essential for the unfolding activity of p97. In vitro and cell-based assays showed that the disulfiram derivative bis-(diethyldithiocarbamate)-copper (CuET) can bypass the copper transporter system and inhibit the function of p97 in the cytoplasm by releasing cupric ions under oxidative conditions, which disrupt the zinc finger motifs of Npl4, locking the essential conformational switch of the complex.


Asunto(s)
Coenzimas/química , Ditiocarba/análogos & derivados , Péptidos y Proteínas de Señalización Intracelular/química , Proteínas Nucleares/química , Compuestos Organometálicos/química , Ubiquitina/química , Proteína que Contiene Valosina/química , Adenosina Trifosfato/análogos & derivados , Adenosina Trifosfato/química , Adenosina Trifosfato/metabolismo , Sitios de Unión , Clonación Molecular , Coenzimas/genética , Coenzimas/metabolismo , Microscopía por Crioelectrón , Disulfiram/química , Disulfiram/metabolismo , Ditiocarba/química , Ditiocarba/metabolismo , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Expresión Génica , Vectores Genéticos/química , Vectores Genéticos/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Modelos Moleculares , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Compuestos Organometálicos/metabolismo , Unión Proteica , Conformación Proteica , Dominios y Motivos de Interacción de Proteínas , Desplegamiento Proteico , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Especificidad por Sustrato , Ubiquitina/genética , Ubiquitina/metabolismo , Proteína que Contiene Valosina/antagonistas & inhibidores , Proteína que Contiene Valosina/genética , Proteína que Contiene Valosina/metabolismo , Dedos de Zinc
19.
Cells ; 9(10)2020 09 25.
Artículo en Inglés | MEDLINE | ID: mdl-32993012

RESUMEN

We attempted to design an ophthalmic in situ gel formulation incorporating disulfiram (DIS) nanoparticles (Dis-NPs/ISG) and demonstrated the therapeutic effect of Dis-NPs/ISG on retinal dysfunction in 15-month-old Otsuka Long-Evans Tokushima Fatty (OLETF) rats, a rat model of diabetes. The DIS particles were crushed using a bead mill to prepare the nanoparticles, and the Dis-NPs/ISG was prepared using a combination of the DIS nanoparticles and an in situ gelling system based on methylcellulose (MC). The particle size of the Dis-NPs/ISG was 80-250 nm, and there was no detectable precipitation or aggregation for 1 month. Moreover, the Dis-NPs/ISG was gelled at 37 °C, and the drug was delivered into the retina by instillation. Only diethyldithiocarbamate (DDC) was detected in the retina (DIS was not detected) when the Dis-NPs/ISG was instilled in the right eye, and the DDC levels in the right retina were significantly higher than those in the left retina. In addition, the retinal residence time of the drug was prolonged by the application of the in situ gelling system, since the DDC levels in the retinas of rats instilled with Dis-NPs/ISG were higher than those in DIS nanoparticles without MC. Furthermore, repetitive instillation of the Dis-NPs/ISG attenuated the deterioration of electroretinograms (ERGs) in 15-month-old OLETF rats by preventing the collapse of ATP production via excessive nitric oxide and recovered the decrease in retinal function. These findings provide important information for the development of novel therapeutic approaches to diabetic retinopathy.


Asunto(s)
Retinopatía Diabética/tratamiento farmacológico , Geles/farmacología , Nanopartículas/uso terapéutico , Enfermedades de la Retina/tratamiento farmacológico , Adenosina Trifosfato/metabolismo , Animales , Retinopatía Diabética/patología , Modelos Animales de Enfermedad , Disulfiram/química , Disulfiram/farmacología , Geles/química , Humanos , Nanopartículas/química , Ratas , Ratas Endogámicas OLETF , Retina/efectos de los fármacos , Retina/patología , Enfermedades de la Retina/patología
20.
Mol Pharm ; 17(10): 3857-3869, 2020 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-32833457

RESUMEN

Disulfiram (DSF) is an FDA-approved anti-alcoholic drug that has recently proven to be effective in cancer treatment. However, the short half-life in the bloodstream and the metal ion-dependent antitumor activity significantly limited the further application of DSF in the clinical field. To this end, we constructed a silk fibroin modified disulfiram/zinc oxide nanocomposites (SF/DSF@ZnO) to solubilize and stabilize DSF, and, more importantly, achieve pH triggered Zn2+ release and subsequent synergistic antitumor activity. The prepared SF/DSF@ZnO nanocomposites were spherical and had a high drug loading. Triggered by the lysosomal pH, SF/DSF@ZnO could induce the rapid release of Zn2+ under the acidic conditions and caused nanoparticulate disassembly along with DSF release. In vitro experiments showed that cytotoxicity of DSF could be enhanced by the presence of Zn2+, and further amplified when encapsulated into SF/DSF@ZnO nanocomposites. It was confirmed that the significantly amplified cytotoxicity of SF/DSF@ZnO was resulted from pH-triggered Zn2+ release, inhibited cell migration, and increased ROS production. In vivo study showed that SF/DSF@ZnO nanocomposites significantly increased the tumor accumulation and prolonged the retention time. In vivo antitumor experiments in the xenograft model showed that SF/DSF@ZnO exerted the highest tumor-inhibition rate among all the drug treatments. Therefore, this exquisite study established silk fibroin-modified disulfiram/zinc oxide nanocomposites, SF/DSF@ZnO, where ZnO not only acted as a delivery carrier but also served as a metal ion reservoir to achieve synergistic antitumor efficacy. The established DSF nanoformulation displayed excellent therapeutic potential in future cancer treatment.


Asunto(s)
Antineoplásicos/farmacocinética , Nanocompuestos/administración & dosificación , Neoplasias/tratamiento farmacológico , Zinc/farmacocinética , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/química , Bombyx/química , Cationes Bivalentes/farmacocinética , Línea Celular Tumoral/trasplante , Modelos Animales de Enfermedad , Disulfiram/administración & dosificación , Disulfiram/química , Disulfiram/farmacocinética , Composición de Medicamentos/métodos , Liberación de Fármacos , Ensayos de Selección de Medicamentos Antitumorales , Sinergismo Farmacológico , Femenino , Fibroínas/química , Semivida , Humanos , Concentración de Iones de Hidrógeno , Ratones , Neoplasias/patología , Óxido de Zinc/administración & dosificación , Óxido de Zinc/química , Óxido de Zinc/farmacocinética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...